1
|
Liao Y, Gao IH, Kusakabe T, Lin WY, Grier A, Pan X, Morzhanaeva O, Shea TP, Yano H, Karo-Atar D, Olsen KA, Oh JH, Vandegrift KJ, King IL, Cuomo CA, Artis D, Rehermann B, Lipman N, Iliev ID. Fungal symbiont transmitted by free-living mice promotes type 2 immunity. Nature 2024; 636:697-704. [PMID: 39604728 PMCID: PMC11733984 DOI: 10.1038/s41586-024-08213-2] [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: 04/28/2023] [Accepted: 10/14/2024] [Indexed: 11/29/2024]
Abstract
The gut mycobiota is crucial for intestinal homeostasis and immune function1. Yet its variability and inconsistent fungal colonization of laboratory mice hinders the study of the evolutionary and immune processes that underpin commensalism2,3. Here, we show that Kazachstania pintolopesii is a fungal commensal in wild urban and rural mice, with an exceptional ability to colonize the mouse gastrointestinal tract and dominate the gut mycobiome. Kazachstania pintolopesii colonization occurs in a bacteria-independent manner, results in enhanced colonization resistance to other fungi and is shielded from host immune surveillance, allowing commensal presence. Following changes in the mucosal environment, K. pintolopesii colonization triggers a type 2 immune response in mice and induces gastrointestinal eosinophilia. Mechanistically, we determined that K. pintolopesii activates type 2 immunity via the induction of epithelial IL-33 and downstream IL-33-ST2 signalling during mucus fluctuations. Kazachstania pintolopesii-induced type 2 immunity enhanced resistance to helminth infections or aggravated gastrointestinal allergy in a context-dependent manner. Our findings indicate that K. pintolopesii is a mouse commensal and serves as a valuable model organism for studying gut fungal commensalism and immunity in its native host. Its unnoticed presence in mouse facilities highlights the need to evaluate its influence on experimental outcomes and phenotypes.
Collapse
Affiliation(s)
- Yun Liao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Iris H Gao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Takato Kusakabe
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Woan-Yu Lin
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alexander Grier
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Xiangyu Pan
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Olga Morzhanaeva
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Terrance P Shea
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hiroshi Yano
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Kaitlin A Olsen
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Ji Hoon Oh
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Kurt J Vandegrift
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Neil Lipman
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
2
|
Bavbek S, Kepil Özdemir S, Bonadonna P, Atanaskovic-Markovic M, Barbaud A, Brockow K, Laguna Martinez J, Nakonechna A, Pagani M, Arcolacı A, Lombardo C, Torres MJ. Hypersensitivity reactions to proton pump inhibitors. An EAACI position paper. Allergy 2024; 79:552-564. [PMID: 38013608 DOI: 10.1111/all.15961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023]
Abstract
Proton pump inhibitors (PPIs) are invaluable therapeutic options in a variety of dyspeptic diseases. In addition to their well-known risk profile, PPI consumption is related to food and environmental allergies, dysbiosis, osteoporosis, as well as immediate and delayed hypersensitivity reactions (HSRs). The latter, although a rare event, around 1%-3%, due to the extraordinarily high rate of prescription and consumption of PPIs are related to a substantial risk. In this Position Paper, we provide clinicians with practical evidence-based recommendations for the diagnosis and management of HSRs to PPIs. Furthermore, the unmet needs proposed in the document aim to stimulate more in-depth investigations in the topic.
Collapse
Affiliation(s)
- Sevim Bavbek
- Division of Immunology and Allergy, Department of Chest Diseases, School of Medicine, Ankara University, Ankara, Turkey
| | - Seçil Kepil Özdemir
- Department of Chest Diseases, Allergy and Immunology Unit, İzmir Faculty of Medicine, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, University of Health Sciences, İzmir, Turkey
| | | | - Marina Atanaskovic-Markovic
- Department of Allergology and Pulmonology, Faculty of Medicine, University of Belgrade, University Children's Hospital, Belgrade, Serbia
| | - Annick Barbaud
- Département de dermatologie et allergologie, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, AP-HP, Sorbonne Université, Hôpital Tenon, Paris, France
| | - Knut Brockow
- Department of Dermatology and Allergy Biederstein, Faculty of Medicine, Technical University of Munich, Munich, Germany
| | - Jose Laguna Martinez
- Allergy Unit, Allergo-Anaesthesia Unit, Faculty of Medicine, Hospital Central de la Cruz Roja, Alfonso X El Sabio University, Madrid, Spain
| | - Alla Nakonechna
- Allergy and Clinical Immunology Department, University of Liverpool, Royal Preston Hospital, Lancashire Teaching Hospitals, NHS Foundation Trust, Liverpool, UK
| | - Mauro Pagani
- Medicine Department, Medicine Ward Mantova Hospital, ASST di Mantova, Mantova, Italy
| | | | - Carla Lombardo
- Division of Dermatology and Allergy, APSS - Trento Hospital, Trento, Italy
| | - Maria J Torres
- Allergy Unit, Regional University Hospital of Malaga, IBIMA-UMA-ARADyAL, Malaga, Spain
| |
Collapse
|
3
|
Bartra J, Turner PJ, Muñoz-Cano RM. Cofactors in food anaphylaxis in adults. Ann Allergy Asthma Immunol 2023; 130:733-740. [PMID: 36958469 DOI: 10.1016/j.anai.2023.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
Around 25% to 50% of food-induced allergic reactions in adults cause anaphylaxis, and epidemiologic evidence suggests that food is the most common cause of anaphylaxis. Reaction severity is unpredictable, and patients will often experience reactions of variable severity, even to an identical exposure (both dose and allergen). A common explanation for this phenomenon has been the impact of "cofactors"-factors that might contribute to reaction severity independent of the allergen exposure. Cofactors can influence reaction severity in 2 ways: either by reducing the reaction threshold (ie, the dose needed to trigger any symptoms) so that patients have no symptoms in the absence of the cofactor and only react with the cofactor present, or by increasing reaction severity such that individuals have only mild symptoms in the absence of the cofactor, but a more severe reaction when the cofactor is present. Indeed, the same patient may have reactions with different cofactors or even need more than one cofactor to develop a severe reaction. Cofactors reportedly play a role in approximately 30% of anaphylaxis reactions in adults. Exercise, nonsteroidal, anti-inflammatory drugs, alcohol, and sleep deprivation are the most frequent cofactors reported. Routine evaluation of the possible involvement of cofactors is essential in managing patients with food anaphylaxis: in patients with a suggestive history but a negative oral food challenge, cofactors should be taken into account to provide appropriate advice to reduce the risk of future anaphylaxis.
Collapse
Affiliation(s)
- Joan Bartra
- Department of Allergy, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), RETIC ARADyAL, RICORs REI, Barcelona, Spain
| | - Paul J Turner
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Rosa M Muñoz-Cano
- Department of Allergy, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), RETIC ARADyAL, RICORs REI, Barcelona, Spain
| |
Collapse
|
4
|
Wang K, Crevel RWR, Mills ENC. An in vitro protocol to characterise the resistance of food proteins to intestinal digestion. Food Chem Toxicol 2023; 173:113590. [PMID: 36584934 DOI: 10.1016/j.fct.2022.113590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022]
Abstract
In vitro digestion tests provide data on the form in which dietary proteins maybe presented to the gut mucosal immune system, one of many strands of evidence used in allergenicity risk assessment. A 96-well plate format in vitro intestinal digestion protocol has been developed with a high and low enzyme activity test executed at pH 6.5 and 8.0. It was applied to the systematic analysis of test proteins (including six allergens and one non-allergenic comparator) which were either completely resistant to pepsinolysis or gave rise to large persistent fragments following in vitro gastric digestion. Digestion was monitored using SDS-PAGE and densitometry. Proteins resistant to pepsin were also resistant to intestinal digestion irrespective of the protocol applied and gave rise to large persistent digestion fragments. In contrast persistent fragments from pepsin digestion were readily digested. Bile salts enhanced the digestibility of two highly resistant proteins, lysozyme ad β-lactoglobulin, changing the rank order of protein digestibility. Intestinal digestion tests that include bile salts provide a more physiologically relevant system for future investigation into how digestion products may influence the balance between tolerance and sensitization - and hence contribute to future development of a more effective allergenicity risk assessment process.
Collapse
Affiliation(s)
- Kai Wang
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Institute of Biotechnology, 131 Princess Street, Manchester, M1 7DN, UK.
| | - Rene W R Crevel
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Institute of Biotechnology, 131 Princess Street, Manchester, M1 7DN, UK; René Crevel Consulting Ltd, 3 Woodlands Close, Cople, Bedford, MK44 3UE, UK.
| | - E N Clare Mills
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Institute of Biotechnology, 131 Princess Street, Manchester, M1 7DN, UK; School of Biosciences and Medicine, The University of Surrey, Guildford, UK.
| |
Collapse
|
5
|
The Interaction of Food Allergy and Diabetes: Food Allergy Effects on Diabetic Mice by Intestinal Barrier Destruction and Glucagon-like Peptide 1 Reduction in Jejunum. Foods 2022; 11:foods11233758. [PMID: 36496564 PMCID: PMC9741085 DOI: 10.3390/foods11233758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The increase in food allergies and diabetes leads to the assumption that they are related. This study aimed to (1) verify the interaction between food allergy and diabetes and (2) explore the potential mechanisms by which food allergy promotes diabetes. Female BALB/c mice were grouped into a control group (CK), an ovalbumin-sensitized group (OVA), a diabetes group (STZ), and a diabetic allergic group (STZ + OVA) (Mice were modeled diabetes with STZ first, then were given OVA to model food allergies), and an allergic diabetic group (OVA + STZ) (Mice were modeled food allergies with OVA first, then were given STZ to model diabetes). The results showed that OVA + STZ mice exhibited a more serious Th2 humoral response, and they were more susceptible to diabetes. Furthermore, when the OVA + STZ mice were in the sensitized state, the intestinal barrier function was severely impaired, and mast cell activation was promoted. Moreover, we found that the effect of food allergy on diabetes is related to the inhibition of GLP-1 secretion and the up-regulation of the PI3K/Akt/mTOR/NF-κB P65 signaling pathway in the jejunum. Overall, our results suggest that food allergies have interactions with diabetes, which sheds new light on the importance of food allergies in diabetes.
