1
|
Yang T, Li T, Xing Y, Cao M, Zhang M, Leng Q, Qiu J, Song X, Chen J, Hu G, Qian Y. Dietary nucleic acids promote oral tolerance through innate sensing pathways in mice. Nat Commun 2024; 15:9461. [PMID: 39487135 PMCID: PMC11530426 DOI: 10.1038/s41467-024-53814-0] [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: 02/26/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
Oral tolerance is essential for intestinal homeostasis and systemic immune function. However, our understanding of how oral tolerance is maintained is inadequate. Here we report that food-derived nucleic acids promote oral tolerance through innate sensing pathways. We find that dietary nucleic acids, but not microbiota, expand the natural intraepithelial lymphocyte (IEL) pool, specifically in the small intestine. TGF-β1, produced by natural IELs, then promotes activation of gut CD103+ dendritic cells to support the induction of antigen-specific Treg cells in a mouse model of OVA-induced oral tolerance. Mechanistically, MAVS and STING are redundantly required for sensing dietary RNAs and DNAs to activate downstream TBK1 signalling to induce IL-15 production, which results in the accumulation of natural IELs. Thus, our study demonstrates a key role of food-triggered innate sensing pathways in the maintenance of natural IELs and oral tolerance.
Collapse
Affiliation(s)
- Tao Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tian Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yingying Xing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengtao Cao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingxiang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Qibin Leng
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510180, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xinyang Song
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Youcun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China.
| |
Collapse
|
2
|
Sanidad KZ, Rager SL, Carrow HC, Ananthanarayanan A, Callaghan R, Hart LR, Li T, Ravisankar P, Brown JA, Amir M, Jin JC, Savage AR, Luo R, Rowdo FM, Martin ML, Silver RB, Guo CJ, Krumsiek J, Inohara N, Zeng MY. Gut bacteria-derived serotonin promotes immune tolerance in early life. Sci Immunol 2024; 9:eadj4775. [PMID: 38489352 PMCID: PMC11328322 DOI: 10.1126/sciimmunol.adj4775] [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: 06/30/2023] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
The gut microbiota promotes immune system development in early life, but the interactions between the gut metabolome and immune cells in the neonatal gut remain largely undefined. Here, we demonstrate that the neonatal gut is uniquely enriched with neurotransmitters, including serotonin, and that specific gut bacteria directly produce serotonin while down-regulating monoamine oxidase A to limit serotonin breakdown. We found that serotonin directly signals to T cells to increase intracellular indole-3-acetaldehdye and inhibit mTOR activation, thereby promoting the differentiation of regulatory T cells, both ex vivo and in vivo in the neonatal intestine. Oral gavage of serotonin into neonatal mice resulted in long-term T cell-mediated antigen-specific immune tolerance toward both dietary antigens and commensal bacteria. Together, our study has uncovered an important role for specific gut bacteria to increase serotonin availability in the neonatal gut and identified a function of gut serotonin in shaping T cell response to dietary antigens and commensal bacteria to promote immune tolerance in early life.
