1
|
Djeghim H, Bellil I, Benslama O, Lekmine S, Temim E, Boufendi H, Postigo I, Sánchez P, Khelifi D. Effects of genetic diversity on the allergenicity of peanut (Arachis hypogaea) proteins: identification of the hypoallergenic accessions using BALB/c mice model and in silico analysis of Ara h 3 allergen cross-reactivity. J Proteomics 2024; 306:105264. [PMID: 39047939 DOI: 10.1016/j.jprot.2024.105264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
This study investigated the effects of genetic diversity in the allergenicity of peanut and assessed the allergenic capacity of six Arachis hypogaea accessions using a Balb/c mouse model. It also explored potential cross-reactivities between Ara h 3 (peanut allergen) and Gly m (soybean allergen) using computational tools. Female Balb/c mice were injected with peanut protein extracts and alum. Serum-specific antibodies (IgE, IgGt, IgG1, IgG2a) were measured using ELISA, and allergic protein profiles were examined via western blot. Structural homology, B cell epitopes, and molecular interactions between Ara h 3 and Gly m with human IgE were also investigated. The mice developed high sIgE and sIgG1 responses, with antibodies recognizing 19 bands on western blot. Notably, Saharan accessions showed unique features such as no bands on western blot profiles, reduced anaphylactic symptoms, lower IgE titers, and less intestinal tissue damage. Molecular docking results suggest significant cross-allergenicity, supported by allergenicity predictions and structural homology analysis. This comprehensive analysis provides insights into shared epitopes, potential competition for binding sites, and molecular dynamics of cross-reactive responses, enhancing understanding of food allergen interactions. The study recommends using Algerian Sahara peanut accessions in breeding, genomics studies, and industry for safer peanut options for individuals with allergies. SIGNIFICANCE: The significance of this study lies in its contribution to addressing a major public health issue: peanut allergy, which represents a significant cause of anaphylaxis affecting numerous individuals and families worldwide. By exploring the genetic diversity of peanut proteins and identifying hypoallergenic accessions through experimental and computational approaches, this research offers valuable insights for mitigating allergic reactions. The findings highlight that certain accessions from the Saharan region exhibit reduced allergenicity, resulting in attenuated anaphylactic symptoms, lower IgE levels, and reduced intestinal damage in murine models. Furthermore, the study's in silico analysis sheds light on the issue of cross-reactivity between peanut and soybean allergens, providing crucial information for understanding allergen interactions at the molecular level. Overall, this research contributes to advancing knowledge in the field of food allergen research and has practical implications for improving the quality of life for individuals allergic to peanuts, particularly through the selection of safer peanut varieties and their cultivation.
Collapse
Affiliation(s)
- Hanène Djeghim
- Laboratoire de Génétique Biochimie et Biotechnologie Végétale, Faculté des Sciences de la Nature et de la Vie, Université Frères Mentouri Constantine 1, Constantine 25000, Algeria; Laboratoire de Biochimie, Division de Biotechnologie et Santé, Centre de Recherche en Biotechnologie (CRBt), Constantine 25000, Algeria
| | - Ines Bellil
- Laboratoire de Génétique Biochimie et Biotechnologie Végétale, Faculté des Sciences de la Nature et de la Vie, Université Frères Mentouri Constantine 1, Constantine 25000, Algeria
| | - Ouided Benslama
- Laboratory of Natural Substances, Biomolecules, and Biotechnological Applications, Department of Natural and Life Sciences, Larbi Ben M'Hidi University, Oum El Bouaghi 04000, Algeria
| | - Sabrina Lekmine
- Biotechnology, Water, Environment and Health Laboratory, Abbes Laghrour University, Khenchela, Algeria
| | - Esma Temim
- Laboratoire de Microscopie et Diagnostic Génétique, Centre de Recherche en Biotechnologie (CRBt), 25000 Constantine, Algeria
| | - Houda Boufendi
- Laboratoire de Microscopie et Diagnostic Génétique, Centre de Recherche en Biotechnologie (CRBt), 25000 Constantine, Algeria
| | - Idoia Postigo
- Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy and Laboratory of Parasitology and Allergy, Lascaray Research Centre, University of the Basque Country, Vitoria, Spain.
| | - Patricia Sánchez
- Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy and Laboratory of Parasitology and Allergy, Lascaray Research Centre, University of the Basque Country, Vitoria, Spain
| | - Douadi Khelifi
- Laboratoire de Génétique Biochimie et Biotechnologie Végétale, Faculté des Sciences de la Nature et de la Vie, Université Frères Mentouri Constantine 1, Constantine 25000, Algeria; Ecole Nationale Supérieure de Biotechnologie (ENSB), Constantine 25000, Algeria
| |
Collapse
|
2
|
De Paepe E, Plekhova V, Vangeenderhuysen P, Baeck N, Bullens D, Claeys T, De Graeve M, Kamoen K, Notebaert A, Van de Wiele T, Van Den Broeck W, Vanlede K, Van Winckel M, Vereecke L, Elliott C, Cox E, Vanhaecke L. Integrated gut metabolome and microbiome fingerprinting reveals that dysbiosis precedes allergic inflammation in IgE-mediated pediatric cow's milk allergy. Allergy 2024; 79:949-963. [PMID: 38193259 DOI: 10.1111/all.16005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND IgE-mediated cow's milk allergy (IgE-CMA) is one of the first allergies to arise in early childhood and may result from exposure to various milk allergens, of which β-lactoglobulin (BLG) and casein are the most important. Understanding the underlying mechanisms behind IgE-CMA is imperative for the discovery of novel biomarkers and the design of innovative treatment and prevention strategies. METHODS We report a longitudinal in vivo murine model, in which two mice strains (BALB/c and C57Bl/6) were sensitized to BLG using either cholera toxin or an oil emulsion (n = 6 per group). After sensitization, mice were challenged orally, their clinical signs monitored, antibody (IgE and IgG1) and cytokine levels (IL-4 and IFN-γ) measured, and fecal samples subjected to metabolomics. The results of the murine models were further extrapolated to fecal microbiome-metabolome data from our population of IgE-CMA (n = 22) and healthy (n = 23) children (Trial: NCT04249973), on which polar metabolomics, lipidomics and 16S rRNA metasequencing were performed. In vitro gastrointestinal digestions and multi-omics corroborated the microbial origin of proposed metabolic changes. RESULTS During mice sensitization, we observed multiple microbially derived metabolic alterations, most importantly bile acid, energy and tryptophan metabolites, that preceded allergic inflammation. We confirmed microbial dysbiosis, and its associated effect on metabolic alterations in our patient cohort, through in vitro digestions and multi-omics, which was accompanied by metabolic signatures of low-grade inflammation. CONCLUSION Our results indicate that gut dysbiosis precedes allergic inflammation and nurtures a chronic low-grade inflammation in children on elimination diets, opening important new opportunities for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Ellen De Paepe
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Integrative Metabolomics (LIMET), Ghent University, Merelbeke, Belgium
| | - Vera Plekhova
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Integrative Metabolomics (LIMET), Ghent University, Merelbeke, Belgium
| | - Pablo Vangeenderhuysen
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Integrative Metabolomics (LIMET), Ghent University, Merelbeke, Belgium
| | - Nele Baeck
- Department of Pediatrics, Pediatric Gastroenterology, AZ Jan Palfijn Ghent, Ghent, Belgium
| | - Dominique Bullens
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Clinical Division of Pediatrics, UZ Leuven, Leuven, Belgium
| | - Tania Claeys
- Department of Pediatrics, Pediatric Gastroenterology and Nutrition & General Pediatric Medicine, AZ Sint-Jan Bruges, Bruges, Belgium
| | - Marilyn De Graeve
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Integrative Metabolomics (LIMET), Ghent University, Merelbeke, Belgium
| | - Kristien Kamoen
- Department of Pediatrics, Maria Middelares Ghent, Ghent, Belgium
| | - Anneleen Notebaert
- Department of Pediatrics, Sint-Vincentius Hospital Deinze, Deinze, Belgium
| | - Tom Van de Wiele
- Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent University, Ghent, 9000, Belgium
| | - Wim Van Den Broeck
- Faculty of Veterinary Medicine, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Ghent University, Merelbeke, Belgium
| | - Koen Vanlede
- Department of General Pediatrics, VITAZ, Sint-Niklaas, Belgium
| | - Myriam Van Winckel
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Lars Vereecke
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent, Belgium
| | - Chris Elliott
- School of Biological Sciences, Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| | - Eric Cox
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Immunology, Ghent University, Merelbeke, Belgium
| | - Lynn Vanhaecke
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Laboratory of Integrative Metabolomics (LIMET), Ghent University, Merelbeke, Belgium
- School of Biological Sciences, Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
3
|
Issa M, Michaudel C, Guinot M, Grauso-Culetto M, Guillon B, Lecardonnel J, Jouneau L, Chapuis C, Bernard H, Hazebrouck S, Castelli F, Fenaille F, Gaultier E, Rivière G, Houdeau E, Adel-Patient K. Long-term exposure from perinatal life to food-grade TiO 2 alters intestinal homeostasis and predisposes to food allergy in young mice. Allergy 2024; 79:471-484. [PMID: 38010857 DOI: 10.1111/all.15960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Food allergy (FA) is an inappropriate immunological response to food proteins resulting from an impaired induction of oral tolerance. Various early environmental factors can affect the establishment of intestinal homeostasis, predisposing to FA in early life. In this context, we aimed to assess the effect of chronic perinatal exposure to food-grade titanium dioxide (fg-TiO2 ), a common food additive. METHODS Dams were fed a control versus fg-TiO2 -enriched diet from preconception to weaning, and their progeny received the same diet at weaning. A comprehensive analysis of baseline intestinal and systemic homeostasis was performed in offspring 1 week after weaning by assessing gut barrier maturation and microbiota composition, and local and systemic immune system and metabolome. The effect of fg-TiO2 on the susceptibility of progeny to develop oral tolerance versus FA to cow's milk proteins (CMP) was performed starting at the same baseline time-point, using established models. Sensitization to CMP was investigated by measuring β-lactoglobulin and casein-specific IgG1 and IgE antibodies, and elicitation of the allergic reaction by measuring mouse mast cell protease (mMCP1) in plasma collected after an oral food challenge. RESULTS Perinatal exposure to fg-TiO2 at realistic human doses led to an increased propensity to develop FA and an impaired induction of oral tolerance only in young males, which could be related to global baseline alterations in intestinal barrier, gut microbiota composition, local and systemic immunity, and metabolism. CONCLUSIONS Long-term perinatal exposure to fg-TiO2 alters intestinal homeostasis establishment and predisposes to food allergy, with a clear gender effect.