Collapse
|
6
|
Brishti A, Germundson-Hermanson DL, Smith NA, Kearney AE, Warda Y, Nagamoto-Combs K. Asymptomatic sensitization to a cow's milk protein induces sustained neuroinflammation and behavioral changes with chronic allergen exposure. FRONTIERS IN ALLERGY 2022; 3:870628. [PMID: 36157272 PMCID: PMC9490182 DOI: 10.3389/falgy.2022.870628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Mouse models of food allergy have contributed to our understanding of various aspects of the disease, including susceptibilities, symptom spectra, cellular mechanisms, and therapeutic approaches. Previously, we used a mouse model of non-anaphylactic cow's milk allergy (CMA) and investigated sex- and strain-dependent differences in immunological, neurological, and behavioral sequelae. We showed that male C57BL/6J mice sensitized to a bovine whey protein, β-lactoglobulin (BLG; Bos d 5), exhibited anxiety- and depression-like behavior upon acute allergen challenge. Systemic levels of BLG-specific immunoglobulins, cytokines and chemokines were also elevated in the sensitized mice. Furthermore, neuroinflammation and intestinal dysbiosis were evident as the possible causes of the altered behavior. To assess whether frequent allergen exposure influences CMA-associated pathologies over an extended period in this subclinical model, we placed BLG-sensitized mice on a whey protein (WP)-containing or whey-free control (CTL) diet for 3 months. As expected, allergen-specific IgE was significantly elevated in the plasma after completing the 5-week sensitization phase. However, the IgE levels declined in both diet groups after 3 months. In contrast, allergen-specific IgG1 stayed elevated in sensitized mice with the CTL diet, and the WP diet to a lesser extent. Interestingly, BLG-sensitized mice on the WP diet exhibited anxiety-like behavior and a trend toward spatial memory decline compared to the sham or the sensitized mice on the CTL diet. Moreover, increased immunoreactivities for GFAP and Iba1 and elevated levels of CXCL13 and CCL12, the chemokines involved in central leukocyte recruitment and other neurological diseases, were also observed in the brain. We demonstrated that sensitization to the whey protein, particularly with continuous allergen exposure, resulted in persistent neuroinflammation and associated behavioral changes despite lowered allergen-specific immunoglobulin levels. These results suggested that continuous consumption of the offending allergen may lead to adverse consequences in the brain even after desensitization.
Collapse
Affiliation(s)
- Afrina Brishti
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Danielle L. Germundson-Hermanson
- Clinical and Translational Science Graduate Program, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Nicholas A. Smith
- Clinical and Translational Science Graduate Program, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Angela E. Kearney
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Yassmine Warda
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Kumi Nagamoto-Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
7
|
Molecular dynamics simulations of ovalbumin adsorption at squalene/water interface. Chin J Chem Eng 2022. [DOI: 10.1016/j.cjche.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
8
|
Shin M. Food allergies and food-induced anaphylaxis: role of cofactors. Clin Exp Pediatr 2021; 64:393-399. [PMID: 33181008 PMCID: PMC8342881 DOI: 10.3345/cep.2020.01088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/08/2020] [Indexed: 11/29/2022] Open
Abstract
Food allergies and food-induced anaphylaxis are important health problems. Several cofactors modulating the onset of anaphylaxis have been identified. In the presence of cofactors, allergic reactions may be induced at lower doses of food allergens and/or become severe. Exercise and concomitant infections are well-documented cofactors of anaphylaxis in children. Other factors such as consumption of nonsteroidal anti-inflammatory drugs, alcohol ingestion, and stress have been reported. Cofactors reportedly play a role in approximately 30% of anaphylactic reactions in adults and 14%-18.3% in children. Food-dependent exercise-induced anaphylaxis (FDEIA) is the best-studied model of cofactor-induced anaphylaxis. Wheat-dependent exercise-induced anaphylaxis, the most common FDEIA condition, has been studied the most. The mechanisms of action of cofactors have not yet been fully identified. This review aims to educate clinicians on recent developments in the role of cofactors and highlight the importance of recognizing cofactors in food allergies and food-induced anaphylaxis.
Collapse
Affiliation(s)
- Meeyong Shin
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| |
Collapse
|
9
|
Gastric Enzyme Supplementation Inhibits Food Allergy in a BALB/c Mouse Model. Nutrients 2021; 13:nu13030738. [PMID: 33652629 PMCID: PMC7996948 DOI: 10.3390/nu13030738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Impaired gastric digestion due to suppressed gastric acidity enhances the risk for food allergy development. In the current study, we aimed to evaluate the impact of a supported gastric digestion via application of a pharmaceutical gastric enzyme solution (GES) on food allergy development and allergic reactions in a BALB/c mouse model. The ability of the GES to restore hypoacidic conditions was tested in mice treated with gastric acid suppression medication. To evaluate the impact on allergic symptoms, mice were orally sensitized with ovalbumin (OVA) under gastric acid suppression and subjected to oral challenges with or without GES. The immune response was evaluated by measurement of antibody titers, cytokine levels, mucosal allergy effector cell influx and regulatory T-cell counts. Clinical response was objectified by core body temperature measurements after oral OVA challenge. Supplementation of GES transiently restored physiological pH levels in the stomach after pharmaceutical gastric acid suppression. During oral sensitization, supplementation of gastric enzymes significantly reduced systemic IgE, IgG1 and IgG2a levels and allergic symptoms. In food allergic mice, clinical symptoms were reduced by co-administration of the gastric enzyme solution. Support of gastric digestion efficiently prevents food allergy induction and alleviates clinical symptoms in our food allergy model.
Collapse
|
10
|
Samadi N, Klems M, Heiden D, Bauer R, Kitzmüller C, Weidmann E, Ret D, Ondracek AS, Duschl A, Horejs‐Hoeck J, Untersmayr E. Nitrated food proteins induce a regulatory immune response associated with allergy prevention after oral exposure in a Balb/c mouse food allergy model. Allergy 2020; 75:412-422. [PMID: 31444907 PMCID: PMC7064937 DOI: 10.1111/all.14030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/09/2019] [Accepted: 07/23/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Food allergy is associated with a high personal health and economic burden. For immunomodulation toward tolerance, food compounds could be chemically modified, for example, by posttranslational protein nitration, which also occurs via diet-derived nitrating agents in the gastrointestinal tract. OBJECTIVE We sought to analyze the effect of pretreatment with nitrated food proteins on the immune response in a mouse food allergy model and on human monocyte-derived dendritic cells (moDCs) and PBMCs. METHODS The model allergen ovalbumin (OVA) was nitrated in different nitration degrees, and the secondary structures of proteins were determined by circular dichroism (CD). Allergy-preventive treatment with OVA, nitrated OVA (nOVA), and maximally nitrated OVA (nOVAmax) were performed before mice were immunized with or without gastric acid-suppression medication. Antibody levels, regulatory T-cell (Treg) numbers, and cytokine levels were evaluated. Human moDCs or PBMCs were incubated with proteins and evaluated for expression of surface markers, cytokine production, and proliferation of Th2 as well as Tregs. RESULTS In contrast to OVA and nOVA, the conformation of nOVAmax was substantially changed. nOVAmax pretreated mice had decreased IgE as well as IgG1 and IgG2a levels and Treg numbers were significantly elevated, while cytokine levels remained at baseline level. nOVAmax induced a regulatory DC phenotype evidenced by a decrease of the activation marker CD86 and an increase in IL-10 production and was associated with a higher proliferation of memory Tregs. CONCLUSION Oral pretreatment with highly nitrated proteins induces a tolerogenic immune response in the food allergy model and in human immune cells.
Collapse
Affiliation(s)
- Nazanin Samadi
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Martina Klems
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Denise Heiden
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Renate Bauer
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Claudia Kitzmüller
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Eleonore Weidmann
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Davide Ret
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Anna S. Ondracek
- Anna Spiegel Research Center Medical University of Vienna Vienna Austria
| | - Albert Duschl
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Eva Untersmayr
- Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| |
Collapse
|
11
|
Castan L, Bøgh KL, Maryniak NZ, Epstein MM, Kazemi S, O'Mahony L, Bodinier M, Smit JJ, Bilsen JHM, Blanchard C, Głogowski R, Kozáková H, Schwarzer M, Noti M, Wit N, Bouchaud G, Bastiaan‐Net S. Overview of in vivo and ex vivo endpoints in murine food allergy models: Suitable for evaluation of the sensitizing capacity of novel proteins? Allergy 2020; 75:289-301. [PMID: 31187876 PMCID: PMC7065134 DOI: 10.1111/all.13943] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/12/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Significant efforts are necessary to introduce new dietary protein sources to feed a growing world population while maintaining food supply chain sustainability. Such a sustainable protein transition includes the use of highly modified proteins from side streams or the introduction of new protein sources that may lead to increased clinically relevant allergic sensitization. With food allergy being a major health problem of increasing concern, understanding the potential allergenicity of new or modified proteins is crucial to ensure public health protection. The best predictive risk assessment methods currently relied on are in vivo models, making the choice of endpoint parameters a key element in evaluating the sensitizing capacity of novel proteins. Here, we provide a comprehensive overview of the most frequently used in vivo and ex vivo endpoints in murine food allergy models, addressing their strengths and limitations for assessing sensitization risks. For optimal laboratory‐to‐laboratory reproducibility and reliable use of predictive tests for protein risk assessment, it is important that researchers maintain and apply the same relevant parameters and procedures. Thus, there is an urgent need for a consensus on key food allergy parameters to be applied in future food allergy research in synergy between both knowledge institutes and clinicians.