Collapse
Affiliation(s)
- Katherine Z Sanidad
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stephanie L Rager
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hannah C Carrow
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, USA
| | - Aparna Ananthanarayanan
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ryann Callaghan
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, USA
| | - Lucy R Hart
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tingting Li
- Jill Roberts Institute for Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10065, USA
| | - Purnima Ravisankar
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, USA
| | - Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mohammed Amir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jenny C Jin
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexandria Rose Savage
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ryan Luo
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - M Laura Martin
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Randi B Silver
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chun-Jun Guo
- Jill Roberts Institute for Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jan Krumsiek
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Naohiro Inohara
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, USA
| |
Collapse
|
3
|
Zhang J, Yang X, Chen G, Hu J, He Y, Ma J, Ma Z, Chen H, Huang Y, Wu Q, Liu Y, Yu L, Zhang H, Lai H, Zhang J, Zhai J, Huang M, Zou Z, Tao A. Efficacy and safety of intratonsillar immunotherapy for allergic rhinitis: A randomized, double-blind, placebo-controlled clinical trial. Ann Allergy Asthma Immunol 2024; 132:346-354.e1. [PMID: 37913839 DOI: 10.1016/j.anai.2023.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND A lower adherence rate existed in patients receiving allergen-specific immunotherapy due to its lengthy period and adverse effects even though it is the only curative treatment for IgE-mediated allergies. Therefore, exploring innovative allergen-specific immunotherapy routes is necessary. OBJECTIVE To explore the efficacy and safety of the intratonsillar injection of house dust mite (HDM) extract in patients with HDM-induced allergic rhinitis (AR). METHODS A randomized, double-blind, placebo-controlled clinical trial was conducted. A total of 80 patients with HDM-induced AR were randomized to receive 6 intratonsillar injections with HDM extract or placebo in 3 months. The total nasal symptom score (TNSS), visual analogue scale of nasal symptoms, combined symptom and medication score, mini rhinoconjunctivitis quality of life questionnaire, and serum allergen-specific IgG4 to Dermatophagoides pteronyssinus were all monitored at baseline and 3 months, 6 months, and 12 months after the treatment was finished. The intent-to-treat and per-protocol set (PPS) are both analyzed. RESULTS The primary end points TNSS and ΔTNSS were improved significantly at 3 months after the patients with AR finished a 3-month 6-injection intratonsillar immunotherapy compared with those in the placebo treatment in both intent-to-treat and PPS. Results of visual analogue scale, combined symptom and medication score, and mini rhinoconjunctivitis quality of life questionnaire were also improved significantly at 3 months after the treatment in PPS. However, the improvement effect of intratonsillar immunotherapy at 6 and 12 months was limited and uncertain based on the data. The increase of serum Der p IgG4 in the active group was significantly higher than that in the placebo group at 3, 6, and 12 months after the treatment was finished. Adverse events were monitored, and no systemic adverse reactions were observed. CONCLUSION The clinical trial revealed that intratonsillar injection with HDM extract was safe and effective in patients with AR. Optimizing the protocol and allergen formulations is expected to increase and maintain the efficacy of this novel approach. TRIAL REGISTRATION https://www.chictr.org.cn/index.html, identifier: ChiCTR-TRC-13003600.
Collapse
Affiliation(s)
- Junyan Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaobin Yang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guangui Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jintao Hu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ying He
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jinxiang Ma
- Department of Epidemiology and Health Statistics, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhaoen Ma
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Huifang Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuyi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qiurong Wu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yongping Liu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Lu Yu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Hong Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - He Lai
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianguo Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jinming Zhai
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Minqi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zehong Zou
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
4
|
White TLA, Jin Y, Roberts SDA, Gable MJ, Morel PA. Phosphorylation of hnRNP A1-Serine 199 Is Not Required for T Cell Differentiation and Function. Immunohorizons 2024; 8:136-146. [PMID: 38334757 PMCID: PMC10916359 DOI: 10.4049/immunohorizons.2300074] [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: 09/11/2023] [Accepted: 01/05/2024] [Indexed: 02/10/2024] Open
Abstract
hnRNP A1 is an important RNA-binding protein that influences many stages of RNA processing, including transcription, alternative splicing, mRNA nuclear export, and RNA stability. However, the role of hnRNP A1 in immune cells, specifically CD4+ T cells, remains unclear. We previously showed that Akt phosphorylation of hnRNP A1 was dependent on TCR signal strength and was associated with Treg differentiation. To explore the impact of hnRNP A1 phosphorylation by Akt on CD4+ T cell differentiation, our laboratory generated a mutant mouse model, hnRNP A1-S199A (A1-MUT) in which the major Akt phosphorylation site on hnRNP A1 was mutated to alanine using CRISPR Cas9 technology. Immune profiling of A1-MUT mice revealed changes in the numbers of Tregs in the mesenteric lymph node. We found no significant differences in naive CD4+ T cell differentiation into Th1, Th2, Th17, or T regulatory cells (Tregs) in vitro. In vivo, Treg differentiation assays using OTII-A1-Mut CD4+ T cells exposed to OVA food revealed migration and homing defects in the A1-MUT but no change in Treg induction. A1-MUT mice were immunized with NP- keyhole limpet hemocyanin, and normal germinal center development, normal numbers of NP-specific B cells, and no change in Tfh numbers were observed. In conclusion, Akt phosphorylation of hnRNP A1 S199 does not play a role in CD4+ T cell fate or function in the models tested. This hnRNP A1-S199A mouse model should be a valuable tool to study the role of Akt phosphorylation of hnRNP A1-S199 in different cell types or other mouse models of human disease.