Collapse
Affiliation(s)
- Mohammad Issa
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Chloé Michaudel
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Marine Guinot
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Marta Grauso-Culetto
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Blanche Guillon
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Jérôme Lecardonnel
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Luc Jouneau
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Céline Chapuis
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Hervé Bernard
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Stephane Hazebrouck
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| | - Florence Castelli
- Université Paris-Saclay, CEA, INRAE - UMR Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Innovation en Spectrométrie de Masse, Saclay, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE - UMR Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Innovation en Spectrométrie de Masse, Saclay, France
| | - Eric Gaultier
- Toxalim UMR1331 (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Gilles Rivière
- Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail (ANSES), Direction de l'Evaluation des Risques, Maisons-Alfort, France
| | - Eric Houdeau
- Toxalim UMR1331 (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Karine Adel-Patient
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI/Laboratoire d'Immuno-Allergie Alimentaire, Gif-sur-Yvette, France
| |
Collapse
|
4
|
Enzymatic hydrolysis of silkworm pupa and its allergenicity evaluation by animal model with different immunization routes. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
5
|
Tao Y, Zhou E, Li F, Meng L, Li Q, Wu L. Allergenicity Alleviation of Bee Pollen by Enzymatic Hydrolysis: Regulation in Mice Allergic Mediators, Metabolism, and Gut Microbiota. Foods 2022; 11:foods11213454. [PMID: 36360070 PMCID: PMC9658975 DOI: 10.3390/foods11213454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Bee pollen as a nutrient-rich functional food has been considered for use as an adjuvant for chronic disease therapy. However, bee pollen can trigger food-borne allergies, causing a great concern to food safety. Our previous study demonstrated that the combined use of cellulase, pectinase and papain can hydrolyze allergens into peptides and amino acids, resulting in reduced allergenicity of bee pollen based on in vitro assays. Herein, we aimed to further explore the mechanisms behind allergenicity alleviation of enzyme-treated bee pollen through a BALB/c mouse model. Results showed that the enzyme-treated bee pollen could mitigate mice scratching frequency, ameliorate histopathological injury, decrease serum IgE level, and regulate bioamine production. Moreover, enzyme-treated bee pollen can modulate metabolic pathways and gut microbiota composition in mice, further supporting the alleviatory allergenicity of enzyme-treated bee pollen. The findings could provide a foundation for further development and utilization of hypoallergenic bee pollen products.
Collapse
Affiliation(s)
- Yuxiao Tao
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China
| | - Enning Zhou
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China
| | - Fukai Li
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China
| | - Lifeng Meng
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China
| | - Qiangqiang Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China
- Correspondence: ; Tel.: +86-132-6949-5300
| | - Liming Wu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China
| |
Collapse
|
6
|
Immunomodulatory Role of BLG-Derived Peptides Based on Simulated Gastrointestinal Digestion and DC-T Cell from Mice Allergic to Cow's Milk. Foods 2022; 11:foods11101450. [PMID: 35627020 PMCID: PMC9140701 DOI: 10.3390/foods11101450] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
Peptides, but not whole protein, elicit an allergic reaction since food allergens should be consumed by digestion. In this study, we explored the remaining peptides after simulated digestion of cow’s milk in order to search for β-lactoglobulin (BLG)-derived peptides that could play an immunomodulatory role. As a major allergen in milk, BLG-derived peptides, 109 in total, were identified both from simulated infant and adult digestion in vitro. These peptides were mainly located in four regions, and they were synthesized as five peptides, namely, BLG1–14, BLG24–35, BLG40–60, BLG82–101, and BLG123–139. Then, the effect of peptides on the Caco-2 cell’s transport absorption, the co-stimulatory molecules of DC, and the T-cell phenotype was explored. The results suggested all peptides showed better transport absorption capacity with the apparent permeability coefficient higher than 2 × 10−6 cm·s−1. The ability of BLG40–60 for promoting lamina propria-derived DC cell (LPDC) maturation was observed by the increase in MHC II. Moreover, BLG1–14 and BLG40–60 directed activation of T lymphocytes towards a Th1 phenotype. This is the first report of the immunomodulatory potential of peptides in the sensitization of allergic reaction, and one peptide, BLG40–60, was regarded as an immunomodulatory peptide, one that should be further explored in an animal model in depth.
Collapse
|
7
|
Route of Sensitization to Peanut Influences Immune Cell Recruitment at Various Mucosal Sites in Mouse: An Integrative Analysis. Nutrients 2022; 14:nu14040790. [PMID: 35215440 PMCID: PMC8875549 DOI: 10.3390/nu14040790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Symptom occurrence at the first ingestion suggests that food allergy may result from earlier sensitization via non-oral routes. We aimed to characterize the cellular populations recruited at various mucosal and immune sites after experimental sensitization though different routes. BALB/cJ mice were exposed to a major allergenic food (peanut) mixed with cholera toxin via the intra-gastric (i.g.), respiratory, cutaneous, or intra-peritoneal (i.p.) route. We assessed sensitization and elicitation of the allergic reaction and frequencies of T cells, innate lymphoid cells (ILC), and inflammatory and dendritic cells (DC) in broncho-alveolar lavages (BAL), lungs, skin, intestine, and various lymph nodes. All cellular data were analyzed through non-supervised and supervised uni/multivariate analysis. All exposure routes, except cutaneous, induced sensitization, but intestinal allergy was induced only in i.g.- and i.p.-exposed mice. Multivariate analysis of all cellular constituents did not discriminate i.g. from control mice. Conversely, respiratory-sensitized mice constituted a distinct cluster, characterized by high local inflammation and immune cells recruitment. Those mice also evidenced changes in ILC frequencies at distant site (intestine). Despite absence of sensitization, cutaneous-exposed mice evidenced comparable changes, albeit less intense. Our study highlights that the initial route of sensitization to a food allergen influences the nature of the immune responses at various mucosal sites. Interconnections of mucosal immune systems may participate in the complexity of clinical manifestations as well as in the atopic march.
Collapse
|
8
|
Zhang Y, Liu JY, Shao JW, Luo QQ, Zhang YQ, Song G, Wang CY, Zhao SY, Wan C, Du XH, Xu LZ. Effective Model of Food Allergy in Mice Sensitized with Ovalbumin and Freud's Adjuvant. Bull Exp Biol Med 2021; 171:352-356. [PMID: 34297293 DOI: 10.1007/s10517-021-05226-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 12/29/2022]
Abstract
To better explore the pathophysiology of FA and its therapy, we aimed to establish a simple and practicable FA model with Freund's adjuvant and introduce an easy and reliable laboratory evaluation method for assessment of inflammation in intestinal segments at different anatomical locations. BALB/c mice were sensitized with ovalbumin combined with Freund's adjuvant. Complete Freund's adjuvant was chosen for the first sensitization and two weeks later incomplete Freund's adjuvant was used for a second sensitization. Two weeks later, the sensitized mice were challenged with 50 mg ovalbumin every other day. After the 6 challenge, all mice were assessed for systemic anaphylaxis, and then sacrificed for sample collection. All sensitized mice showed anaphylactic symptoms and markedly increased levels of serum ovalbumin-specific IgE and IgG1. The activity of mast cell protease-1 (mMCPT-1) was significantly increased in the serum and interstitial fluid of the duodenum, jejunum, ileum, and colon. A successful FA model was established, of which inflammation occurred in the duodenum, jejunum, ileum, and colon. This model provides a reliable and simple tool for analysis of the mechanism of FA and methods of immunotherapy. Moreover, combined detection of ovalbumin-specific antibody and local mMCPT-1 levels could potentially be used as the major indicator for assessment of food allergy.
Collapse
Affiliation(s)
- Y Zhang
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - J Y Liu
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - J W Shao
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Q Q Luo
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Y Q Zhang
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - G Song
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - C Y Wang
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - S Y Zhao
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - C Wan
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - X H Du
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - L Z Xu
- Key Lab for Immunology, Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China.
| |
Collapse
|
9
|
Benedé S, Berin MC. Applications of Mouse Models to the Study of Food Allergy. Methods Mol Biol 2021; 2223:1-17. [PMID: 33226583 DOI: 10.1007/978-1-0716-1001-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mouse models of allergic disease offer numerous advantages when compared to the models of other animals. However, selection of appropriate mouse models is critical to advance the field of food allergy by revealing mechanisms of allergy and for testing novel therapeutic approaches. All current mouse models for food allergy have weaknesses that may limit their applicability to human disease. Aspects such as the genetic predisposition to allergy or tolerance from the strain of mouse used, allergen dose, route of exposure (oral, intranasal, intraperitoneal, or epicutaneous), damage of the epithelial barrier, use of adjuvants, food matrix effects, or composition of the microbiota should be considered prior to the selection of a specific murine model and contemplated according to the intended purpose of the study. This chapter reviews our current knowledge on the application of mouse models to food allergy research and the variables that may influence the successful development of each type of model.
Collapse
Affiliation(s)
- Sara Benedé
- Instituto de Investigación en Ciencias de la Alimentación (CIAL), CSIC-UAM, Madrid, Spain
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Cecilia Berin
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Induction of Hypersensitivity with Purified Beta-Lactoglobulin as a Mouse Model of Cow's Milk Allergy. Methods Mol Biol 2021; 2223:67-78. [PMID: 33226587 DOI: 10.1007/978-1-0716-1001-5_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cow's milk allergy is one of the most prevalent food allergies in both children and adults. As dairy products are common dietary ingredients and the prevalence of chronic conditions is on the rise, milk allergy is a growing public health concern. To elucidate underlying mechanisms and develop therapeutic strategies, reliable animal models are essential research tools. Sensitization to a milk protein is the principal procedure for establishing animal models of cow's milk allergy. However, the methods of sensitization vary from laboratory to laboratory, using different milk proteins with different amounts, routes, and durations of allergen exposure during sensitization of varying sex and strains of mice, likely resulting in diverse immunological and physical responses. Furthermore, the sources and potential impurities of milk protein may also produce variable responses. Thus, standardization of sensitization protocol is important, particularly when results are compared across studies. Here, we describe a method to generate a mouse model of cow's milk allergy using purified β-lactoglobulin as the milk allergen with cholera toxin as an adjuvant in a 5-week oral sensitization protocol.