Collapse
Affiliation(s)
| | - Katrine L. Bøgh
- National Food Institute Technical University of Denmark Kgs. Lyngby Denmark
| | | | - Michelle M. Epstein
- Experimental Allergy Laboratory, Department of Dermatology Medical University of Vienna Vienna Austria
| | - Sahar Kazemi
- Experimental Allergy Laboratory, Department of Dermatology Medical University of Vienna Vienna Austria
| | - Liam O'Mahony
- Department of Medicine, APC Microbiome Ireland National University of Ireland Cork Ireland
- Department of Microbiology, APC Microbiome Ireland National University of Ireland Cork Ireland
| | | | - Joost J. Smit
- Institute for Risk Assessment Sciences Utrecht University Utrecht The Netherlands
| | | | | | - Robert Głogowski
- Department of Animal Breeding and Production Warsaw University of Life Sciences Warsaw Poland
| | - Hana Kozáková
- Institute of Microbiology Czech Academy of Sciences Nový Hrádek Czech Republic
| | - Martin Schwarzer
- Institute of Microbiology Czech Academy of Sciences Nový Hrádek Czech Republic
| | - Mario Noti
- Institute of Pathology University of Bern Bern Switzerland
| | - Nicole Wit
- Wageningen Food and Biobased Research Wageningen The Netherlands
| | | | | |
Collapse
|
12
|
Immune Effects of the Nitrated Food Allergen Beta-Lactoglobulin in an Experimental Food Allergy Model. Nutrients 2019; 11:nu11102463. [PMID: 31618852 PMCID: PMC6835712 DOI: 10.3390/nu11102463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/08/2019] [Accepted: 10/12/2019] [Indexed: 12/18/2022] Open
Abstract
Food proteins may get nitrated by various exogenous or endogenous mechanisms. As individuals might get recurrently exposed to nitrated proteins via daily diet, we aimed to investigate the effect of repeatedly ingested nitrated food proteins on the subsequent immune response in non-allergic and allergic mice using the milk allergen beta-lactoglobulin (BLG) as model food protein in a mouse model. Evaluating the presence of nitrated proteins in food, we could detect 3-nitrotyrosine (3-NT) in extracts of different foods and in stomach content extracts of non-allergic mice under physiological conditions. Chemically nitrated BLG (BLGn) exhibited enhanced susceptibility to degradation in simulated gastric fluid experiments compared to untreated BLG (BLGu). Gavage of BLGn to non-allergic animals increased interferon-γ and interleukin-10 release of stimulated spleen cells and led to the formation of BLG-specific serum IgA. Allergic mice receiving three oral gavages of BLGn had higher levels of mouse mast cell protease-1 (mMCP-1) compared to allergic mice receiving BLGu. Regardless of the preceding immune status, non-allergic or allergic, repeatedly ingested nitrated food proteins seem to considerably influence the subsequent immune response.
Collapse
|
13
|
Identification and characterization of major IgE binding of purified allergenic protein (11 kDa) from Buchanania lanzan. Food Res Int 2019; 125:108640. [PMID: 31554061 DOI: 10.1016/j.foodres.2019.108640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
Abstract
Tree nut along with peanut are among the most potent food allergens, responsible for frequently inducing the IgE-mediated hypersensitivity reaction. Our aim was identification, purification of Buchanania lanzan (Bl-11 kDa) protein along with characterization and assessment of allergenic potential of clinically relevant allergen. Further study was executed in clinical samples of sensitive patients, BALB/c mice, and in-vitro. A major IgE binding 11-kDa protein from Buchanania lanzan was purified by anion exchange chromatography, reverse phase high pressure liquid chromatography (RP-HPLC) and characterized using peptide mass fingerprinting (PMF). Buchanania lanzan (Bl-11 kDa) protein shows the pepsin resistance and depicts IgE interacting capacity to Buchanania lanzan allergic patient's sera as well as sensitized mice sera. It also showed increase in the allergic mediator's like IgE, IgG1, histamine levels in sensitized mice sera. Further study was carried out in-vitro (RBL-2H3 cells) and increased release mast cell degranulation mediators such as β-hexosaminidase, histamine, CysL and PGD2 in the culture supernatant was found. The activation of Th2 cytokines/transcription factors and expression of molecular markers in the downstream of mast cell signaling were up-regulated while the Th1 transcriptional factor (T-bet) was decreased in Bl-11 kDa protein treated mice. Conclusively, our study demonstrates Buchanania lanzan purified protein to be potential allergen that may generate an allergic reaction in sensitized individuals, and one of the most important IgE binding protein responsible for its allergenicity.
Collapse
|
14
|
Country-wide medical records infer increased allergy risk of gastric acid inhibition. Nat Commun 2019; 10:3298. [PMID: 31363098 PMCID: PMC6667461 DOI: 10.1038/s41467-019-10914-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric acid suppression promotes allergy in mechanistic animal experiments and observational human studies, but whether gastric acid inhibitors increase allergy incidence at a population level remains uncharacterized. Here we aim to assess the use of anti-allergic medication following prescription of gastric acid inhibitors. We analyze data from health insurance records covering 97% of Austrian population between 2009 and 2013 on prescriptions of gastric acid inhibitors, anti-allergic drugs, or other commonly prescribed (lipid-modifying and antihypertensive) drugs as controls. Here we show that rate ratios for anti-allergic following gastric acid-inhibiting drug prescriptions are 1.96 (95%CI:1.95–1.97) and 3.07 (95%-CI:2.89–3.27) in an overall and regional Austrian dataset. These findings are more prominent in women and occur for all assessed gastric acid-inhibiting substances. Rate ratios increase from 1.47 (95%CI:1.45–1.49) in subjects <20 years, to 5.20 (95%-CI:5.15–5.25) in > 60 year olds. We report an epidemiologic relationship between gastric acid-suppression and development of allergic symptoms. Gastric acid inhibitors promote experimental allergy in animals, and have been linked to allergy risk in observational human studies. Here the authors show in a country-wide medical record analysis that allergy development risk is doubled in gastric acid inhibitor users, and is higher in women and in older age.
Collapse
|
15
|
Abstract
Recent studies have reported a higher prevalence of eosinophilic esophagitis in children with esophageal atresia. Under recognition of eosinophilic esophagitis in these patients may lead to excessive use of antireflux therapy and an escalation of interventions, including fundoplication, as symptoms may be attributed to gastroesophageal reflux disease. In addition, long-term untreated eosinophilic esophagitis may lead to recurrent strictures due to transmural esophageal inflammation, necessitating repeated dilatations. Eosinophilic esophagitis should be considered when children with esophageal atresia show persistent symptoms on standard antireflux treatment, increasing dysphagia, and recurrent strictures. Treatment has been found to not only significantly reduce intraepithelial eosinophil count, but also to improve symptoms, and to lower the occurrence of strictures and the need for dilatations. Future prospective studies are warranted in this area.
Collapse
Affiliation(s)
- Usha Krishnan
- Department of Pediatric Gastroenterology, Sydney Children's Hospital, Sydney, NSW, Australia.,School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
16
|
Weidmann E, Samadi N, Klems M, Heiden D, Seppova K, Ret D, Untersmayr E. Mouse Chow Composition Influences Immune Responses and Food Allergy Development in a Mouse Model. Nutrients 2018; 10:nu10111775. [PMID: 30453476 PMCID: PMC6266012 DOI: 10.3390/nu10111775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Our diet is known to substantially influence the immune response not only by support of mucosal barriers but also via direct impact on immune cells. Thus, it was of great interest to compare the immunological effect of two mouse chows with substantial differences regarding micro-, macronutrient, lipid and vitamin content on the food allergic response in our previously established mouse model. As the two mouse chows of interest, we used a soy containing feed with lower fatty acid (FA) amount (soy-containing feed) and compared it to a soy free mouse chow (soy-free feed) in an established protocol of oral immunizations with Ovalbumin (OVA) under gastric acid suppression. In the animals receiving soy-containing feed, OVA-specific IgE, IgG1, IgG2a antibody levels were significantly elevated and food allergy was evidenced by a drop of body temperature after oral immunizations. In contrast, mice on soy-free diet had significantly higher levels of IL-10 and were protected from food allergy development. In conclusion, soy-containing feed was auxiliary during sensitizations, while soy-free feed supported oral tolerance development and food allergy prevention.
Collapse
Affiliation(s)
- Eleonore Weidmann
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Nazanin Samadi
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Martina Klems
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Denise Heiden
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Klara Seppova
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Davide Ret
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
- Division of Macromolecular Chemistry, Institute of Applied Synthetic Chemistry, Vienna University of Technology, 1060 Vienna, Austria.