Collapse
Affiliation(s)
- Tristan L. A. White
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sean D. A. Roberts
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Matthew J. Gable
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
5
|
Turkalj M, Miletić Gospić A, Višekruna Džidić I, Banić I. Food Allergen Immunotherapy in the Treatment of Patients with IgE-Mediated Food Allergy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:121. [PMID: 38256382 PMCID: PMC10820435 DOI: 10.3390/medicina60010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024]
Abstract
The prevalence of allergic diseases, including food allergy, is increasing, especially in developed countries. Implementation of an elimination diet is not a sufficient therapeutic strategy in patients with food allergy, whose quality of life is significantly impaired. In recent years, new effective therapeutic strategies have been developed, such as the application of oral, sublingual, and epicutaneous immunotherapy. Oral immunotherapy is the most often applied strategy because of its effectiveness and ease of application, with an acceptable safety profile. The effectiveness of oral immunotherapy in patients with egg, cow's milk, and peanut allergy has been proven both in terms of raising of the threshold and the development of tolerance, and in some patients, the development of sustainable unresponsiveness. Although oral immunotherapy is an effective treatment for food allergy, several limitations, including a long duration and a significant rate of reported adverse events, reduces its success. Therefore, new therapeutic options, such as treatment with biologicals, either as combinations with food allergen immunotherapy or as monotherapy with the aim of improving the efficacy and safety of treatment, are being investigated.
Collapse
Affiliation(s)
- Mirjana Turkalj
- Srebrnjak Children’s Hospital, HR-10000 Zagreb, Croatia; (M.T.); (I.V.D.); (I.B.)
- Faculty of Medicine, J.J. Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- School of Medicine, Catholic University of Croatia, HR-10000 Zagreb, Croatia
| | | | | | - Ivana Banić
- Srebrnjak Children’s Hospital, HR-10000 Zagreb, Croatia; (M.T.); (I.V.D.); (I.B.)
| |
Collapse
|
6
|
Crespo JF, Cabanillas B. Recent advances in cellular and molecular mechanisms of IgE-mediated food allergy. Food Chem 2023; 411:135500. [PMID: 36682170 DOI: 10.1016/j.foodchem.2023.135500] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Food allergy is a public health issue the prevalence of which is steadily increasing. New discoveries have contributed to the understanding of the molecular and cellular mechanisms that lead to IgE-mediated food allergy. Novel scientific findings have defined roles for specific cell types, such as T follicular helper cells, in induction of high-affinity IgE by B cells. Also, not only mast cells and basophils contribute to food anaphylaxis, but also other cell types, such as neutrophils and macrophages. Elucidation of mechanisms involved in sensitization to food allergens through organs including the skin is key to deepening our understanding of the "dual exposure" hypothesis, which suggests that allergic sensitization is mainly acquired through inflamed skin while the oral route induces tolerance. This review considers the latest scientific knowledge about the molecular and cellular mechanisms of IgE-mediated food allergy. It reveals crucial components involved in the sensitization and elicitation phases and emerging approaches in anaphylaxis pathophysiology.