Collapse
|
11
|
Adel-Patient K, Guinot M, Guillon B, Bernard H, Chikhi A, Hazebrouck S, Junot C. Administration of Extensive Hydrolysates From Caseins and Lactobacillus rhamnosus GG Probiotic Does Not Prevent Cow's Milk Proteins Allergy in a Mouse Model. Front Immunol 2020; 11:1700. [PMID: 33042105 PMCID: PMC7516991 DOI: 10.3389/fimmu.2020.01700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/25/2020] [Indexed: 11/18/2022] Open
Abstract
Background Early nutrition may influence the development of food allergies later in life. In the absence of breastfeeding, hydrolysates from cow's milk proteins (CMP) were indicated as a prevention strategy in at risk infants, but their proof of effectiveness in clinical and pre-clinical studies is still insufficient. Thanks to a validated mouse model, we then assessed specific and nonspecific preventive effects of administration of extensive hydrolysates from caseins (eHC) on the development of food allergy to CMP. The additional nonspecific effect of the probiotic Lactobacillus GG (LGG), commonly used in infant formula, was also assessed. Methods Groups of young BALB/cByJ female mice were pretreated by repeated gavage either with PBS (control mice), or with PBS solution containing non-hydrolyzed milk protein isolate (MPI), eHC or eHC+LGG (eq. of 10 mg of protein/gavage). All mice were then experimentally sensitized to CMP by gavage with whole CM mixed with the Th2 mucosal adjuvant Cholera toxin. All mice were further chronically exposed to cow's milk. A group of mice was kept naïve. Sensitization to both caseins and to the non-related whey protein β-lactoglobulin (BLG) was evaluated by measuring specific antibodies in plasma and specific ex vivo Th2/Th1/Th17 cytokine secretion. Elicitation of the allergic reaction was assessed by measuring mMCP1 in plasma obtained after oral food challenge (OFC) with CMP. Th/Treg cell frequencies in gut-associated lymphoid tissue and spleen were analyzed by flow cytometry at the end of the protocol. Robust statistical procedure combining non-supervised and supervised multivariate analyses and univariate analyses, was conducted to reveal any effect of the pretreatments. Results PBS pretreated mice were efficiently sensitized and demonstrated elicitation of allergic reaction after OFC, whereas mice pretreated with MPI were durably protected from allergy to CMP. eHC+/-LGG pretreatments had no protective effect on sensitization to casein (specific) or BLG (non-specific), nor on CMP-induced allergic reactions. Surprisingly, eHC+LGG mice demonstrated significantly enhanced humoral and cellular immune responses after sensitization with CMP. Only some subtle changes were evidenced by flow cytometry. Conclusion Neither specific nor nonspecific preventive effects of administration of casein-derived peptides on the development of CMP food allergy were evidenced in our experimental setup. Further studies should be conducted to delineate the mechanisms involved in the immunostimulatory potential of LGG and to clarify its significance in clinical use.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Marine Guinot
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Blanche Guillon
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hervé Bernard
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Amina Chikhi
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université d’Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Stéphane Hazebrouck
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Christophe Junot
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
12
|
Miteva D, Solak A, Dyankova S, Nacheva I, Dimov K. Assessment of allergenicity of irradiated dairy products in a Balb/c mice model. PHARMACIA 2020. [DOI: 10.3897/pharmacia.67.e53886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The objective of the presentin vivostudy was to analyze the changes in the immune response of the sensitized Ваlb/c mice against milk allergens in lyophilized cow’s milk (LCM) and whey curd (LC) before and after gamma irradiation (10 kGy). The results showed lower levels of IgE in the group treated with irradiated LCM (ICM) compared to the group treated with non-irradiated LCM (NiCM). Hence, it could be suggested that gamma irradiation influenced the epitopes of the major milk proteins and was associated with lower allergenicity of the lyophilized irradiated milk. The gamma irradiation in the whey curd, however, did not significantly change the level of IgE antibodies in IC (treated with irradiated LC) compared to NiC (treated with non-irradiated LC) group.
Collapse
|
13
|
Adel-Patient K, Bernard H, Fenaille F, Hazebrouck S, Junot C, Verhasselt V. Prevention of Allergy to a Major Cow's Milk Allergen by Breastfeeding in Mice Depends on Maternal Immune Status and Oral Exposure During Lactation. Front Immunol 2020; 11:1545. [PMID: 32849523 PMCID: PMC7396486 DOI: 10.3389/fimmu.2020.01545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background: The high incidence of food allergy in childhood points to the need of elucidating early life factors dictating allergy susceptibility. Here, we aim to address in a mouse model how the exposure to a major cow's milk allergen through breastmilk of mothers with different immune status influences food allergy outcome in offspring. Methods: BALB/cJ future dams were either kept naïve, or sensitized through the oral route using cholera toxin (“orally sensitized”) or through the i.p. route using alum (“i.p. sensitized”), or rendered fully tolerant (oral gavage without any adjuvant) to bovine β-lactoglobulin (BLG). After mating with naïve males and delivery, mothers were orally exposed or not to BLG during the whole lactation. Then, eight groups of lactating mothers were considered: naïve, i.p. sensitized, orally sensitized, or tolerant, each exposed or not during lactation. In order to specifically address breastmilk effects on their allergy susceptibility, pups from naïve-synchronized mothers were cross-fostered by the different groups of treated dams and lactating mothers at delivery. In some experiments, mothers kept their own pups to address a possible in utero effect. BLG antigen, BLG-specific antibodies, and BLG-immune complexes were measured in breastmilk from the different lactating mother groups. Allergic sensitization was monitored in 5-weeks old female offspring (n = 7–8/group of lactating mothers) by determining BLG-specific antibodies in plasma and splenocytes cytokine secretion after i.p. injections of BLG/alum. Allergic reaction to oral BLG challenge was evaluated by measuring mMCP1 in plasma. Results: Offspring was protected from one allergic i.p. sensitization when nursed by i.p. sensitized mothers, independently of BLG exposure during lactation. Orally sensitized dams conferred protection in offspring solely when exposed to BLG during lactation, while naïve mothers did not provide any protection upon BLG exposure. The levels of protection correlated with the levels of BLG-specific antibodies and BLG-immune complex in breastmilk. There was a trend for decreased sensitization in offspring breastfed by tolerant and exposed mothers, which was not associated with transfer of specific antibodies through breastmilk. Protection provided by nursing by treated/exposed mothers was not persistent after a boost i.p. injection of the progeny and then did not protect them from an allergic reaction induced at this time point. No additional in utero effects were evidenced. Conclusion: Our study demonstrates the strong potential of breastmilk to modulate immune response to a major cow's milk allergen in the progeny. It highlights the importance of maternal immune status and of her consumption of the allergen during lactation in dictating the outcomes in offspring. This opens perspectives where modulating maternal immune status might increase the chance of cow's milk allergy prevention in breastfed children.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hervé Bernard
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - François Fenaille
- Laboratoire du Métabolisme des Médicaments, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé, CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Stéphane Hazebrouck
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Christophe Junot
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France.,Laboratoire du Métabolisme des Médicaments, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé, CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Valérie Verhasselt
- Chair of Human Lactology, School of Molecular Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
14
|
Tong P, Xiong L, Gao J, Li X, Wu Z, Yang A, Yuan J, Wu Y, Chen H. Influence of heat treatment and egg matrix on the physicochemical and allergenic properties of egg custard. J Food Sci 2020; 85:789-799. [PMID: 32078753 DOI: 10.1111/1750-3841.15065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 11/27/2022]
Abstract
To investigate the influence of heat treatment and egg matrix on egg custard (EC) proteins, 12 different kinds of ECs with different egg/water ratios (1:1, 1:1.5, 1:2, or 1:3, v/v) and different heating temperatures (80, 90, or 100 °C) and times (10, 15, or 20 min) were prepared and evaluated for the digestibility, structure, eliciting capacity and sensitizing capacity using SDS-PAGE, fluorescence spectra, ELISA, and a BALB/c mouse model, respectively. The physicochemical properties of EC proteins were significantly affected by heat treatment and egg matrix, which showed the increased digestibility and partially unfolded structure. The eliciting capacity of EC evaluated by IgE binding to sera from egg-allergic patients was reduced after heat treatment, and the EC made by heating at 100 °C for 20 min with a whole egg/water ratio of 1:2 (v/v) was the weakest. The sensitizing capacity of EC was also reduced in the BALB/c mouse model, which showed the significantly decreased levels of specific IgE, IgG, IgG1 and IgG2a, mMCP-1 and histamine in the mouse sera, as well as cytokine secretions of IL-4, IL-5, and IL-13, compared with the raw egg (RE) group. Results demonstrate that heat treatment and egg matrix significantly reduced the eliciting and sensitizing capacity of EC by changing the tertiary structure and increasing the digestibility of EC proteins. PRACTICAL APPLICATION: Egg custard (EC) is one kind of savory food suitable for all ages, and is also a traditional supplementary food for infants and young children in China. However, limited information is available on the allergenicity of egg custard. In this work, we evaluated how the structure, digestibility, and allergenic potential of egg allergens in EC were altered by the degree of thermal treatment and egg matrix, and elucidated the links between the physicochemical properties and allergenic potential of EC affected by heat treatment and egg matrix. Our results demonstrate that heat treatment and egg matrix significantly reduced the eliciting and sensitizing capacity of EC by changing the tertiary structure and increasing the digestibility of EC proteins.
Collapse
Affiliation(s)
- Ping Tong
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China.,School of Food Science & Technology, Nanchang Univ., Nanchang, 330047, P. R. China
| | - Liji Xiong
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China
| | - Jinyan Gao
- School of Food Science & Technology, Nanchang Univ., Nanchang, 330047, P. R. China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China.,School of Food Science & Technology, Nanchang Univ., Nanchang, 330047, P. R. China
| | - Zhihua Wu
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China.,Sino-German Joint Research Inst., Nanchang Univ., Nanchang, 330047, P. R. China
| | - Anshu Yang
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China.,Sino-German Joint Research Inst., Nanchang Univ., Nanchang, 330047, P. R. China
| | - Juanli Yuan
- School of Pharmaceutical Science, Nanchang Univ., Nanchang, 330047, P. R. China
| | - Yong Wu
- Sino-German Joint Research Inst., Nanchang Univ., Nanchang, 330047, P. R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang Univ., Nanchang, 330047, P. R. China.,Sino-German Joint Research Inst., Nanchang Univ., Nanchang, 330047, P. R. China
| |
Collapse
|
15
|
Mouse Models for Food Allergies: Where Do We Stand? Cells 2019; 8:cells8060546. [PMID: 31174293 PMCID: PMC6627293 DOI: 10.3390/cells8060546] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Food allergies are a steadily increasing health and economic problem. Immunologically, food allergic reactions are caused by pathological, allergen-specific Th2 responses resulting in IgE-mediated mast cell degranulation and associated inflammatory reactions. Clinically, food allergies are characterized by local inflammation of the mouth mucosa, the face, the throat, the gastrointestinal tract, are frequently paralleled by skin reactions, and can result in life-threatening anaphylactic reactions. To better understand food allergies and establish novel treatment options, mouse models are indispensable. This review discusses the available mouse food allergy models, dividing them into four categories: (1) adjuvant-free mouse models, (2) mouse models relying on adjuvants to establish allergen-specific Th2 responses, (3) mouse models using genetically-modified mouse strains to allow for easier sensitization, and (4) humanized mouse models in which different immunodeficient mouse strains are reconstituted with human immune or stem cells to investigate humanized immune responses. While most of the available mouse models can reproducibly portray the immunological parameters of food allergy (Th2 immune responses, IgE production and mast cell activation/expansion), so far, the recreation of the clinical parameters has proven more difficult. Therefore, up to now none of the available mouse models can reproduce the complete human pathology.