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
17
|
The Effect of Digestion and Digestibility on Allergenicity of Food. Nutrients 2018; 10:nu10091129. [PMID: 30134536 PMCID: PMC6164088 DOI: 10.3390/nu10091129] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/03/2018] [Accepted: 08/13/2018] [Indexed: 01/08/2023] Open
Abstract
Food allergy prevalence numbers are still on the rise. Apart from environmental influences, dietary habits, food availability and life-style factors, medication could also play a role. For immune tolerance of food, several contributing factors ensure that dietary compounds are immunologically ignored and serve only as source for energy and nutrient supply. Functional digestion along the gastrointestinal tract is essential for the molecular breakdown and a prerequisite for appropriate uptake in the intestine. Digestion and digestibility of carbohydrates and proteins thus critically affect the risk of food allergy development. In this review, we highlight the influence of amylases, gastric acid- and trypsin-inhibitors, as well as of food processing in the context of food allergenicity.
Collapse
|
18
|
Yu JE, Mallapaty A, Miller RL. It's not just the food you eat: Environmental factors in the development of food allergies. ENVIRONMENTAL RESEARCH 2018; 165:118-124. [PMID: 29689456 DOI: 10.1016/j.envres.2018.03.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
The dramatic rise in the prevalence of food allergy and food allergy-associated anaphylaxis in the past few decades has fueled investigative interest into understanding this puzzling trend. Here, we review the question as to whether important external environmental determinants beyond dietary habits and exposure to food allergens are involved. This review will summarize our current understanding of these environment determinants, derived from the latest experimental and epidemiological research. Specifically, we will review the role of exposures that affect skin barrier function, development of a diverse microbiome, and food processing. Additional exposures of concern are insufficient sunlight, endocrine disrupting chemicals and pesticides, and use of specific pharmaceutical agents that may drive or modify the risk for food allergy. Despite limitations in the quantity and quality of research to date, many new epidemiological associations and experimental data in support of this paradigm have emerged.
Collapse
Affiliation(s)
- Joyce E Yu
- Division of Allergy, Immunology, Rheumatology, Department of Pediatrics, PH8E-101, Columbia University Medical Center, 630 W. 168th St., New York, NY 10032, USA
| | - Anu Mallapaty
- Division of Allergy, Immunology, Rheumatology, Department of Pediatrics, PH8E-101, Columbia University Medical Center, 630 W. 168th St., New York, NY 10032, USA
| | - Rachel L Miller
- Division of Allergy, Immunology, Rheumatology, Department of Pediatrics, PH8E-101, Columbia University Medical Center, 630 W. 168th St., New York, NY 10032, USA; Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, PH8E-101, Columbia University Medical Center, 630 W. 168th St., New York, NY 10032, USA; Department of Environmental Health Sciences, Mailman School of Public Health, 722 W. 168th St., New York, NY 10032, USA.
| |
Collapse
|
19
|
Abstract
Immunoglobulin E-mediated food allergy is rapidly developing into a global health problem. Publicly available therapeutic intervention strategies are currently restricted to allergen avoidance and emergency treatments. To gain a better understanding of the disease pathophysiology so that new therapies can be developed, major research efforts have been put into studying food allergy in mice. Animal models should reflect the human pathology as closely as possible to allow for a rapid translation of basic science observations to the bedside. In this regard, experimental models of food allergy provide significant challenges for research because of discrepancies between the presentation of disease in humans and mice. The goal of this review is to give a summary of commonly used murine disease models and to discuss how they relate to the human condition. We will focus on epicutaneous sensitization models, on mouse strains that sensitize spontaneously to food as seen in humans, and on models in humanized animals. In summary, expanding the research toolbox of experimental food allergy provides an important step toward closing gaps in our understanding of the derailing immune mechanism that underlies the human disease. The availability of additional experimental models will provide exciting opportunities to discover new intervention points for the treatment of food allergies. (Cell Mol Gastroenterol Hepatol 2018;x:x).
Collapse
Key Words
- Allergen Challenge
- Allergen Sensitization
- Anaphylaxis
- EPIT, epicutaneous immunotherapy
- Epictutaneous Sensitization
- FCER1A, high-affinity immunoglobulin epsilon receptor subunit alpha
- FCERIA
- FcεRI, high-affinity immunoglobulin E receptor
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HSC, hematopoietic stem cell
- Humanized Model
- IL, interleukin
- Ig, immunoglobulin
- IgE
- LCT, long chain triglycerides
- MCPT, mouse mast cell protease
- MCT, medium chain triglycerides
- Murine Models of Food Allergy
- OIT, oral immunotherapy
- PBMC, peripheral blood mononuclear cell
- Spontaneous Sensitization
- TSLP, thymic stromal lymphopoietin
- Th, T helper
- Treg, regulatory T cell
- WASP, Wiskott–Aldrich syndrome protein
Collapse
|
20
|
Samadi N, Klems M, Untersmayr E. The role of gastrointestinal permeability in food allergy. Ann Allergy Asthma Immunol 2018; 121:168-173. [PMID: 29803708 DOI: 10.1016/j.anai.2018.05.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/04/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Nazanin Samadi
- Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Martina Klems
- Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Eva Untersmayr
- Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
21
|
Diesner SC, Bergmayr C, Wang XY, Heiden D, Exenberger S, Roth-Walter F, Starkl P, Ret D, Pali-Schöll I, Gabor F, Untersmayr E. Characterization of Vibrio cholerae neuraminidase as an immunomodulator for novel formulation of oral allergy immunotherapy. Clin Immunol 2018; 192:30-39. [PMID: 29608970 DOI: 10.1016/j.clim.2018.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 09/05/2017] [Accepted: 03/28/2018] [Indexed: 01/22/2023]
Abstract
To improve current mucosal allergen immunotherapy Vibrio cholerae neuraminidase (NA) was evaluated as a novel epithelial targeting molecule for functionalization of allergen-loaded, poly(D,L-lactide-co-glycolide) (PLGA) microparticles (MPs) and compared to the previously described epithelial targeting lectins wheat germ agglutinin (WGA) and Aleuria aurantia lectin (AAL). All targeters revealed binding to Caco-2 cells, but only NA had high binding specificity to α-L fucose and monosialoganglioside-1. An increased transepithelial uptake was found for NA-MPs in a M-cell co-culture model. NA and NA-MPs induced high levels of IFN-γ and IL10 in naive mouse splenocytes and CCL20 expression in Caco-2. Repeated oral gavage of NA-MPs resulted in a modulated, allergen-specific immune response. In conclusion, NA has enhanced M-cell specificity compared to the other targeters. NA functionalized MPs induce a Th1 and T-regulatory driven immune response and avoid allergy effector cell activation. Therefore, it is a promising novel, orally applied formula for allergy therapy.
Collapse
Affiliation(s)
- Susanne C Diesner
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Cornelia Bergmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Xue-Yan Wang
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Denise Heiden
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sarah Exenberger
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franziska Roth-Walter
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Philipp Starkl
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Davide Ret
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
| | - Isabella Pali-Schöll
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franz Gabor
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
22
|
A Kudoa septempunctata antigen induces production of IgE in BALB/c mice. Parasitol Res 2018; 117:303-306. [DOI: 10.1007/s00436-017-5665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/24/2017] [Indexed: 10/18/2022]
|
23
|
DALLAS DAVIDC, SANCTUARY MEGANR, QU YUNYAO, KHAJAVI SHABNAMHAGHIGHAT, VAN ZANDT ALEXANDRIAE, DYANDRA MELISSA, FRESE STEVENA, BARILE DANIELA, GERMAN JBRUCE. Personalizing protein nourishment. Crit Rev Food Sci Nutr 2017; 57:3313-3331. [PMID: 26713355 PMCID: PMC4927412 DOI: 10.1080/10408398.2015.1117412] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Proteins are not equally digestible-their proteolytic susceptibility varies by their source and processing method. Incomplete digestion increases colonic microbial protein fermentation (putrefaction), which produces toxic metabolites that can induce inflammation in vitro and have been associated with inflammation in vivo. Individual humans differ in protein digestive capacity based on phenotypes, particularly disease states. To avoid putrefaction-induced intestinal inflammation, protein sources, and processing methods must be tailored to the consumer's digestive capacity. This review explores how food processing techniques alter protein digestibility and examines how physiological conditions alter digestive capacity. Possible solutions to improving digestive function or matching low digestive capacity with more digestible protein sources are explored. Beyond the ileal digestibility measurements of protein digestibility, less invasive, quicker and cheaper techniques for monitoring the extent of protein digestion and fermentation are needed to personalize protein nourishment. Biomarkers of protein digestive capacity and efficiency can be identified with the toolsets of peptidomics, metabolomics, microbial sequencing and multiplexed protein analysis of fecal and urine samples. By monitoring individual protein digestive function, the protein component of diets can be tailored via protein source and processing selection to match individual needs to minimize colonic putrefaction and, thus, optimize gut health.