Collapse
Affiliation(s)
- Jesus F Crespo
- Department of Allergy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| |
Collapse
|
7
|
Huang M, Shao H, Wang Z, Chen H, Li X. Specific and nonspecific nutritional interventions enhance the development of oral tolerance in food allergy. Crit Rev Food Sci Nutr 2023; 64:10303-10318. [PMID: 37313721 DOI: 10.1080/10408398.2023.2222803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The goal of food allergy (FA) prevention and treatment is to induce oral tolerance (OT). Appropriate nutritional interventions are essential to induce OT to food allergens. This review introduces the mechanism of OT and the importance of early nutritional interventions, and then firstly summarizes specific nutritional factors to induce the development of OT of FA, including proteins, vitamins, fatty acids, saccharides and probiotics. The regulatory mechanism mainly induces the development of tolerance by increasing local or systemic protective regulatory T cells (Tregs) to suppress FA, while the gut microbiota may also be changed to maintain intestinal homeostasis. For allergens-specific OT, the disruption to the structure of proteins and epitopes is critical for the induction of tolerance by hydrolyzed and heated proteins. Vitamins (vitamin A, D), fatty acids, saccharides and probiotics as allergens nonspecific OT also induce the development of OT through immunomodulatory effects. This review contributes to our understanding of OT in FA through nutritional interventions. Nutritional interventions play an important role in the induction of OT, and offer promising approaches to reduce allergy risk and alleviate FA. Moreover, due to the importance and diversity of nutrition, it must be the future trend of induction of OT in FA.
Collapse
Affiliation(s)
- Meijia Huang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Huming Shao
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Zhongliang Wang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| | - Xin Li
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| |
Collapse
|
8
|
Nguyen NH, Chak V, Keller K, Wu H, Balu-Iyer SV. Phosphatidylserine-mediated oral tolerance. Cell Immunol 2023; 384:104660. [PMID: 36586393 PMCID: PMC11034824 DOI: 10.1016/j.cellimm.2022.104660] [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: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 12/26/2022]
Abstract
Phosphatidylserine (PS) is an anionic phospholipid exposed on the surface of apoptotic cells. The exposure of PS typically recruits and signals phagocytes to engulf and silently clear these dying cells to maintain tolerance via immunological ignorance. However, recent and emerging evidence has demonstrated that PS converts an "immunogen" into a "tolerogen", and PS exposure on the surface of cells or vesicles actively promotes a tolerogenic environment. This tolerogenic property depends on the biophysical characteristics of PS-containing vesicles, including PS density on the particle surface to effectively engage tolerogenic receptors, such as TIM-4, which is exclusively expressed on the surface of antigen-presenting cells. We harnessed the cellular and molecular mechanistic insight of PS-mediated immune regulation to design an effective oral tolerance approach. This immunotherapy has been shown to prevent/reduce immune response against life-saving protein-based therapies, food allergens, autoantigens, and the antigenic viral capsid peptide commonly used in gene therapy, suggesting a broad spectrum of potential clinical applications. Given the good safety profile of PS together with the ease of administration, oral tolerance achieved with PS-based nanoparticles has a very promising therapeutic impact.
Collapse
Affiliation(s)
| | - Vincent Chak
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Katherine Keller
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Helen Wu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
9
|
Puricelli C, Boggio E, Gigliotti CL, Stoppa I, Sutti S, Rolla R, Dianzani U. Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review. Pharmaceutics 2022; 14:pharmaceutics14091782. [PMID: 36145531 PMCID: PMC9501480 DOI: 10.3390/pharmaceutics14091782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional therapies for immune-mediated diseases, including autoimmune disorders, transplant reactions, and allergies, have undergone a radical evolution in the last few decades; however, they are still not specific enough to avoid widespread immunosuppression. The idea that vaccine usage could be extended beyond its traditional immunogenic function by encompassing the ability of vaccines to induce antigen-specific tolerance may revolutionize preventive and therapeutic strategies in several clinical fields that deal with immune-mediated disorders. This approach has been supported by improved data relating to the several mechanisms involved in controlling unwanted immune responses and allowing peripheral tolerance. Given these premises, several approaches have been developed to induce peripheral tolerance against the antigens that are involved in the pathological immune response, including allergens, autoantigens, and alloantigens. Technological innovations, such as nucleic acid manipulation and the advent of micro- and nanoparticles, have further supported these novel preventive and therapeutic approaches. This review focuses on the main strategies used in the development of tolerogenic vaccines, including the technological issues used in their design and the role of “inverse adjuvants”. Even though most studies are still limited to the preclinical field, the enthusiasm generated by their results has prompted some initial clinical trials, and they show great promise for the future management of immune-mediated pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberta Rolla
- Correspondence: ; Tel.: +39-0321-3733583; Fax: +39-0321-3733987
| | | |
Collapse
|
10
|
Benne N, Ter Braake D, Stoppelenburg AJ, Broere F. Nanoparticles for Inducing Antigen-Specific T Cell Tolerance in Autoimmune Diseases. Front Immunol 2022; 13:864403. [PMID: 35392079 PMCID: PMC8981588 DOI: 10.3389/fimmu.2022.864403] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Autoimmune diseases affect many people worldwide. Current treatment modalities focus on the reduction of disease symptoms using anti-inflammatory drugs which can lead to side effects due to systemic immune suppression. Restoration of immune tolerance by down-regulating auto-reactive cells in an antigen-specific manner is currently the “holy grail” for the treatment of autoimmune diseases. A promising strategy is the use of nanoparticles that can deliver antigens to antigen-presenting cells which in turn can enhance antigen-specific regulatory T cells. In this review, we highlight some promising cell targets (e.g. liver sinusoidal endothelial cells and splenic marginal zone macrophages) for exploiting natural immune tolerance processes, and several strategies by which antigen-carrying nanoparticles can target these cells. We also discuss how nanoparticles carrying immunomodulators may be able to activate tolerance in other antigen-presenting cell types. Finally, we discuss some important aspects that must be taken into account when translating data from animal studies to patients.
Collapse
Affiliation(s)
- Naomi Benne
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Daniëlle Ter Braake
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Arie Jan Stoppelenburg
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
11
|
Mao R, Yang M, Yang R, Chen Y, Diao E, Zhang T, Li D, Chang X, Chi Z, Wang Y. Oral delivery of the intracellular domain of the insulinoma-associated protein 2 (IA-2ic) by bacterium-like particles (BLPs) prevents type 1 diabetes mellitus in NOD mice. Drug Deliv 2022; 29:925-936. [PMID: 35311607 PMCID: PMC8942491 DOI: 10.1080/10717544.2022.2053760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Antigen-specific immune tolerance, which possesses great potential in preventing or curing type 1 diabetes mellitus (T1DM), can be induced by oral vaccination with T1DM-related autoantigens. However, direct administration of autoantigens via oral route exhibits a low tolerance-inducing effect as a result of the digestion of protein antigens in the gastrointestinal tract (GIT) and therefore, a large dosage of autoantigens may be needed. In this study, bacterium-like particles (BLPs) made from food-grade lactic acid bacteria were used to deliver the intracellular domain of the insulinoma-associated protein 2 (IA-2ic). For this purpose, BLPs-IA-2ic vaccine in which IA-2ic bound to the surface of BLPs was constructed. BLPs enhanced the stability of the delivered IA-2ic based on the stability analysis in vitro. Oral administration of BLPs-IA-2ic significantly reduced T1DM incidence in NOD mice. The mice fed BLPs-IA-2ic exhibited a significant reduction in insulitis and preserved the ability to secrete insulin. Immunologic analysis showed that oral vaccination with BLPs-IA-2ic induced antigen-specific T cell tolerance. The results revealed that the successful induction of immune tolerance was dependent on the immune deviation (in favor of T helper 2 responses) and CD4+CD25+FoxP3+ regulatory T cells. Hence, oral vaccination with BLPs-IA-2ic shows potential for application in preventing T1DM.