Collapse
|
16
|
Chikhi A, Elmecherfi KE, Bernard H, Cortes-Perez N, Kheroua O, Saidi D, Adel-Patient K. Evaluation of the efficiency of hydrolyzed whey formula to prevent cow's milk allergy in the BALB/c mouse model. Pediatr Allergy Immunol 2019; 30:370-377. [PMID: 30672606 DOI: 10.1111/pai.13017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Partially hydrolyzed milk formulas have been proposed for primary prevention in at-risk infants, but evidence of their efficiency and elucidation of the underlying mechanisms are still lacking. Thanks to a Th2-biased mouse model mimicking at-risk patients, we aimed to assess the potency of a partially hydrolyzed whey formula (pHWF) to induce oral tolerance thus preventing further cow's milk (CM) allergy. METHODS BALB/c mice were gavaged with pHWF, standard milk formula (SF), or vehicle only (PBS+). All mice were then orally sensitized to CM using cholera toxin and further chronically exposed to CM. Humoral (IgE, IgG1, IgG2a) and cellular (Th2/Th1/Th17 cytokine secretion; frequency of CD4+GATA3+ and CD4+CD25+Foxp3+ T cells in the spleen) responses against β-lactoglobulin (BLG) and whole caseins (CAS) were assessed, as well as a marker of elicitation of allergic reaction (mMCP-1) released after an oral challenge with CM. RESULTS All markers of sensitization and of allergic reaction were evidenced in the PBS+ mice and were significantly enhanced upon chronic exposure. Gavage with SF totally and durably prevented sensitization and elicitation of the allergic reaction. Conversely, pre-treatment with pHWF only reduced BLG-specific sensitization (IgE, Th2 cytokines), with no significant effect on sensitization to caseins. However, pHWF pre-treatment significantly reduced mMCP-1 concentration in plasma after CM challenges. CD4+CD25+Foxp3+ Treg cell frequency could not be correlated with tolerance efficiency. CONCLUSION Partially hydrolyzed whey formula only partially prevents the further development of CM allergy in this Th2-biased model. A hydrolysate from both whey and casein fractions may be more efficient.
Collapse
Affiliation(s)
- Amina Chikhi
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Kamel-Eddine Elmecherfi
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Hervé Bernard
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Naima Cortes-Perez
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Omar Kheroua
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Djamel Saidi
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Karine Adel-Patient
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
17
|
Mondoulet L, Dioszeghy V, Busato F, Plaquet C, Dhelft V, Bethune K, Leclere L, Daviaud C, Ligouis M, Sampson H, Dupont C, Tost J. Gata3 hypermethylation and Foxp3 hypomethylation are associated with sustained protection and bystander effect following epicutaneous immunotherapy in peanut-sensitized mice. Allergy 2019; 74:152-164. [PMID: 29779209 PMCID: PMC6585762 DOI: 10.1111/all.13479] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2018] [Indexed: 12/17/2022]
Abstract
Background Epicutaneous immunotherapy (EPIT) is a promising method for treating food allergies. In animal models, EPIT induces sustained unresponsiveness and prevents further sensitization mediated by Tregs. Here, we elucidate the mechanisms underlying the therapeutic effect of EPIT, by characterizing the kinetics of DNA methylation changes in sorted cells from spleen and blood and by evaluating its persistence and bystander effect compared to oral immunotherapy (OIT). Methods BALB/c mice orally sensitized to peanut proteins (PPE) were treated by EPIT using a PPE‐patch or by PPE‐OIT. Another set of peanut‐sensitized mice treated by EPIT or OIT were sacrificed following a protocol of sensitization to OVA. DNA methylation was analyzed during immunotherapy and 8 weeks after the end of treatment in sorted cells from spleen and blood by pyrosequencing. Humoral and cellular responses were measured during and after immunotherapy. Results Analyses showed a significant hypermethylation of the Gata3 promoter detectable only in Th2 cells for EPIT from the 4th week and a significant hypomethylation of the Foxp3 promoter in CD62L+ Tregs, which was sustained only for EPIT. In addition, mice treated with EPIT were protected from subsequent sensitization and maintained the epigenetic signature characteristic for EPIT. Conclusions Our study demonstrates that EPIT leads to a unique and stable epigenetic signature in specific T‐cell compartments with downregulation of Th2 key regulators and upregulation of Treg transcription factors, likely explaining the sustainability of protection and the observed bystander effect.
Collapse
Affiliation(s)
| | | | - F. Busato
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | | | | | - K. Bethune
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | - L. Leclere
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | - C. Daviaud
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | | | - H. Sampson
- DBV Technologies Montrouge France
- Icahn School of Medicine at Mont Sinai New York NY USA
| | - C. Dupont
- Université Paris Descartes Hôpital Necker‐Enfants Malades Paris France
| | - J. Tost
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| |
Collapse
|
18
|
Assessment of the Sensitizing Potential of Proteins in BALB/c Mice: Comparison of Three Protocols of Intraperitoneal Sensitization. Nutrients 2018; 10:nu10070903. [PMID: 30011888 PMCID: PMC6073454 DOI: 10.3390/nu10070903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 11/16/2022] Open
Abstract
Most food allergy cases are associated with a limited group of allergens. This could be attributed to an increased ability of some foods to sensitize and trigger allergic reactions. However, there are no validated animal models to evaluate the sensitizing or allergenic potentials of proteins. Our aim was to evaluate three protocols of adjuvant-free intraperitoneal sensitization that differ in the time points for sample collection (days 14, 28 and 35 from beginning of the sensitization) and also in the number of immunizations (2, 5 and 3, respectively). Ovalbumin (OVA; 0.05 mg), cow milk proteins (CMP; 0.025, 0.05 and 0.25 mg), and potato acid phosphatase (PAP; low allergenic protein; 250.0 mg) were administered intraperitoneally (ip) to BALB/c mice (n = 4–6) and the protein-specific IgE and IgG antibody responses were evaluated using ELISA. Additional serum protein-specific IgE antibodies evaluations were carried out after IgG depletion. Anti-OVA IgE antibodies were detected in mice from all three protocols. The responses were higher in the group of mice that underwent the 28-day protocol than in those that underwent the 14- or 35-day protocols (p < 0.01 and p < 0.05, respectively). Anti-CMP IgE antibodies were detected in both the 14- and 28-day protocols, but the response was higher in the group that underwent the 28-day protocol (p < 0.001). The anti-CMP IgE antibody response detection was improved after serum IgG depletion (p < 0.001). Anti-PAP IgE antibodies were not detected. Mice with undetectable serum levels of protein-specific IgE triggered anti-OVA, -CMP, and -PAP IgG responses. An adjuvant-free 28-day protocol with five ip immunizations seems appropriate for evaluation of the inherent sensitizing or allergenic capacity of the studied proteins. Reproducible results were obtained utilizing the BALB/c mouse strain. Inter-laboratory studies including a larger number of proteins should be carried out to validate this model.
Collapse
|
19
|
Guendouz M, Haddi A, Grar H, Kheroua O, Saidi D, Kaddouri H. Preventive effects of royal jelly against anaphylactic response in a murine model of cow's milk allergy. PHARMACEUTICAL BIOLOGY 2017; 55:2145-2152. [PMID: 28982287 PMCID: PMC6130715 DOI: 10.1080/13880209.2017.1383487] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/15/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Royal jelly (RJ) has long been used to promote human health. OBJECTIVE The current study investigated the preventive effects of RJ against the development of a systemic and intestinal immune response in mice allergic to cow's milk proteins. MATERIALS AND METHODS Balb/c mice treated orally for seven days with RJ at doses of 0.5, 1 and 1.5 g/kg were sensitized intraperitoneally with β-lactoglobulin (β-Lg). Serum IgG and IgE anti-β-Lg were determined by an enzyme-linked immunosorbent assay (ELISA). Plasma histamine levels, symptom scores and body temperature were determined after in vivo challenge to β-Lg. Jejunums were used for assessment of local anaphylactic responses by an ex vivo study in Ussing chambers and morphologic changes by histological analysis. RESULTS RJ significantly decreased serum IgG (31.15-43.78%) and IgE (64.28-66.6%) anti-β-Lg and effectively reduced plasma histamine level (66.62-67.36%) (p < 0.001) at all the doses tested. Additionally, no clinical symptoms or body temperature drops were observed in RJ-pretreated mice. Interestingly, RJ significantly reduced (p < 0.001) intestinal dysfunction by abolishing the secretory response (70.73-72.23%) induced by sensitization and prevented length aberrations of jejunal villi by 44.32-59.01% (p < 0.001). DISCUSSION AND CONCLUSIONS We speculate that using RJ may help prevent systemic and anaphylactic response in allergic mice. These effects may be related to its inhibitory effects on the degranulation of mast cells.