Collapse
Affiliation(s)
- DAVID C. DALLAS
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - MEGAN R. SANCTUARY
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - YUNYAO QU
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - SHABNAM HAGHIGHAT KHAJAVI
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Food Science and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - ALEXANDRIA E. VAN ZANDT
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - MELISSA DYANDRA
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - STEVEN A. FRESE
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - DANIELA BARILE
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - J. BRUCE GERMAN
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
- Foods for Health Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| |
Collapse
|
24
|
Kiewiet MBG, van Esch BCAM, Garssen J, Faas MM, de Vos P. Partially hydrolyzed whey proteins prevent clinical symptoms in a cow's milk allergy mouse model and enhance regulatory T and B cell frequencies. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201700340] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 01/06/2023]
Affiliation(s)
- Mensiena B. Gea Kiewiet
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen; University Medical Center Groningen; RB Groningen The Netherlands
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science; Utrecht University; Utrecht The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science; Utrecht University; Utrecht The Netherlands
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen; University Medical Center Groningen; RB Groningen The Netherlands
- Department of Obstetrics and Gynecology, University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen; University Medical Center Groningen; RB Groningen The Netherlands
| |
Collapse
|
25
|
Aitoro R, Paparo L, Amoroso A, Di Costanzo M, Cosenza L, Granata V, Di Scala C, Nocerino R, Trinchese G, Montella M, Ercolini D, Berni Canani R. Gut Microbiota as a Target for Preventive and Therapeutic Intervention against Food Allergy. Nutrients 2017; 9:nu9070672. [PMID: 28657607 PMCID: PMC5537787 DOI: 10.3390/nu9070672] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/15/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota plays a pivotal role in immune system development and function. Modification in the gut microbiota composition (dysbiosis) early in life is a critical factor affecting the development of food allergy. Many environmental factors including caesarean delivery, lack of breast milk, drugs, antiseptic agents, and a low-fiber/high-fat diet can induce gut microbiota dysbiosis, and have been associated with the occurrence of food allergy. New technologies and experimental tools have provided information regarding the importance of select bacteria on immune tolerance mechanisms. Short-chain fatty acids are crucial metabolic products of gut microbiota responsible for many protective effects against food allergy. These compounds are involved in epigenetic regulation of the immune system. These evidences provide a foundation for developing innovative strategies to prevent and treat food allergy. Here, we present an overview on the potential role of gut microbiota as the target of intervention against food allergy.
Collapse
Affiliation(s)
- Rosita Aitoro
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Lorella Paparo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Antonio Amoroso
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Margherita Di Costanzo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Linda Cosenza
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Viviana Granata
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Carmen Di Scala
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Rita Nocerino
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Giovanna Trinchese
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Mariangela Montella
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Danilo Ercolini
- Department of Agricultural Sciences, Division of Microbiology, University of Naples "Federico II", 80055 Portici, Italy.
- Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Roberto Berni Canani
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
- Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy.
- European Laboratory for the Investigation of Food Induced Diseases, University of Naples "Federico II", 80131 Naples, Italy.
- CEINGE Advanced Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
26
|
Kumar Gupta R, Kumar S, Gupta K, Sharma A, Roy R, Kumar Verma A, Chaudhari BP, Das M, Ahmad Ansari I, Dwivedi PD. Cutaneous exposure to clinically-relevant pigeon pea (Cajanus cajan) proteins promote TH2-dependent sensitization and IgE-mediated anaphylaxis in Balb/c mice. J Immunotoxicol 2016; 13:827-841. [DOI: 10.1080/1547691x.2016.1205159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Rinkesh Kumar Gupta
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
- Department of Biosciences, Integral University, Lucknow, India
| | - Sandeep Kumar
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, USA
| | - Kriti Gupta
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Akanksha Sharma
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Ruchi Roy
- Pharmacology and Chemical Biology Department, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alok Kumar Verma
- Section of Department of Medicine, Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University, New Orleans, LA, USA
| | - Bhushan P. Chaudhari
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| | - Mukul Das
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | | | - Premendra D. Dwivedi
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| |
Collapse
|
27
|
Diesner SC, Bergmayr C, Pfitzner B, Assmann V, Krishnamurthy D, Starkl P, Endesfelder D, Rothballer M, Welzl G, Rattei T, Eiwegger T, Szépfalusi Z, Fehrenbach H, Jensen-Jarolim E, Hartmann A, Pali-Schöll I, Untersmayr E. A distinct microbiota composition is associated with protection from food allergy in an oral mouse immunization model. Clin Immunol 2016; 173:10-18. [PMID: 27789346 PMCID: PMC5464391 DOI: 10.1016/j.clim.2016.10.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 01/08/2023]
Abstract
In our mouse model, gastric acid-suppression is associated with antigen-specific IgE and anaphylaxis development. We repeatedly observed non-responder animals protected from food allergy. Here, we aimed to analyse reasons for this protection. Ten out of 64 mice, subjected to oral ovalbumin (OVA) immunizations under gastric acid-suppression, were non-responders without OVA-specific IgE or IgG1 elevation, indicating protection from allergy. In these non-responders, allergen challenges confirmed reduced antigen uptake and lack of anaphylactic symptoms, while in allergic mice high levels of mouse mast-cell protease-1 and a body temperature reduction, indicative for anaphylaxis, were determined. Upon OVA stimulation, significantly lower IL-4, IL-5, IL-10 and IL-13 levels were detected in non-responders, while IL-22 was significantly higher. Comparison of fecal microbiota revealed differences of bacterial communities on single bacterial Operational-Taxonomic-Unit level between the groups, indicating protection from food allergy being associated with a distinct microbiota composition in a non-responding phenotype in this mouse model.
Collapse
Affiliation(s)
- Susanne C. Diesner
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Cornelia Bergmayr
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Barbara Pfitzner
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Department of Environmental Sciences, Research Unit Microbe-Plant Interactions, Research Group Molecular Microbial Ecology, Neuherberg, Germany
| | - Vera Assmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Durga Krishnamurthy
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Philipp Starkl
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - David Endesfelder
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Scientific Computing Research Unit, Neuherberg, Germany
| | - Michael Rothballer
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Department of Environmental Sciences, Research Unit Microbe-Plant Interactions, Research Group Molecular Microbial Ecology, Neuherberg, Germany
| | - Gerhard Welzl
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Department of Environmental Sciences, Research Unit Environmental Genomics, Neuherberg, Germany
| | - Thomas Rattei
- University of Vienna, Division of Computational Systems Biology, Vienna, Austria
| | - Thomas Eiwegger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Zsolt Szépfalusi
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Heinz Fehrenbach
- Division of Experimental Pneumology, Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Erika Jensen-Jarolim
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Comparative Medicine, Messerli Research Institute of the Veterinary University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Anton Hartmann
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Department of Environmental Sciences, Research Unit Microbe-Plant Interactions, Research Group Molecular Microbial Ecology, Neuherberg, Germany
| | - Isabella Pali-Schöll
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Comparative Medicine, Messerli Research Institute of the Veterinary University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Eva Untersmayr
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Versluis A, van Os-Medendorp H, Kruizinga AG, Blom WM, Houben GF, Knulst AC. Cofactors in allergic reactions to food: physical exercise and alcohol are the most important. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:392-400. [PMID: 27980774 PMCID: PMC5134724 DOI: 10.1002/iid3.120] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 12/03/2022]
Abstract
Introduction Involvement of cofactors, like physical exercise, alcohol consumption and use of several types of medication, are associated with more severe food allergic symptoms. However, there is limited evidence on how often cofactors play a role in food allergic reactions. The study aimed to get more insight into the frequency of exposure to cofactors and how often cofactors are associated with more severe symptoms in food allergic patients. Methods A questionnaire was completed by patients visiting the Allergology outpatient clinic. Patients with food allergy were included. Outcome measures were the frequency of medication use of medication groups that might act as cofactor and the frequency that physical exercise, alcohol consumption and use of analgesics are associated with more severe food allergic symptoms. Results Four hundred ninety‐six patients were included in the study. The frequency with which patients used one or more types of medication that might act as cofactors was 7.7%: antacids/acid neutralizing medication (5%), NSAIDs (2%), beta blockers (0.6%), angiotensin‐converting enzyme inhibitors (0.6%), and angiotensin receptor blockers (0.2%). Of all patients, 13% reported more severe symptoms to food after involvement of one or more of the cofactors: physical exercise (10%), alcohol consumption (5%), and use of analgesics (0.6%). Sixty‐five percent did not know if these cofactors caused more severe symptoms; 22% reported that these cofactors had no effect. Conclusions Only a small percentage of patients (7.7%) used medication that might aggravate food allergic reactions. Physical exercise and alcohol consumption were the most frequently reported cofactors, but occurring still in only 10% or less.
Collapse
|
29
|
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
|
30
|
Verma AK, Sharma A, Kumar S, Gupta RK, Kumar D, Gupta K, Giridhar B, Das M, Dwivedi PD. Purification, characterization and allergenicity assessment of 26 kDa protein, a major allergen from Cicer arietinum. Mol Immunol 2016; 74:113-24. [DOI: 10.1016/j.molimm.2016.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 04/21/2016] [Accepted: 05/04/2016] [Indexed: 12/24/2022]
|
31
|
Molina-Infante J, Prados-Manzano R, Gonzalez-Cordero PL. The role of proton pump inhibitor therapy in the management of eosinophilic esophagitis. Expert Rev Clin Immunol 2016; 12:945-52. [PMID: 27097787 DOI: 10.1080/1744666x.2016.1178574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Eosinophilic esophagitis (EoE) is a chronic esophageal disease characterized by a Th2 inflammatory response triggered by food/environmental allergens. Solid data confirm that up to half of patients with suspected EoE achieve complete remission on proton pump inhibitors (PPI) therapy. This disease phenotype is currently labelled as PPI-responsive esophageal eosinophilia (PPI-REE). Albeit initially believed to represent gastro-esophageal reflux disease (GERD), evolving evidence has underscored that PPI-REE and EoE show a significant overlap regarding clinic, endoscopic, histologic, Th2 immune-mediated inflammation and gene expression features. Moreover, PPI therapy can effectively reverse Th2 inflammation and the EoE transcriptome expression in PPI-REE patients. Therefore, EoE and PPI-REE likely represent a common allergic disorder, where PPI therapy should be considered a short- and long-term therapeutic asset, along with diet and topical steroids.