Collapse
Affiliation(s)
- Ruifeng Mao
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Menglan Yang
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Rui Yang
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Yingying Chen
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Enjie Diao
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Tong Zhang
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Dengchao Li
- School of Life Sciences, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huai'an, China
| | - Xin Chang
- Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Zhenjing Chi
- Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Yefu Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
12
|
林 冬, 杨 利, 王 智. [Research Updates: The Role of Interaction between Oral Microbiota, Immune Cells, and Epithelial Barrier in Oral Mucosal Homeostasis and Pathogenesis]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:188-193. [PMID: 35332716 PMCID: PMC10409361 DOI: 10.12182/20220360501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Indexed: 06/14/2023]
Abstract
In a healthy state, the interaction between the oral microorganisms, mucosal immune cells and epithelial barrier can maintain the oral microecological stability. However, the oral microecology is disrupted under a diseased state and various pathogenic bacteria and their virulence factors and metabolites irritate the immune system, which causes direct or indirect damage to the epithelial barrier, promotes the pathogenesis and progression of oral mucosal diseases, and triggers immune inflammatory response or the irreversible transformation from inflammation into cancer. We herein reviewed the interaction between oral microorganisms, immune cells and epithelial barrier from two perspectives, the maintenance of the oral homeostasis and the pathogenesis of oral mucosal diseases. We intended to gain further understanding of the oral mucosal homeostasis and the mechanism of action of the pathogenesis and progression of oral mucosal diseases, and to provide thereby ideas and scientific and theoretical basis for developing new strategies for the diagnosis and treatment of oral mucosal diseases through re-establishing mucosal homeostasis.
Collapse
Affiliation(s)
- 冬佳 林
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - 利洒 杨
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - 智 王
- 中山大学光华口腔医学院·附属口腔医院 (广州 510055)Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- 广东省口腔医学重点实验室 (广州 510055)Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
13
|
Mechler-Dreibi ML, Almeida HMS, Sonalio K, Martines MAC, Petri FAM, Zambotti BB, Ferreira MM, Storino GY, Martins TS, Montassier HJ, Sant'Anna OA, Fantini MCA, de Oliveira LG. Oral vaccination of piglets against Mycoplasma hyopneumoniae using silica SBA-15 as an adjuvant effectively reduced consolidation lung lesions at slaughter. Sci Rep 2021; 11:22377. [PMID: 34789792 PMCID: PMC8599662 DOI: 10.1038/s41598-021-01883-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Mycoplasma (M.) hyopneumoniae is the main pathogen of porcine enzootic pneumonia (PEP). Its controlling is challenging, and requires alternative strategies. This study aimed to develop an oral vaccine against M. hyopneumoniae using a nanostructured mesoporous silica (SBA-15) as an adjuvant, and compare its effect with an intramuscular (IM) commercial vaccine (CV). Fifty 24 day-old M. hyopneumoniae-free piglets composed five equal groups for different immunization protocols, consisting of a CV and/or oral immunization (OI). Control piglets did not receive any form of immunization. All piglets were challenged with M. hyopneumoniae strain 232 on D49 by tracheal route. IgA antibody response in the respiratory tract, bacterial shedding and serum IgG were evaluated. The piglets were euthanized on 28 (D77) and 56 (D105) days post-infection. Lung lesions were macroscopically evaluated; lung fragments and bronchoalveolar fluid (BALF) were collected for estimation of bacterial loads by qPCR and/or histopathology examination. All immunization protocols induced reduction on Mycoplasma-like macroscopic lung lesions. IgA Ab responses anti-M. hyopneumoniae, the expression of IL-4 cytokine and a lower expression of IL-8 were induced by CV and OI vaccines, while IgG was induced only by CV. Oral immunization using silica as a carrier-adjuvant can be viable in controlling M. hyopneumoniae infection.
Collapse
Affiliation(s)
- Marina L Mechler-Dreibi
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Henrique M S Almeida
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Karina Sonalio
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Mariela A C Martines
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Fernando A M Petri
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Beatriz B Zambotti
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Marcela M Ferreira
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Gabriel Y Storino
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - Tereza S Martins
- Department of Chemistry, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Hélio J Montassier
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | | | | | - Luís Guilherme de Oliveira
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil.