Collapse
Affiliation(s)
- Malika Guendouz
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Abir Haddi
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Hadria Grar
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Omar Kheroua
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Djamel Saidi
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Hanane Kaddouri
- Laboratory of Physiology of the Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Sciences, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| |
Collapse
|
20
|
Villa C, Costa J, Oliveira MBP, Mafra I. Bovine Milk Allergens: A Comprehensive Review. Compr Rev Food Sci Food Saf 2017; 17:137-164. [DOI: 10.1111/1541-4337.12318] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Caterina Villa
- REQUIMTE-LAQV, Faculdade de Farmácia; Univ. do Porto; Porto Portugal
| | - Joana Costa
- REQUIMTE-LAQV, Faculdade de Farmácia; Univ. do Porto; Porto Portugal
| | | | - Isabel Mafra
- REQUIMTE-LAQV, Faculdade de Farmácia; Univ. do Porto; Porto Portugal
| |
Collapse
|
21
|
Gamazo C, García-Azpíroz M, Souza Rebouças JD, Gastaminza G, Ferrer M, Irache JM. Oral immunotherapy using polymeric nanoparticles loaded with peanut proteins in a murine model of fatal anaphylaxis. Immunotherapy 2017; 9:1205-1217. [DOI: 10.2217/imt-2017-0111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Carlos Gamazo
- Department of Microbiology, University of Navarra, Instituto de Investigación Sanitaria de Navarra (Idisna), C/Irunlarrea, 1; 31080 - Pamplona, Spain
| | - Maddi García-Azpíroz
- Department of Microbiology, University of Navarra, Instituto de Investigación Sanitaria de Navarra (Idisna), C/Irunlarrea, 1; 31080 - Pamplona, Spain
| | - Juliana De Souza Rebouças
- Department of Microbiology, University of Navarra, Instituto de Investigación Sanitaria de Navarra (Idisna), C/Irunlarrea, 1; 31080 - Pamplona, Spain
- Laboratory of Microbiology & Immunoregulation, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Gabriel Gastaminza
- Department of Allergology & Clinical Immunology, Clínica Universidad de Navarra, Navarra, Spain
| | - Marta Ferrer
- Department of Allergology & Clinical Immunology, Clínica Universidad de Navarra, Navarra, Spain
| | - Juan M Irache
- Department of Pharmacy & Pharmaceutical Technology, University of Navarra, Navarra, Spain
| |
Collapse
|
22
|
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; 60:2722-2735. [PMID: 27374416 PMCID: PMC5213772 DOI: 10.1002/mnfr.201500923] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/27/2016] [Accepted: 06/08/2016] [Indexed: 11/11/2022]
Abstract
SCOPE Processing of food has been shown to impact IgE binding and functionality of food allergens. In the present study, we investigated the impact of heat processing on the sensitization capacity of Ara h 6, a major peanut allergen and one of the most potent elicitors of the allergic reaction. METHODS AND RESULTS Peanut extracts obtained from raw or heat-processed peanut and some fractions thereof were biochemically and immunochemically characterized. These extracts/fractions, purified Ara h 6, or recombinant Ara h 6 including Ara h 6 mutants lacking disulfide bridges were used in in vitro digestion tests and mouse models of experimental sensitization. Peanut roasting led to the formation of complexes of high molecular weight, notably between Ara h 6 and Ara h 1, which supported the induction of IgE specific to native Ara h 6. On the contrary, a fraction containing free monomeric 2S albumins or purified native Ara h 6 displayed no intrinsic allergenicity. In addition to complex formation, heat denaturation and/or partial destabilization enhanced Ara h 6 immunogenicity and increased its sensitivity to digestion. CONCLUSION These results suggest that sensitization potency and IgE binding capacity can be supported by different structures, modified and/or produced during food processing in interaction with other food constituents.
Collapse
Affiliation(s)
- Blanche Guillon
- UMR CEA-INRA Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hervé Bernard
- UMR CEA-INRA Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Marie-Françoise Drumare
- UMR CEA-INRA Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Stéphane Hazebrouck
- UMR CEA-INRA Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Karine Adel-Patient
- UMR CEA-INRA Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
23
|
Cecchi R. Diagnosis of anaphylactic death in forensics: Review and future perspectives. Leg Med (Tokyo) 2016; 22:75-81. [PMID: 27591544 DOI: 10.1016/j.legalmed.2016.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 12/18/2022]
Abstract
The diagnosis of anaphylaxis in a pre- or post-mortal phase involves the formulation of problems not yet solved by the international scientific literature, due to the complexity of pathogenic factors and pathophysiological processes that characterizes it. For forensic autopsies, further problems of differential diagnosis arise and often leave the forensic pathologist unable to express an opinion of certainty, as a result of lack of case history, circumstantial and autoptical-histopathological data. Nevertheless, in routine cases the postmortem diagnosis of anaphylactic death continues to be based on exclusion and circumstantial evidence. The author, after an extensive review of the literature relating to deaths from anaphylaxis of forensic pathological interest, and a discussion of the microscopical and biochemical findings, proposes a diagnostic protocol for forensic purposes and evaluates the diagnostic perspectives enabled by the newly available analytic techniques and markers. Maybe, the application of omics methodologies could help in the future for anaphylaxis diagnosis.
Collapse
Affiliation(s)
- Rossana Cecchi
- Department of Biomedical, Biotechnological and Translational Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy.
| |
Collapse
|
24
|
Bøgh KL, van Bilsen J, Głogowski R, López-Expósito I, Bouchaud G, Blanchard C, Bodinier M, Smit J, Pieters R, Bastiaan-Net S, de Wit N, Untersmayr E, Adel-Patient K, Knippels L, Epstein MM, Noti M, Nygaard UC, Kimber I, Verhoeckx K, O'Mahony L. Current challenges facing the assessment of the allergenic capacity of food allergens in animal models. Clin Transl Allergy 2016; 6:21. [PMID: 27313841 PMCID: PMC4910256 DOI: 10.1186/s13601-016-0110-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/09/2016] [Indexed: 01/16/2023] Open
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.
Collapse
Affiliation(s)
| | | | | | - Iván López-Expósito
- Department of Bioactivity and Food Analysis, Institute for Food Science Research (CIAL) (CSIC-UAM), Madrid, Spain
| | | | | | | | - Joost Smit
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Raymond Pieters
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Shanna Bastiaan-Net
- Food and Biobased Research, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Nicole de Wit
- Food and Biobased Research, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Eva Untersmayr
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Karine Adel-Patient
- UMR-INRA-CEA, Service de Pharmacologie et d'Immunoanalyse, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Leon Knippels
- Danone Nutricia Research, Utrecht, The Netherlands ; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Michelle M Epstein
- Experimental Allergy Laboratory, DIAID, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Mario Noti
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Ian Kimber
- University of Manchester, Manchester, UK
| | | | - Liam O'Mahony
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos Platz, Switzerland
| |
Collapse
|
25
|
Negaoui H, El Mecherfi KE, Tadjer SA, Grar H, Kheroua O, Saidi D. Bovine lactoferrin allergenicity as studied in murine model of allergy. FOOD AGR IMMUNOL 2016. [DOI: 10.1080/09540105.2016.1160365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Hanane Negaoui
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| | - Kamel Eddine El Mecherfi
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| | - Sid Ahmed Tadjer
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| | - Hadria Grar
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| | - Omar Kheroua
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| | - Djamel Saidi
- Laboratory of the Physiology of Nutrition and Food Safety, Department of Biology, Faculty of Natural and Life Science, University of Oran 1, Ahmed Ben Bella, Oran, Algeria
| |
Collapse
|
26
|
Liu T, Navarro S, Lopata AL. Current advances of murine models for food allergy. Mol Immunol 2016; 70:104-17. [DOI: 10.1016/j.molimm.2015.11.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/21/2015] [Accepted: 11/28/2015] [Indexed: 12/16/2022]
|
27
|
Yoo JM, Park YW, Yoon SY, Son JY, Jeong SG, Park BY, Kim JW, Nam MS. Thymic Stromal Lymphopoietin Induction Is Mediated by the Major Whey Proteins α-Lactalbumin and β-Lactoglobulin through the NF-κB Pathway in Immune Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:10803-10810. [PMID: 26618482 DOI: 10.1021/acs.jafc.5b04790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
α-Lactalbumin and β-lactoglobulin are two major whey proteins that specifically bind immunoglobulin E and are suspected as major allergens causing cow's milk allergy (CMA). Recent studies have shown that thymic stromal lymphopoietin is a critical factor linking at the interface of the body and environment to the T-helper 2 response. However, it is not known whether thymic stromal lymphopoietin expression is changed by α-lactalbumin and β-lactoglobulin in immune cells. Using RT-PCR and ELISA, the present study was conducted to examine if intravenous injection of α-lactalbumin and β-lactoglobulin increased pro-inflammatory cytokines, T-helper 2 cytokines, and thymic stromal lymphopoietin expression in several immune cells, including macrophages, mast cells, and keratinocytes. Results showed that α-lactalbumin and β-lactoglobulin induced thymic stromal lymphopoietin, interleukin-6, and tumor necrosis factor-α expression. It was concluded that the allergenicity of α-lactalbumin and β-lactoglobulin may be attributed to thymic stromal lymphopoietin induction, T-helper 2 cytokines, and pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Jae-Min Yoo
- Department of Animal Biosystem Science, Chungnam National University (CNU) , 99 Daehak-ro, Yusung-gu, Daejeon 34134, Republic of Korea
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology , Daejeon 34141, Republic of Korea
| | - Young W Park
- Agricultural Research Station, Fort Valley State University , Fort Valley, Georgia 31030, United States
- Department of Food Science & Technology, University of Georgia , Athens, Georgia 30602, United States
| | - Sun Young Yoon
- ENZYCHEM Life Sciences, 741, KAIST-ICC 193, Munji-Ro, Daejeon 34051, Republic of Korea
| | - Ji Yoon Son
- Department of Animal Biosystem Science, Chungnam National University (CNU) , 99 Daehak-ro, Yusung-gu, Daejeon 34134, Republic of Korea
| | - Seok Geun Jeong
- National Institute of Animal Science, Rural Development Administration , Wanju 55365, Republic of Korea
| | - Beom-Young Park
- National Institute of Animal Science, Rural Development Administration , Wanju 55365, Republic of Korea
| | - Jae Wha Kim
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology , Daejeon 34141, Republic of Korea
| | - Myoung Soo Nam
- Department of Animal Biosystem Science, Chungnam National University (CNU) , 99 Daehak-ro, Yusung-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
28
|
Wu Z, Zhou N, Xiong F, Li X, Yang A, Tong P, Tang R, Chen H. Allergen composition analysis and allergenicity assessment of Chinese peanut cultivars. Food Chem 2015; 196:459-65. [PMID: 26593515 DOI: 10.1016/j.foodchem.2015.09.070] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 08/25/2015] [Accepted: 09/16/2015] [Indexed: 11/17/2022]
Abstract
Peanut (Arachis hypogaea) is among the eight major food allergens in the world. Several attempts have been made to decrease or eliminate the allergenicity of peanut. Systemic screening of thousands of peanut cultivars may identify peanut with low allergenicity. In this study, the allergen compositions of 53 Chinese peanut cultivars were characterized, and their allergenicity to sera IgE of Chinese patients and in a mouse model was assessed. Contents of total protein and allergens were quantified by SDS-PAGE and densitometry analysis on gel. Although the contents of allergens broadly varied among cultivars, they were related to one another. The IgE binding capacity of cultivars was tested by ELISA, and their allergenicity was further evaluated in a mouse model by oral sensitization. Results showed that the allergenicity of peanut was affected by allergen composition rather than a single allergen. Peanut cultivars with low allergenicity may contain more Ara h 3/4 (24 kDa), Ara h 2 and less Ara h 3/4 (43, 38, and 36 kDa), Ara h 6. Screening based on allergen composition would facilitate the identification of low-allergenic peanut.