Collapse
Affiliation(s)
- Javier Molina-Infante
- a Department of Gastroenterology , Hospital San Pedro de Alcantara , Caceres , Spain
| | - Raul Prados-Manzano
- a Department of Gastroenterology , Hospital San Pedro de Alcantara , Caceres , Spain
| | | |
Collapse
|
32
|
Intestinal barrier dysfunction: implications for chronic inflammatory conditions of the bowel. Nutr Res Rev 2016; 29:40-59. [DOI: 10.1017/s0954422416000019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AbstractThe intestinal epithelium of adult humans acts as a differentially permeable barrier that separates the potentially harmful contents of the lumen from the underlying tissues. Any dysfunction of this boundary layer that disturbs the homeostatic equilibrium between the internal and external environments may initiate and sustain a biochemical cascade that results in inflammation of the intestine. Key to such dysfunction are genetic, microbial and other environmental factors that, singularly or in combination, result in chronic inflammation that is symptomatic of inflammatory bowel disease (IBD). The aim of the present review is to assess the scientific evidence to support the hypothesis that defective transepithelial transport mechanisms and the heightened absorption of intact antigenic proinflammatory oligopeptides are important contributing factors in the pathogenesis of IBD.
Collapse
|
33
|
Manzano-Szalai K, Pali-Schöll I, Krishnamurthy D, Stremnitzer C, Flaschberger I, Jensen-Jarolim E. Anaphylaxis Imaging: Non-Invasive Measurement of Surface Body Temperature and Physical Activity in Small Animals. PLoS One 2016; 11:e0150819. [PMID: 26963393 PMCID: PMC4786094 DOI: 10.1371/journal.pone.0150819] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/19/2016] [Indexed: 11/18/2022] Open
Abstract
In highly sensitized patients, the encounter with a specific allergen from food, insect stings or medications may rapidly induce systemic anaphylaxis with potentially lethal symptoms. Countless animal models of anaphylaxis, most often in BALB/c mice, were established to understand the pathophysiology and to prove the safety of different treatments. The most common symptoms during anaphylactic shock are drop of body temperature and reduced physical activity. To refine, improve and objectify the currently applied manual monitoring methods, we developed an imaging method for the automated, non-invasive measurement of the whole-body surface temperature and, at the same time, of the horizontal and vertical movement activity of small animals. We tested the anaphylaxis imaging in three in vivo allergy mouse models for i) milk allergy, ii) peanut allergy and iii) egg allergy. These proof-of-principle experiments suggest that the imaging technology represents a reliable non-invasive method for the objective monitoring of small animals during anaphylaxis over time. We propose that the method will be useful for monitoring diseases associated with both, changes in body temperature and in physical behaviour.
Collapse
Affiliation(s)
- Krisztina Manzano-Szalai
- Comparative Medicine, Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- Comparative Medicine, Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Durga Krishnamurthy
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Caroline Stremnitzer
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Erika Jensen-Jarolim
- Comparative Medicine, Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
34
|
Pablos-Tanarro A, López-Expósito I, Lozano-Ojalvo D, López-Fandiño R, Molina E. Antibody Production, Anaphylactic Signs, and T-Cell Responses Induced by Oral Sensitization With Ovalbumin in BALB/c and C3H/HeOuJ Mice. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2016; 8:239-45. [PMID: 26922934 PMCID: PMC4773212 DOI: 10.4168/aair.2016.8.3.239] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/30/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022]
Abstract
Purpose Two mouse strains, BALB/c and C3H/HeOuJ, broadly used in the field of food allergy, were compared for the evaluation of the allergenic potential of ovalbumin (OVA). Methods Sensitization was made by administering 2 different OVA doses (1 and 5 mg), with cholera toxin as Th2-polarizing adjuvant. Antibody levels, severity of anaphylaxis, and Th1 and Th2 responses induced by the allergen were assessed. In addition, because the mice selected had functional toll-like receptor 4, the influence of contamination with lipopolysaccharide (LPS) on the immunostimulating capacity of OVA on spleen cells was also evaluated. Results Both strains exhibited similar susceptibility to OVA sensitization. The 2 protein doses generated similar OVA-specific IgE and IgG1 levels in both strains, whereas C3H/HeOuJ mice produced significantly more IgG2a. Oral challenge provoked more severe manifestations in C3H/HeOuJ mice as indicated by the drop in body temperature and the severity of the anaphylactic scores. Stimulation of splenocytes with OVA led to significantly higher levels of Th2 and Th1 cytokines in BALB/c, and these were less affected by protein contamination with LPS. Conclusions The antibody and cytokine levels induced by OVA in BALB/c mice and the observation that BALB/c spleen cell cultures were more resistant than those of C3H/HeOuJ mice to the stimulus of LPS make this strain prone to exhibit Th2-mediated food allergic reactions and very adequate for the study of the features of OVA that make it allergenic.
Collapse
Affiliation(s)
- Alba Pablos-Tanarro
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera, Madrid, Spain
| | - Ivan López-Expósito
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera, Madrid, Spain
| | - Daniel Lozano-Ojalvo
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera, Madrid, Spain
| | - Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera, Madrid, Spain
| | - Elena Molina
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera, Madrid, Spain.
| |
Collapse
|
35
|
Untersmayr E. The influence of gastric digestion on the development of food allergy. REVUE FRANCAISE D ALLERGOLOGIE 2015; 55:444-447. [PMID: 28616101 DOI: 10.1016/j.reval.2015.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Food allergy represents an increasing health concern worldwide. To identify mechanisms and risk factors associated with food allergy development major research efforts are ongoing. For a long time only food allergens that are resistant to gastric enzymes were accepted to harbor sensitizing capacity via the oral route. However, over the past years several studies reported that even important food allergens can be readily degraded by digestive enzymes. Interestingly, a number of in vitro experiments confirmed that impairment of physiological gastric digestion by elevating gastric pH levels was associated with protein resistance. Additionally, pharmacological gastric acid suppression was found to be a risk factor for food allergy induction. In contrast, protein modifications resulting in increased susceptibility to digestive enzymes were reported to decrease the sensitization capacity via the oral route. The here reviewed data highlight the important gate keeping function of physiological gastric digestion in food allergy.
Collapse
Affiliation(s)
- Eva Untersmayr
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Abstract
For over 100 years it was believed that dietary protein must be completely hydrolysed before its constituent amino acids could be absorbed via specific amino acid transport systems. It is now known that the uptake of di- and tripeptides into the enterocyte is considerable, being transported across the intestinal endothelium by the PepT1 H+/peptide co-transporter. There is also evidence that some di- and tripeptides may survive cytosolic hydrolysis and be transported intact across the basolateral membrane. However, other than antigen sampling, the transport of larger intact macromolecules across the intestinal endothelium of the healthy adult human remains a controversial issue as there is little unequivocal in vivo evidence to support this postulation. The aim of the present review was to critically evaluate the scientific evidence that peptides/proteins are absorbed by healthy intestinal epithelia and pass intact into the hepatic portal system. The question of the absorption of oliogopeptides is paramount to the emerging science of food-derived bioactive peptides, their mode of action and physiological effects. Overall, we conclude that there is little unequivocal evidence that dietary bioactive peptides, other than di- and tripeptides, can cross the gut wall intact and enter the hepatic portal system in physiologically relevant concentrations.
Collapse
|
37
|
Diesner SC, Schultz C, Ackaert C, Oostingh GJ, Ondracek A, Stremnitzer C, Singer J, Heiden D, Roth-Walter F, Fazekas J, Assmann VE, Jensen-Jarolim E, Stutz H, Duschl A, Untersmayr E. Nitration of β-Lactoglobulin but Not of Ovomucoid Enhances Anaphylactic Responses in Food Allergic Mice. PLoS One 2015; 10:e0126279. [PMID: 25955653 PMCID: PMC4425501 DOI: 10.1371/journal.pone.0126279] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/31/2015] [Indexed: 12/31/2022] Open
Abstract
Background We revealed in previous studies that nitration of food proteins reduces the risk of de novo sensitization in a murine food allergy model. In contrast, in situations with preformed specific IgE antibodies, in vitro experiments suggested an increased capacity of effector cell activation by nitrated food proteins. Objective The aim of this study was to investigate the influence of protein nitration on the effector phase of food allergy. Design BALB/c mice were immunized intraperitoneally (i.p.) with the milk allergen β-lactoglobulin (BLG) or the egg allergen ovomucoid (OVM), followed by intragastric (i.g.) gavages to induce a strong local inflammatory response and allergen-specific antibodies. Subsequently, naïve and allergic mice were intravenously (i.v.) challenged with untreated, sham-nitrated or nitrated BLG or OVM. Anaphylaxis was monitored by measuring core body temperature and determination of mouse mast cell protease-1 (mMCP-1) levels in blood. Results A significant drop of body temperature accompanied with significantly elevated concentrations of the anaphylaxis marker mMCP-1 were only observed in BLG allergic animals challenged with nitrated BLG and not in OVM allergic mice challenged with nitrated OVM. SDS-PAGE and circular dichroism analysis of the differentially modified allergens revealed an effect of nitration on the secondary protein structure exclusively for BLG together with enhanced protein aggregation. Conclusion Our data suggest that nitration affects differently the food allergens BLG and OVM. In the case of BLG, structural changes favored dimerization possibly explaining the increased anaphylactic reactivity in BLG allergic animals.