Collapse
Affiliation(s)
- Zhihua Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Ningling Zhou
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China; Department of Food Science, Nanchang University, Nanchang 330047, China
| | - Faqian Xiong
- Cash Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Department of Food Science, Nanchang University, Nanchang 330047, China
| | - Anshu Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Ronghua Tang
- Cash Crops Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
29
|
Mondoulet L, Dioszeghy V, Puteaux E, Ligouis M, Dhelft V, Plaquet C, Dupont C, Benhamou PH. Specific epicutaneous immunotherapy prevents sensitization to new allergens in a murine model. J Allergy Clin Immunol 2015; 135:1546-57.e4. [DOI: 10.1016/j.jaci.2014.11.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 11/04/2014] [Accepted: 11/12/2014] [Indexed: 01/06/2023]
|
30
|
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; 5:3. [PMID: 25671077 PMCID: PMC4322461 DOI: 10.1186/s13601-015-0047-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/07/2015] [Indexed: 02/04/2023] Open
Abstract
Background Allergic sensitisation to food may occur through non-gastrointestinal routes such as via skin or lung. We recently demonstrated in mice that cutaneous or respiratory pre-exposures to peanut proteins on intact epithelia induce a Th2 priming and allow subsequent oral sensitization without the use of adjuvant. We then aimed to assess the impact of a similar pattern of exposure to another relevant food allergen, cows’ milk. Findings The humoral and cellular immune response induced in BALB/cJ mice after repeated cutaneous applications on intact skin or after intranasal administration of cows’ milk proteins was analysed. In order to assess the potential effect of the food matrix, we used either a purified major cows’ milk allergen, β-lactoglobulin (BLG), or whole cows’ milk containing the same amount of BLG. We then studied the impact of these pre-exposures on a subsequent oral exposure to milk in the presence or absence of the mucosal Th2 adjuvant, Cholera toxin (CT). Cutaneous applications of milk induced production of BLG-specific IgE and IgG1 in 5 and 8 mice out of 20 respectively, whereas purified BLG alone did not. Intranasal exposure to milk, but not to BLG, led to BLG-specific IgG1 production in 8 out of 20 mice. Notably, cutaneous pre-exposure to milk favours further oral sensitisation without CT, while intra-nasal pre-exposure to BLG prevents further experimental sensitisation. Conclusions Altogether, our results thus demonstrated that the immune response induced after non-gastrointestinal exposure to food depends on the allergen, the matrix and the route of exposure.
Collapse
Affiliation(s)
- Sophie Wavrin
- Unité INRA d'Immuno-Allergie Alimentaire, IBiTec-S - SPI, Bât. 136 - CEA de Saclay, 91191 Gif-sur-Yvette Cedex, France
| | - Herve Bernard
- Unité INRA d'Immuno-Allergie Alimentaire, IBiTec-S - SPI, Bât. 136 - CEA de Saclay, 91191 Gif-sur-Yvette Cedex, France
| | - Jean-Michel Wal
- AgroParisTech - Department SVS, 16 rue Claude Bernard, F-75231 Paris Cedex 05, France
| | - Karine Adel-Patient
- Unité INRA d'Immuno-Allergie Alimentaire, IBiTec-S - SPI, Bât. 136 - CEA de Saclay, 91191 Gif-sur-Yvette Cedex, France
| |
Collapse
|
31
|
El Mecherfi KE, Rouaud O, Curet S, Negaoui H, Chobert JM, Kheroua O, Saidi D, Haertlé T. Peptic hydrolysis of bovine beta-lactoglobulin under microwave treatment reduces its allergenicity in anex vivomurine allergy model. Int J Food Sci Technol 2014. [DOI: 10.1111/ijfs.12653] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kamel-Eddine El Mecherfi
- Université d'Oran; Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire; Équipe Allergie; BP 1524 EL M'Naouer 31000 Oran Algéria
- UR 1268 Biopolymères Interactions Assemblages; Institut National de Recherche Agronomique; Équipe Fonctions et Interactions des Protéines; rue de la Géraudière BP 71627 44316 Nantes Cedex 3 France
| | - Olivier Rouaud
- ONIRIS; CNRS; GEPEA; UMR 6144 rue de la Géraudière CS82225 44322 Nantes France
| | - Sébastien Curet
- ONIRIS; CNRS; GEPEA; UMR 6144 rue de la Géraudière CS82225 44322 Nantes France
| | - Hanane Negaoui
- Université d'Oran; Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire; Équipe Allergie; BP 1524 EL M'Naouer 31000 Oran Algéria
| | - Jean-Marc Chobert
- UR 1268 Biopolymères Interactions Assemblages; Institut National de Recherche Agronomique; Équipe Fonctions et Interactions des Protéines; rue de la Géraudière BP 71627 44316 Nantes Cedex 3 France
| | - Omar Kheroua
- Université d'Oran; Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire; Équipe Allergie; BP 1524 EL M'Naouer 31000 Oran Algéria
| | - Djamel Saidi
- Université d'Oran; Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire; Équipe Allergie; BP 1524 EL M'Naouer 31000 Oran Algéria
| | - Thomas Haertlé
- UR 1268 Biopolymères Interactions Assemblages; Institut National de Recherche Agronomique; Équipe Fonctions et Interactions des Protéines; rue de la Géraudière BP 71627 44316 Nantes Cedex 3 France
| |
Collapse
|
32
|
No adjuvant effect of Bacillus thuringiensis-maize on allergic responses in mice. PLoS One 2014; 9:e103979. [PMID: 25084284 PMCID: PMC4118972 DOI: 10.1371/journal.pone.0103979] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 07/09/2014] [Indexed: 11/19/2022] Open
Abstract
Genetically modified (GM) foods are evaluated carefully for their ability to induce allergic disease. However, few studies have tested the capacity of a GM food to act as an adjuvant, i.e. influencing allergic responses to other unrelated allergens at acute onset and in individuals with pre-existing allergy. We sought to evaluate the effect of short-term feeding of GM Bacillus thuringiensis (Bt)-maize (MON810) on the initiation and relapse of allergic asthma in mice. BALB/c mice were provided a diet containing 33% GM or non-GM maize for up to 34 days either before ovalbumin (OVA)-induced experimental allergic asthma or disease relapse in mice with pre-existing allergy. We observed that GM-maize feeding did not affect OVA-induced eosinophilic airway and lung inflammation, mucus hypersecretion or OVA-specific antibody production at initiation or relapse of allergic asthma. There was no adjuvant effect upon GM-maize consumption on the onset or severity of allergic responses in a mouse model of allergic asthma.
Collapse
|
33
|
Wavrin S, Bernard H, Wal JM, Adel-Patient K. Cutaneous or respiratory exposures to peanut allergens in mice and their impacts on subsequent oral exposure. Int Arch Allergy Immunol 2014; 164:189-99. [PMID: 25034179 DOI: 10.1159/000363444] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 05/07/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recent data suggested that non-gastrointestinal exposure can lead to sensitisation to food allergens. We thus assessed the immune impact of respiratory or cutaneous exposure to peanut proteins on non-altered epithelium and investigated the effect of such pre-exposure on subsequent oral administration of peanut. METHODS BALB/cJ mice were exposed to purified Ara h 1 or to a non-defatted roasted peanut extract (PE) by simple deposit of allergens solutions on non-altered skin or in the nostrils. Exposures were performed 6 times at weekly intervals. Pre-exposed mice then received intra-gastric administrations of PE alone or in the presence of the Th2 mucosal adjuvant cholera toxin (CT). The specific humoral and cellular immune response was assessed throughout the protocol. RESULTS Both cutaneous and respiratory exposures led to the production of specific IgG1. Local and systemic IL-5 and IL-13 production were also evidenced, demonstrating activation of specific Th2 cells. This effect was dose-dependent and most efficient via the respiratory route. Moreover, these pre-exposures led to the production of specific IgE antibodies after gavage with PE, whatever the presence of CT. CONCLUSIONS Cutaneous or respiratory exposures to peanut induce Th2 priming in mice. Moreover, pre-exposures promote further sensitisation via the oral route without the use of CT; this proposes a new adjuvant-free experimental model of sensitisation to food that may reflect a realistic exposure pattern in infants. These results also suggest that non-gastrointestinal peanut exposure should be minimised in high-risk infants, even those with non-altered skin, to potentially reduce allergic sensitisation to this major food allergen.
Collapse
Affiliation(s)
- Sophie Wavrin
- INRA, UR496 Immuno-Allergie Alimentaire, CEA/IBiTeC-S/SPI, CEA de Saclay, Gif-sur-Yvette, France
| | | | | | | |
Collapse
|
34
|
Bernard H, Ah-Leung S, Drumare MF, Feraudet-Tarisse C, Verhasselt V, Wal JM, Créminon C, Adel-Patient K. Peanut allergens are rapidly transferred in human breast milk and can prevent sensitization in mice. Allergy 2014; 69:888-97. [PMID: 24773443 DOI: 10.1111/all.12411] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Food allergens have been evidenced in breast milk under physiological conditions, but the kinetic and the role of this passage in food allergies are still unclear. We then aimed to analyze the passage of peanut allergens in human breast milk and their allergenicity/immunomodulatory properties. METHODS Human breast milk was collected from two non-atopic peanut-tolerant mothers before and at different time points after ingestion of 30 g of commercial roasted peanut. Ara h 6, Ara h 6 immune complexes, and the IgE binding capacity of breast milk samples were measured using specific immunoassays. Their allergenic functionality was then assessed using cell-based assay. Finally, human breast milk obtained before or after peanut ingestion was administered intragastrically to BALB/c mice at different ages, and mice were further experimentally sensitized to peanut using cholera toxin. RESULTS Ara h 6 is detected as soon as 10 min after peanut ingestion, with peak values observed within the first hour after ingestion. The transfer is long-lasting, small quantities of peanut allergens being detected over a 24-h period. IgG-Ara h 6 and IgA-Ara h 6 immune complexes are evidenced, following a different kinetic of excretion than free allergens. Peanut allergens transferred in milk are IgE reactive and can induce an allergic reaction in vitro. However, administration of human breast milk to young mice, notably before weaning, does not lead to sensitization, but instead to partial oral tolerance. CONCLUSION The low quantities of immunologically active allergens transferred through breast milk may prevent instead of priming allergic sensitization to peanut.
Collapse
Affiliation(s)
- H. Bernard
- INRA; UR496 Unité d'Immuno-Allergie Alimentaire; Jouy-en-Josas France
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - S. Ah-Leung
- INRA; UR496 Unité d'Immuno-Allergie Alimentaire; Jouy-en-Josas France
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - M.-F. Drumare
- INRA; UR496 Unité d'Immuno-Allergie Alimentaire; Jouy-en-Josas France
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - C. Feraudet-Tarisse
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - V. Verhasselt
- EA 6302 Tolérance Immunitaire; Hôpital de l'Archet; Université de Nice Sophia-Antipolis; Nice France
| | - J.-M. Wal
- INRA; UR496 Unité d'Immuno-Allergie Alimentaire; Jouy-en-Josas France
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - C. Créminon
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| | - K. Adel-Patient
- INRA; UR496 Unité d'Immuno-Allergie Alimentaire; Jouy-en-Josas France
- CEA; IBiTecS; Service de Pharmacologie et d'Immunoanalyse; Gif-sur-Yvette France
| |
Collapse
|
35
|
Role of cellular immunity in cow's milk allergy: pathogenesis, tolerance induction, and beyond. Mediators Inflamm 2014; 2014:249784. [PMID: 25002754 PMCID: PMC4070503 DOI: 10.1155/2014/249784] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/22/2014] [Indexed: 12/14/2022] Open
Abstract
Food allergy is an aberrant immune-mediated reaction against harmless food substances, such as cow's milk proteins. Due to its very early introduction, cow's milk allergy is one of the earliest and most common food allergies. For this reason cow's milk allergy can be recognized as one of the first indications of an aberrant inflammatory response in early life. Classically, cow's milk allergy, as is true for most other allergies as well, is primarily associated with abnormal humoral immune responses, that is, elevation of specific immunoglobulin E levels. There is growing evidence indicating that cellular components of both innate and adaptive immunity play significant roles during the pathogenesis of cow's milk allergy. This is true for the initiation of the allergic phenotype (stimulation and skewing towards sensitization), development and outgrowth of the allergic disease. This review discusses findings pertaining to roles of cellular immunity in allergic inflammation, and tolerance induction against cow's milk proteins. In addition, a possible interaction between immune mechanisms underlying cow's milk allergy and other types of inflammation (infections and noncommunicable diseases) is discussed.