Collapse
Affiliation(s)
- Susanne C. Diesner
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Cornelia Schultz
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Chloé Ackaert
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Gertie J. Oostingh
- Biomedical Sciences, Salzburg University of Applied Sciences, Puch/Salzburg, Austria
| | - Anna Ondracek
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Caroline Stremnitzer
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Josef Singer
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Denise Heiden
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Comparative Medicine, Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Judit Fazekas
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Vera E. Assmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Comparative Medicine, Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Hanno Stutz
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Albert Duschl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Eva Untersmayr
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
38
|
Lombardo C, Bonadonna P. Hypersensitivity Reactions to Proton Pump Inhibitors. CURRENT TREATMENT OPTIONS IN ALLERGY 2015. [DOI: 10.1007/s40521-015-0046-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
39
|
Lee MF, Yang KJ, Wang NM, Chiu YT, Chen PC, Chen YH. The development of a murine model for Forcipomyia taiwana (biting midge) allergy. PLoS One 2014; 9:e91871. [PMID: 24651257 PMCID: PMC3961268 DOI: 10.1371/journal.pone.0091871] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/15/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Forcipomyia taiwana (biting midge) allergy is the most prevalent biting insect allergy in Taiwan. An animal model corresponding to the human immuno-pathologic features of midge allergy is needed for investigating the mechanisms and therapies. This study successfully developed a murine model of Forcipomyia taiwana allergy. METHODS BALB/c mice were sensitized intra-peritoneally with midge extract on days 0, 7, 14, 21 then intra-dermally on days 28, 31 and 35. Serum midge-specific IgE, IgG1, and IgG2a were measured every 14 days by indirect ELISA. The mice were challenged intradermally with midge extract at day 40 and then sacrificed. Proliferation and cytokine production of splenocytes after stimulation with midge extract were determined by MTT assay and ELISA, respectively. The cytokine mRNA expression in response to midge stimulation was analyzed by RT-PCR. RESULTS Serum IgE, total IgG, and IgG1 antibody levels against midge extract were significantly higher in the midge-sensitized mice than in the control mice. After the two-step sensitization, all mice in the midge-sensitized group displayed immediate itch and plasma extravasation reactions in response to challenge with midge extract. Skin histology from midge-sensitized mice showed marked eosinophil and lymphocyte infiltrations similar to that observed in humans. Stimulation of murine splenocytes with midge extract elicited significant proliferation, IL-4, IL-10, IL-13 and IFN-γ protein production, and up-regulation of mRNA in a dose-dependent manner in the midge-sensitized group, but not in the control group. CONCLUSIONS A murine model of midge bite allergy has been successfully developed using a two-step sensitization protocol. The sensitized mice have very similar clinical and immunologic reactions to challenge with midge proteins as the reactions of human to midge bites. This murine model may be a useful platform for future research and the development of treatment strategies for insect bite allergy.
Collapse
Affiliation(s)
- Mey-Fann Lee
- Department of Medical Research Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Kai-Jei Yang
- Institute of Biotechnology, National Changhua University of Education, Changhua, Taiwan
| | - Nancy M. Wang
- Institute of Biotechnology, National Changhua University of Education, Changhua, Taiwan
| | - Yung-Tsung Chiu
- Department of Medical Research Taichung Veterans General Hospital, Taichung, Taiwan
| | - Pei-Chih Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
| | - Yi-Hsing Chen
- Department of Life Science, Tunghai University, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
40
|
Kumar S, Sharma A, Neelabh, Singh G, Verma AK, Roy R, Gupta R, Misra A, Tripathi A, Ansari KM, Das M, Shanker R, Dwivedi PD. Allergenic responses of green gram (Vigna radiata L. Millsp) proteins can be vitiated by induction of oral tolerance due to single acute dose in BALB/c mice. Food Res Int 2014. [DOI: 10.1016/j.foodres.2014.01.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
41
|
Phaseolin: A 47.5kDa protein of red kidney bean (Phaseolus vulgaris L.) plays a pivotal role in hypersensitivity induction. Int Immunopharmacol 2014; 19:178-90. [DOI: 10.1016/j.intimp.2014.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/11/2014] [Accepted: 01/13/2014] [Indexed: 12/30/2022]
|
42
|
Kumar D, Kumar S, Verma AK, Sharma A, Tripathi A, Chaudhari BP, Kant S, Das M, Jain SK, Dwivedi PD. Hypersensitivity linked to exposure of broad bean protein(s) in allergic patients and BALB/c mice. Nutrition 2013; 30:903-14. [PMID: 24985010 DOI: 10.1016/j.nut.2013.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/13/2013] [Accepted: 11/25/2013] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Broad bean (Vicia faba L.), a common vegetable, belongs to the family Fabaceae and is consumed worldwide. Limited studies have been done on allergenicity of broad beans. The aim of this study was to determine if broad bean proteins have the ability to elicit allergic responses due to the presence of clinically relevant allergenic proteins. METHODS Simulated gastric fluid (SGF) assay and immunoglobulin E (IgE) immunoblotting were carried out to identify pepsin-resistant and IgE-binding proteins. The allergenicity of broad beans was assessed in allergic patients, BALB/c mice, splenocytes, and RBL-2H3 cells. RESULTS Eight broad bean proteins of approximate molecular weight 70, 60, 48, 32, 23, 19, 15, and 10 kDa that remained undigested in SGF, showed IgE-binding capacity as well. Of 127 allergic patients studied, broad bean allergy was evident in 16 (12%). Mice sensitized with broad bean showed increased levels of histamine, total and specific IgE, and severe signs of systemic anaphylaxis compared with controls. Enhanced levels of histamine, prostaglandin D2, cysteinyl leukotriene, and β-hexosaminidase release were observed in the primed RBL-2H3 cells following broad bean exposure. The levels of interleukin IL-4, IL-5, IL-13 and regulated on activation, normal T-cell expressed and secreted were found enhanced in broad bean-treated splenocytes culture supernatant compared with controls. CONCLUSION This study inferred that broad bean proteins have the ability to elicit allergic responses due to the presence of clinically relevant allergenic proteins.
Collapse
Affiliation(s)
- Dinesh Kumar
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India; Department of Biotechnology, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Sandeep Kumar
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Alok K Verma
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Akanksha Sharma
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Anurag Tripathi
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Bhushan P Chaudhari
- Central Pathology Lab, Gehru Campus, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Surya Kant
- Department of Pulmonary Medicine, King George Medical University, Lucknow, India
| | - Mukul Das
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Swatantra K Jain
- Department of Biotechnology, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Premendra D Dwivedi
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.
| |
Collapse
|
43
|
Van Bilsen JH, Knippels LM, Penninks AH, Nieuwenhuizen WF, De Jongh HH, Koppelman SJ. The protein structure determines the sensitizing capacity of Brazil nut 2S albumin (Ber e1) in a rat food allergy model. Clin Transl Allergy 2013; 3:36. [PMID: 24180644 PMCID: PMC3827886 DOI: 10.1186/2045-7022-3-36] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/18/2013] [Indexed: 12/28/2022] Open
Abstract
It is not exactly known why certain food proteins are more likely to sensitize. One of the characteristics of most food allergens is that they are stable to the acidic and proteolytic conditions in the digestive tract. This property is thought to be a risk factor in allergic sensitization. The purpose of the present study was to investigate the contribution of the protein structure of 2S albumin (Ber e1), a major allergen from Brazil nut, on the sensitizing capacity in vivo using an oral Brown Norway rat food allergy model. Disulphide bridges of 2S albumin were reduced and alkylated resulting in loss of protein structure and an increased pepsin digestibility in vitro. Both native 2S albumin and reduced/alkylated 2S albumin were administered by daily gavage dosing (0.1 and 1 mg) to Brown Norway rats for 42 days. Intraperitoneal administration was used as a positive control. Sera were analysed by ELISA and passive cutaneous anaphylaxis. Oral exposure to native or reduced/alkylated 2S albumin resulted in specific IgG1 and IgG2a responses whereas only native 2S albumin induced specific IgE in this model, which was confirmed by passive cutaneous anaphylaxis. This study has shown that the disruption of the protein structure of Brazil nut 2S albumin decreased the sensitizing potential in a Brown Norway rat food allergy model, whereas the immunogenicity of 2S albumin remained preserved. This observation may open possibilities for developing immunotherapy for Brazil nut allergy.
Collapse
Affiliation(s)
| | - Léon Mj Knippels
- TNO, Zeist, Netherlands.,Nutricia Research, Utrecht, Netherlands
| | | | | | | | - Stef J Koppelman
- TNO, Zeist, Netherlands.,Food Allergy Research and Resource Program, University of Nebraska, Lincoln, Nebraska, USA
| |
Collapse
|
44
|
Phytohemagglutinins augment red kidney bean (Phaseolus vulgaris L.) induced allergic manifestations. J Proteomics 2013; 93:50-64. [DOI: 10.1016/j.jprot.2013.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/31/2013] [Accepted: 02/02/2013] [Indexed: 11/19/2022]
|
45
|
Wölbing F, Fischer J, Köberle M, Kaesler S, Biedermann T. About the role and underlying mechanisms of cofactors in anaphylaxis. Allergy 2013; 68:1085-92. [PMID: 23909934 DOI: 10.1111/all.12193] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2013] [Indexed: 12/20/2022]
Abstract
Anaphylaxis is the systemic and most severe presentation of type I allergy. A number of conditions were identified that modulate the onset of anaphylaxis such as co- or augmentation factors, which significantly lower the allergen dose necessary for triggering anaphylaxis. Next to physical exercise or alcohol consumption, co-administration of nonsteroidal anti-inflammatory drugs (NSAID) or concomitant infectious diseases are well-documented cofactors of anaphylaxis. Registries for anaphylaxis document a role for cofactors in about 30% of anaphylactic reactions. Some disease entities such as 'wheat-dependent exercise-induced anaphylaxis' (WDEIA) are explicitly characterized by elicitation of anaphylaxis only in the presence of at least one such cofactor. Using WDEIA as a model disease, studies demonstrated that exercise increases skin prick test reactivity to and bioavailability of the allergen. Additional data indicate that alcohol consumption and NSAID administration display similar effects. Modulation of the cellular activation threshold is another mechanism underlying cofactor-induced anaphylaxis, most likely also functional when infectious diseases orchestrate elicitation of anaphylaxis. Cofactors are increasingly accepted to play a fundamental role in eliciting anaphylaxis. Consequently, to improve patient management modalities, a better understanding of the underlying mechanisms is warranted. This review aims to update clinicians and clinical scientists on recent developments.