Collapse
|
36
|
Neunkirchner A, Schmetterer KG, Pickl WF. Lymphocyte-based model systems for allergy research: a historic overview. Int Arch Allergy Immunol 2014; 163:259-91. [PMID: 24777172 PMCID: PMC7617143 DOI: 10.1159/000360163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During the last decades, a multitude of studies applying distinct in vitro and in vivo model systems have contributed greatly to our better understanding of the initiation and regulation of inflammatory processes leading to allergic diseases. Over the years, it has become evident that among lymphocytes, not only IgE-producing B cells and allergy-orchestrating CD4(+) helper cells but also cytotoxic CD8(+) T cells, γδ-T cells and innate lymphoid cells, as well as regulatory lymphocytes, might critically shape the immune response towards usually innocuous allergens. In this review, we provide a historic overview of pioneering work leading to the establishment of important lymphocyte-based model systems for allergy research. Moreover, we contrast the original findings with our currently more refined knowledge to appreciate the actual validity of the respective models and to reassess the conclusions obtained from them. Conflicting studies and interpretations are identified and discussed. The tables are intended to provide an easy overview of the field not only for scientists newly entering the field but also for the broader readership interested in updating their knowledge. Along those lines, herein we discuss in vitro and in vivo approaches to the investigation of lymphocyte effector cell activation, polarization and regulation, and describe depletion and adoptive transfer models along with gene knockout and transgenic (tg) methodologies. In addition, novel attempts to establish humanized T cell antigen receptor tg mouse models for allergy research are described and discussed.
Collapse
Affiliation(s)
- Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
37
|
Ladics GS, Fry J, Goodman R, Herouet-Guicheney C, Hoffmann-Sommergruber K, Madsen CB, Penninks A, Pomés A, Roggen EL, Smit J, Wal JM. Allergic sensitization: screening methods. Clin Transl Allergy 2014; 4:13. [PMID: 24739743 PMCID: PMC3990213 DOI: 10.1186/2045-7022-4-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/09/2014] [Indexed: 11/10/2022] Open
Abstract
Experimental in silico, in vitro, and rodent models for screening and predicting protein sensitizing potential are discussed, including whether there is evidence of new sensitizations and allergies since the introduction of genetically modified crops in 1996, the importance of linear versus conformational epitopes, and protein families that become allergens. Some common challenges for predicting protein sensitization are addressed: (a) exposure routes; (b) frequency and dose of exposure; (c) dose-response relationships; (d) role of digestion, food processing, and the food matrix; (e) role of infection; (f) role of the gut microbiota; (g) influence of the structure and physicochemical properties of the protein; and (h) the genetic background and physiology of consumers. The consensus view is that sensitization screening models are not yet validated to definitively predict the de novo sensitizing potential of a novel protein. However, they would be extremely useful in the discovery and research phases of understanding the mechanisms of food allergy development, and may prove fruitful to provide information regarding potential allergenicity risk assessment of future products on a case by case basis. These data and findings were presented at a 2012 international symposium in Prague organized by the Protein Allergenicity Technical Committee of the International Life Sciences Institute's Health and Environmental Sciences Institute.
Collapse
Affiliation(s)
- Gregory S Ladics
- DuPont Pioneer Agricultural Biotechnology, DuPont Experimental Station, 200 Powder Mill Road, Wilmington, DE 19880-0400, USA
| | - Jeremy Fry
- ProImmune Limited, The Magdalen Centre, The Oxford Science Park, Robert Robinson Avenue, Oxford OX4 4GA, United Kingdom
| | - Richard Goodman
- Department of Food Science & Technology, Food Allergy Research and Resource Program, University of Nebraska–Lincoln, 143 Food Industry Complex, Lincoln, Nebraska, USA
| | | | - Karin Hoffmann-Sommergruber
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Charlotte B Madsen
- Department of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, 19, Mørkhøj Bygade, DK-2860 Søborg, Denmark
| | - André Penninks
- TNO Triskelion BV, Utrechtseweg 48, 3700 AV Zeist, Netherlands
| | - Anna Pomés
- Indoor Biotechnologies, Inc, 1216 Harris Street, Charlottesville, Virginia, USA
| | - Erwin L Roggen
- Novozymes AS and 3Rs Management and Consultancy, Krogshoejvej 36, 2880 Bagsvaerd, Denmark
| | - Joost Smit
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 104, 3508 TD Utrecht, Netherlands
| | - Jean-Michel Wal
- AgroParisTech, Department SVS, 16 rue Claude Bernard, F-75231, Paris Cedex 05, France
| |
Collapse
|
38
|
Sun H, Liu X, Wang YZ, Liu JX, Feng J. Allergen-specific immunoglobulin, histamine and T-cell responses induced by soybean glycinin and β-conglycinin in BALB/c mice of oral sensitisation. FOOD AGR IMMUNOL 2013. [DOI: 10.1080/09540105.2012.730501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
39
|
Steele L, Mayer L, Berin MC. Mucosal immunology of tolerance and allergy in the gastrointestinal tract. Immunol Res 2013; 54:75-82. [PMID: 22447352 DOI: 10.1007/s12026-012-8308-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mucosal immune system typically exists in a state of active tolerance to food antigens and commensal bacteria. Tolerance to food proteins is induced in part by dendritic cells residing in the intestinal mucosa and implemented by regulatory T cells. Food allergy occurs when immune tolerance is disrupted and a sensitizing immune response characterized by food-specific IgE production occurs instead. Experimental food allergy in mice requires use of adjuvant or exploitation of alternate routes of sensitization to induce allergic sensitization, and can aid in understanding the mechanisms of sensitization to food allergens and the pathophysiology of gastrointestinal manifestations of food allergy. Recent work in the understanding of mucosal immunology of tolerance and allergy in the gastrointestinal tract will be discussed.
Collapse
Affiliation(s)
- Lauren Steele
- Mount Sinai School of Medicine, Immunology Institute, New York, NY 10029, USA
| | | | | |
Collapse
|
40
|
Cholera toxin induces a shift from inactive to active cyclooxygenase 2 in alveolar macrophages activated by Mycobacterium bovis BCG. Infect Immun 2012; 81:373-80. [PMID: 23147035 DOI: 10.1128/iai.01031-12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intranasal vaccination stimulates formation of cyclooxygenases (COX) and release of prostaglandin E(2) (PGE(2)) by lung cells, including alveolar macrophages. PGE(2) plays complex pro- or anti-inflammatory roles in facilitating mucosal immune responses, but the relative contributions of COX-1 and COX-2 remain unclear. Previously, we found that Mycobacterium bovis BCG, a human tuberculosis vaccine, stimulated increased release of PGE(2) by macrophages activated in vitro; in contrast, intranasal BCG activated no PGE(2) release in the lungs, because COX-1 and COX-2 in alveolar macrophages were subcellularly dissociated from the nuclear envelope (NE) and catalytically inactive. This study tested the hypothesis that intranasal administration of BCG with cholera toxin (CT), a mucosal vaccine component, would shift the inactive, NE-dissociated COX-1/COX-2 to active, NE-associated forms. The results showed increased PGE(2) release in the lungs and NE-associated COX-2 in the majority of COX-2(+) macrophages. These COX-2(+) macrophages were the primary source of PGE(2) release in the lungs, since there was only slight enhancement of NE-associated COX-1 and there was no change in COX-1/COX-2 levels in alveolar epithelial cells following treatment with CT and/or BCG. To further understand the effect of CT, we investigated the timing of BCG versus CT administration for in vivo and in vitro macrophage activations. When CT followed BCG treatment, macrophages in vitro had elevated COX-2-mediated PGE(2) release, but macrophages in vivo exhibited less activation of NE-associated COX-2. Our results indicate that inclusion of CT in the intranasal BCG vaccination enhances COX-2-mediated PGE(2) release by alveolar macrophages and further suggest that the effect of CT in vivo is mediated by other lung cells.
Collapse
|
41
|
Morin S, Fischer R, Przybylski-Nicaise L, Bernard H, Corthier G, Rabot S, Wal JM, Hazebrouck S. Delayed bacterial colonization of the gut alters the host immune response to oral sensitization against cow's milk proteins. Mol Nutr Food Res 2012; 56:1838-47. [PMID: 23065810 DOI: 10.1002/mnfr.201200412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/03/2012] [Accepted: 09/07/2012] [Indexed: 11/07/2022]
Abstract
SCOPE Cow's milk allergy is the most prevalent food allergy in infants whose immune system development is critically stimulated during postnatal gut colonization by commensal bacteria. Allergenic potential of cow's milk β-lactoglobulin (BLG) and caseins (CAS) was investigated in germ-free (GF) BALB/c mice and in GF mice conventionalized (CVd) at 6 weeks of age. METHODS AND RESULTS Oral sensitization to cow's milk in the presence of cholera toxin led to higher BLG-specific IgE, IgG1, and IgG2a responses in GF mice than in conventional (CV) mice. No significant difference was observed for CAS-specific IgE responses although IgG1 responses to αS1- and κ-caseins were higher in GF mice than in CV mice. CVd mice, orally inoculated with fecal preparations from CV mice, also displayed biased antibody responses compared to CV mice. Secretion of Th2 cytokines by BLG- and CAS-reactivated splenocytes of CVd mice was similar to that of GF mice whereas cytokine production by reactivated cells from mesenteric lymph nodes of CVd mice was equivalent to that of CV mice. CONCLUSION Oral sensitization to BLG and CAS was differentially affected by the absence of gut microbiota and delayed bacterial colonization altered persistently the host immune response to oral sensitization against food antigens.