Collapse
Affiliation(s)
- F. Wölbing
- Department of Dermatology; Eberhard-Karls-University of Tübingen; Tübingen; Germany
| | - J. Fischer
- Department of Dermatology; Eberhard-Karls-University of Tübingen; Tübingen; Germany
| | - M. Köberle
- Department of Dermatology; Eberhard-Karls-University of Tübingen; Tübingen; Germany
| | - S. Kaesler
- Department of Dermatology; Eberhard-Karls-University of Tübingen; Tübingen; Germany
| | - T. Biedermann
- Department of Dermatology; Eberhard-Karls-University of Tübingen; Tübingen; Germany
| |
Collapse
|
46
|
A BALB/c mouse model for assessing the potential allergenicity of proteins: comparison of allergen dose, sensitization frequency, timepoint and sex. Food Chem Toxicol 2013; 62:41-7. [PMID: 23969083 DOI: 10.1016/j.fct.2013.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/31/2013] [Accepted: 08/03/2013] [Indexed: 01/28/2023]
Abstract
The present study was to investigate the possibility of using the BALB/c mouse as an animal model for assessing the potential allergenicity of proteins. Specific IgE and IgG1 against ovalbumin were induced by dosing BALB/c mice via intraperitoneal injection (absence of adjuvant). The effects of various allergen doses (5 mg, 0.5 mg or 0.05 mg OVA), sensitization times (twice or five times), timepoints (day 14 or day 28) and sex (male or female) were studied. IL-4, IFN-γ, OVA-specific IgE and IgG1 were measured by enzyme-linked immunosorbent assay (ELISA). A general finding was that mice treated with 0.05 mg OVA had the highest OVA-specific IgE and IgG1, statistically significant higher specific IgE and IgG1 were observed in groups sensitized five times than twice, OVA-specific IgE and IgG1 on day 28 were statistically higher than day 14, and higher IL-4 was observed in OVA-allergic mice than control mice. These results demonstrate that the BALB/c mouse model treated with 0.05 mg OVA intraperitoneally on days 0, 3, 6, 9, 12 might be used for further experiments. OVA-specific IgE and IgG1 should be detected on day 28. Further studies including reproducibility and other conditions were required before using the BALB/c mouse model for assessing the potential allergenicity of proteins.
Collapse
|
47
|
Dhaliwal J, Tobias V, Sugo E, Varjavandi V, Lemberg D, Day A, Bohane T, Ledder O, Jiwane A, Adams S, Henry G, Dilley A, Shi E, Krishnan U. Eosinophilic esophagitis in children with esophageal atresia. Dis Esophagus 2013; 27:340-7. [PMID: 23947919 DOI: 10.1111/dote.12119] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eosinophilic esophagitis (EoE) has only rarely been reported in esophageal atresia (EA) patients. A retrospective case analysis of all EA patients born at our center between January 1999 and April 2012 was performed. A total of 113 of patients were identified; 10 patients were excluded as a result of inadequate data. Eighteen patients (17%) were diagnosed with EoE. The average number of eosinophilis was 30/high-power field (HPF) (19/HPF-80/HPF). The median age for diagnosis of EoE was 1 year and 6 months (8 months-8 years and 7 months). Children with EoE had a significantly greater incidence of reflux symptoms, dysphagia, tracheomalacia, and 'hypoxic spells' (P < 0.05). EoE patients also underwent significantly more surgery including fundoplication and aortopexy when compared with those without EoE (P < 0.0001). Although the incidence of gastrostomy was greater in the EoE group (33% vs. 13%), this was not statistically significant. Half of the EoE patients had a coexisting atopic condition at time of diagnosis. The commonest condition was asthma 7/18 (38%) followed by specific food allergy 6/18 (33%). EoE was treated in 11 patients with either swallowed fluticasone or budesonide slurry. All improved clinically. Histologically, five had complete resolution and six had partial improvement. Six children with EoE were treated with acid suppression alone. All improved clinically, and 5/6 had subsequent histological resolution. One child who received acid suppression and an exclusion diet also improved. Seven patients (38%) had an esophageal stricture at time of EoE diagnosis. Five were dilated at time of the initial endoscopy, prior to the diagnosis of EoE being available. Two patients had resolution of their strictures on medical treatment of their EoE alone and did not require further dilatation. EoE was seen in 17% of children with EA in this study. EoE should be considered in EA patients with persistent symptoms on standard reflux treatment, increasing dysphagia, and recurrent strictures.
Collapse
Affiliation(s)
- J Dhaliwal
- Department of Paediatric Gastroenterology, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kumar S, Sharma A, Verma AK, Chaudhari B, Das M, Jain S, Dwivedi PD. Allergenicity potential of red kidney bean (Phaseolus vulgaris L.) proteins in orally treated BALB/c mice and passively sensitized RBL-2H3 cells. Cell Immunol 2013; 284:37-44. [DOI: 10.1016/j.cellimm.2013.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 06/25/2013] [Accepted: 07/01/2013] [Indexed: 01/10/2023]
|
49
|
Epicutaneous sensitization results in IgE-dependent intestinal mast cell expansion and food-induced anaphylaxis. J Allergy Clin Immunol 2013; 131:451-60.e1-6. [PMID: 23374269 DOI: 10.1016/j.jaci.2012.11.032] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 11/02/2012] [Accepted: 11/08/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sensitization to food antigen can occur through cutaneous exposure. OBJECTIVE We sought to test the hypothesis that epicutaneous sensitization with food antigen predisposes to IgE-mediated anaphylaxis on oral allergen challenge. METHODS BALB/c mice were epicutaneously sensitized by repeated application of ovalbumin (OVA) to tape-stripped skin over 7 weeks or orally immunized with OVA and cholera toxin (CT) weekly for 8 weeks and then orally challenged with OVA. Body temperature was monitored, and serum mouse mast cell protease 1 levels were determined after challenge. Tissue mast cell (MC) counts were examined by using chloroacetate esterase staining. Levels of serum OVA-specific IgE and IgG(1) antibodies and cytokines in supernatants of OVA-stimulated splenocytes were measured by means of ELISA. Serum IL-4 levels were measured by using an in vivo cytokine capture assay. RESULTS Epicutaneously sensitized mice exhibited expansion of connective tissue MCs in the jejunum, increased serum IL-4 levels, and systemic anaphylaxis after oral challenge, as evidenced by decreased body temperature and increased serum mouse mast cell protease 1 levels. Intestinal MC expansion and anaphylaxis were IgE dependent because they did not occur in epicutaneously sensitized IgE(-/-) mice. Mice orally immunized with OVA plus CT did not have increased serum IL-4 levels, expanded intestinal MCs, or anaphylaxis after oral challenge, despite OVA-specific IgE levels and splenocyte cytokine production in response to OVA stimulation, which were comparable with those of epicutaneously sensitized mice. CONCLUSION Epicutaneously sensitized mice, but not mice orally immunized with antigen plus CT, have expansion of intestinal MCs and IgE-mediated anaphylaxis after single oral antigen challenge. IgE is necessary but not sufficient for food anaphylaxis, and MC expansion in the gut can play an important role in the development of anaphylaxis.
Collapse
|
50
|
Digestibility and IgE-binding of glycosylated codfish parvalbumin. BIOMED RESEARCH INTERNATIONAL 2013; 2013:756789. [PMID: 23878817 PMCID: PMC3708386 DOI: 10.1155/2013/756789] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 12/05/2022]
Abstract
Food-processing conditions may alter the allergenicity of food proteins by different means. In this study, the effect of the glycosylation as a result of thermal treatment on the digestibility and IgE-binding of codfish parvalbumin is investigated. Native and glycosylated parvalbumins were digested with pepsin at various conditions relevant for the gastrointestinal tract. Intact proteins and peptides were analysed for apparent molecular weight and IgE-binding. Glycosylation did not substantially affect the digestion. Although the peptides resulting from digestion were relatively large (3 and 4 kDa), the IgE-binding was strongly diminished. However, the glycosylated parvalbumin had a strong propensity to form dimers and tetramers, and these multimers bound IgE intensely, suggesting stronger IgE-binding than monomeric parvalbumin. We conclude that glycosylation of codfish parvalbumin does not affect the digestibility of parvalbumin and that the peptides resulting from this digestion show low IgE-binding, regardless of glycosylation. Glycosylation of parvalbumin leads to the formation of higher order structures that are more potent IgE binders than native, monomeric parvalbumin. Therefore, food-processing conditions applied to fish allergen can potentially lead to increased allergenicity, even while the protein's digestibility is not affected by such processing.
Collapse
|