Collapse
Affiliation(s)
- Stéphanie Morin
- INRA, UR 496 - Immuno-Allergie Alimentaire, CEA/iBiTeC-S/SPI, CEA de Saclay, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Husain M, Golovan S, Rupa P, Mine Y, Boermans H, Karrow N. Spleen transcriptome profiles of BALB/c mouse in response to egg ovomucoid sensitisation and challenge. FOOD AGR IMMUNOL 2012. [DOI: 10.1080/09540105.2011.615062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
43
|
López-Expósito I, Chicón R, Belloque J, López-Fandiño R, Berin MC. In vivo methods for testing allergenicity show that high hydrostatic pressure hydrolysates of β-lactoglobulin are immunologically inert. J Dairy Sci 2012; 95:541-8. [PMID: 22281318 DOI: 10.3168/jds.2011-4646] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/25/2011] [Indexed: 11/19/2022]
Abstract
The major milk allergen β-lactoglobulin (β-LG) exhibits an enhanced susceptibility to proteolysis under high hydrostatic pressure and this may be an efficient method to produce hypoallergenic hydrolysates. The aim of this work was to evaluate the in vivo allergenicity of 3 β-LG hydrolysates produced under atmospheric pressure or high-pressure conditions. Hydrolysates were chosen based on previous experiments that showed that they provide a complete removal of intact β-LG but differed in vitro IgE-binding properties that could be traced to the peptide pattern. The ability to trigger systemic anaphylaxis was assessed using C3H/HeJ mice orally sensitized to β-LG. Outcome measures included symptom score, body temperature, serum mouse mast cell protease 1 (mMCP-1), and quantification of circulating basophils. Mast cell degranulation in vivo was assessed by passive cutaneous anaphylaxis. The 3 tested hydrolysates showed an abrogated allergenicity as revealed by the absence of anaphylactic symptoms and a decrease in body temperature. We demonstrated that the peptides present in the hydrolysates had lost their ability to cross-link 2 human IgE antibodies to induce mast cell degranulation, thus indicating that most of the peptides formed retain just one relevant IgE-binding epitope. The orally sensitized mouse model is a useful tool to address the in vivo allergenicity of novel milk formulas and demonstrates the safety of hydrolysates produced under high-pressure conditions.
Collapse
Affiliation(s)
- I López-Expósito
- Department of Bioactivity and Food Analysis, Instituto de Investigación en Ciencias de la Alimentación (CIAL), UAM-CSIC, Madrid, Spain.
| | | | | | | | | |
Collapse
|
44
|
Intestinal epithelial barrier dysfunction in food hypersensitivity. J Allergy (Cairo) 2011; 2012:596081. [PMID: 21912563 PMCID: PMC3170794 DOI: 10.1155/2012/596081] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/06/2011] [Accepted: 07/08/2011] [Indexed: 02/06/2023] Open
Abstract
Intestinal epithelial barrier plays a critical role in the maintenance of gut homeostasis by limiting the penetration of luminal bacteria and dietary allergens, yet allowing antigen sampling for the generation of tolerance. Undigested proteins normally do not gain access to the lamina propria due to physical exclusion by tight junctions at the cell-cell contact sites and intracellular degradation by lysosomal enzymes in enterocytes. An intriguing question then arises: how do macromolecular food antigens cross the epithelial barrier? This review discusses the epithelial barrier dysfunction in sensitized intestine with special emphasis on the molecular mechanism of the enhanced transcytotic rates of allergens. The sensitization phase of allergy is characterized by antigen-induced cross-linking of IgE bound to high affinity FcεRI on mast cell surface, leading to anaphylactic responses. Recent studies have demonstrated that prior to mast cell activation, food allergens are transported in large quantity across the epithelium and are protected from lysosomal degradation by binding to cell surface IgE and low-affinity receptor CD23/FcεRII. Improved immunotherapies are currently under study including anti-IgE and anti-CD23 antibodies for the management of atopic disorders.
Collapse
|
45
|
Morin S, Bernard H, Przybylski-Nicaise L, Corthier G, Rabot S, Wal JM, Hazebrouck S. Allergenic and immunogenic potential of cow's milk β-lactoglobulin and caseins evidenced without adjuvant in germ-free mice. Mol Nutr Food Res 2011; 55:1700-7. [PMID: 22045656 DOI: 10.1002/mnfr.201100024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 06/01/2011] [Accepted: 06/20/2011] [Indexed: 01/07/2023]
Abstract
SCOPE In most animal models of allergy, the development of an IgE response requires the use of an adjuvant. Germ-free (GF) mice exhibit Th2-polarized antibody responses combined with defective immunosuppressive mechanisms. The sensitizing potential of milk proteins was investigated in GF mice in the absence of adjuvant. METHODS AND RESULTS β-lactoglobulin (BLG) and whole casein (CAS) allergenicity was evaluated by means of intraperitoneal injections without adjuvant. Injections of BLG induced significant IgE and IgG1 responses in GF mice, while CAS injections provoked the production of IgG1 toward κ- and αS1-caseins. No significant antibody response was evidenced in conventional (CV) mice. After in vitro BLG-reactivation, IL-4, IL-5, IL-13 and IFN-γ productions by splenocytes were higher in GF mice than in CV mice. Heat-treatment decreased BLG allergenicity as indicated by the absence of IgE production in GF mice. However, heat-treatment increased protein immunogenicity and led to the production of anti-BLG and anti-κ-casein IgG1 in both GF and CV mice. This correlated with enhanced productions of IL-4, IL-5 and IL-13 in BLG-reactivated splenocytes from CV mice. CONCLUSION Gut colonization by commensal bacteria appeared then to significantly reduce the susceptibility of mice toward the intrinsic allergenic and immunogenic potential of milk proteins.
Collapse
Affiliation(s)
- Stéphanie Morin
- INRA, UR 496, Immuno-Allergie Alimentaire, CEA de Saclay, Gif-sur-Yvette, France.
| | | | | | | | | | | | | |
Collapse
|
46
|
Schiavi E, Barletta B, Butteroni C, Corinti S, Boirivant M, Di Felice G. Oral therapeutic administration of a probiotic mixture suppresses established Th2 responses and systemic anaphylaxis in a murine model of food allergy. Allergy 2011; 66:499-508. [PMID: 21058959 DOI: 10.1111/j.1398-9995.2010.02501.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND No effective treatment is available for food allergy and its primary management still consists of avoiding relevant allergens. Probiotics are claimed to beneficially affect the immune system. We sought to investigate the therapeutic potential of VSL#3 probiotic mixture on specific immune responses and anaphylactic reaction induced in mice by the major food allergen shrimp tropomyosin (ST). METHODS The cytokine production by spleen cell from ST-sensitized mice upon allergen re-stimulation in the presence of VSL#3 was analysed. Next, the effects of oral administration of VSL#3 on allergen-induced anaphylaxis and Th2 response in the murine model of food allergy to ST was investigated by evaluating symptom score and histamine content in the faeces after allergen challenge, antibody response in serum and faeces, and cytokine and transcription factor expression in the jejunum. RESULTS The in vitro studies on mouse spleen cells indicates that the VSL#3 preparation has the capacity to shift a polarized Th2 response to a Th1/T regulatory-type profile. Oral therapeutic administration of VSL#3 to ST-sensitized mice significantly reduces symptom score and histamine release in the faeces following allergen challenge, as well as specific IgE response. In the jejunum, IL-4, IL-5 and IL-13 tissue content was significantly reduced, whereas FOXP3 and IL-27 mRNA expression, IL-10, TGF-β and IFN-γ tissue content were up-regulated. CONCLUSIONS Oral therapeutic treatment with the probiotic mixture VSL#3 is effective in redirecting allergen-specific Th2-polarized immune responses towards Th1-T regulatory responses and in the protection against anaphylactic reactions induced by the allergen in a murine model of food allergy.
Collapse
Affiliation(s)
- E Schiavi
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | |
Collapse
|
47
|
Adel-Patient K, Guimaraes VD, Paris A, Drumare MF, Ah-Leung S, Lamourette P, Nevers MC, Canlet C, Molina J, Bernard H, Créminon C, Wal JM. Immunological and metabolomic impacts of administration of Cry1Ab protein and MON 810 maize in mouse. PLoS One 2011; 6:e16346. [PMID: 21298004 PMCID: PMC3029317 DOI: 10.1371/journal.pone.0016346] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 12/13/2010] [Indexed: 12/24/2022] Open
Abstract
We have investigated the immunological and metabolomic impacts of Cry1Ab administration to mice, either as a purified protein or as the Cry1Ab-expressing genetically modified (GM) MON810 maize. Humoral and cellular specific immune responses induced in BALB/cJ mice after intra-gastric (i.g.) or intra-peritoneal (i.p.) administration of purified Cry1Ab were analyzed and compared with those induced by proteins of various immunogenic and allergic potencies. Possible unintended effects of the genetic modification on the pattern of expression of maize natural allergens were studied using IgE-immunoblot and sera from maize-allergic patients. Mice were experimentally sensitized (i.g. or i.p. route) with protein extracts from GM or non-GM maize, and then anti-maize proteins and anti-Cry1Ab–induced immune responses were analyzed. In parallel, longitudinal metabolomic studies were performed on the urine of mice treated via the i.g. route. Weak immune responses were observed after i.g. administration of the different proteins. Using the i.p. route, a clear Th2 response was observed with the known allergenic proteins, whereas a mixed Th1/Th2 immune response was observed with immunogenic protein not known to be allergenic and with Cry1Ab. This then reflects protein immunogenicity in the BALB/c Th2-biased mouse strain rather than allergenicity. No difference in natural maize allergen profiles was evidenced between MON810 and its non-GM comparator. Immune responses against maize proteins were quantitatively equivalent in mice treated with MON810 vs the non-GM counterpart and no anti-Cry1Ab–specific immune response was detected in mice that received MON810. Metabolomic studies showed a slight “cultivar” effect, which represented less than 1% of the initial metabolic information. Our results confirm the immunogenicity of purified Cry1Ab without evidence of allergenic potential. Immunological and metabolomic studies revealed slight differences in mouse metabolic profiles after i.g. administration of MON810 vs its non-GM counterpart, but no significant unintended effect of the genetic modification on immune responses was seen.
Collapse
|
48
|
Cecchi R. Estimating wound age: looking into the future. Int J Legal Med 2010; 124:523-36. [DOI: 10.1007/s00414-010-0505-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 08/10/2010] [Indexed: 12/16/2022]
|
49
|
Scientific Opinion on the assessment of allergenicity of GM plants and microorganisms and derived food and feed. EFSA J 2010. [DOI: 10.2903/j.efsa.2010.1700] [Citation(s) in RCA: 243] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
50
|
Ladics G, Knippels L, Penninks A, Bannon G, Goodman R, 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. [DOI: 10.1016/j.yrtph.2009.09.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 08/31/2009] [Accepted: 09/24/2009] [Indexed: 01/06/2023]
|