| |
Collapse
|
14
|
Rohrer B, Parsons N, Annamalai B, Nicholson C, Obert E, Jones BW, Dick AD. Peptide-based immunotherapy against oxidized elastin ameliorates pathology in mouse model of smoke-induced ocular injury. Exp Eye Res 2021; 212:108755. [PMID: 34487725 PMCID: PMC9753162 DOI: 10.1016/j.exer.2021.108755] [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: 06/08/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Age-related macular degeneration (AMD), the leading cause of blindness in western populations, is associated with an overactive complement system, and an increase in circulating antibodies against certain epitopes, including elastin. As loss of the elastin layer of Bruch's membrane (BrM) has been reported in aging and AMD, we previously showed that immunization with elastin peptide oxidatively modified by cigarette smoke (ox-elastin), exacerbated ocular pathology in the smoke-induced ocular pathology (SIOP) model. Here we asked whether ox-elastin peptide-based immunotherapy (PIT) ameliorates damage. METHODS C57BL/6J mice were injected with ox-elastin peptide at two doses via weekly subcutaneous administration, while exposed to cigarette smoke for 6 months. FcγR-/- and uninjected C57BL/6J mice served as controls. Retinal morphology was assessed by electron microscopy, and complement activation, antibody deposition and mechanisms of immunological tolerance were assessed by Western blotting and ELISA. RESULTS Elimination of Fcγ receptors, preventing antigen/antibody-dependent cytotoxicity, protected against SIOP. Mice receiving PIT with low dose ox-elastin (LD-PIT) exhibited reduced humoral immunity, reduced complement activation and IgG/IgM deposition in the RPE/choroid, and largely a preserved BrM. While there is no direct evidence of ox-elastin pathogenicity, LD-PIT reduced IFNγ and increased IL-4 within RPE/choroid. High dose PIT was not protective. CONCLUSIONS These data further support ox-elastin role in ocular damage in part via elastin-specific antibodies, and support the corollary that PIT with ox-elastin attenuates ocular pathology. Overall, damage is associated with complement activation, antibody-dependent cell-mediated cytotoxicity, and altered cytokine signature.
Collapse
Affiliation(s)
- Bärbel Rohrer
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, 29401, USA.
| | - Nathaniel Parsons
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Balasubramaniam Annamalai
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Crystal Nicholson
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Elisabeth Obert
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan W Jones
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, 84132, USA
| | - Andrew D Dick
- University of Bristol, Bristol BS8 1TD, UK and University College London-Institute of Ophthalmology and the National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, London, EC1V 9EL, UK.
| |
Collapse
|
15
|
Azarcoya-Barrera J, Field CJ, Goruk S, Makarowski A, Curtis JM, Pouliot Y, Jacobs RL, Richard C. Buttermilk: an important source of lipid soluble forms of choline that influences the immune system development in Sprague-Dawley rat offspring. Eur J Nutr 2021; 60:2807-2818. [PMID: 33416979 DOI: 10.1007/s00394-020-02462-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine the effect of feeding buttermilk-derived choline metabolites on the immune system development in Sprague-Dawley rat pups. METHODS Sprague-Dawley dams were randomized to one of the three diets containing 1.7 g/kg choline: 1-Control (100% free choline (FC)), 2-Buttermilk (BM, 37% phosphatidylcholine (PC), 34% sphingomyelin (SM), 17% glycerophosphocholine (GPC), 7% FC, 5% phosphocholine), and 3-Placebo (PB, 50% PC, 25% FC, 25% GPC) until the end of the lactation period. At weaning, pups continued on the same diet as their mom. Cell phenotypes and cytokine production by mitogen-stimulated splenocytes isolated from 3- and 10-week-old pups were measured. RESULTS At 3 weeks, BM-pups had a higher proportion of cytotoxic T cells (CTL; CD3 + CD8 +) while both BM- and PB-pups had an increased proportion of cells expressing CD28 + , CD86 + and CD27 + (all p > 0.05). Following ConA stimulation, splenocytes from BM- and PB-pups produced more TNF-α and IFN-γ and after LPS stimulation produced more IL-10 and TNF-α (all p > 0.05). Starting at week 6 of age, BM-pups had a higher body weight. At 10 weeks, both the BM- and PB-pups had a higher proportion of CTL expressing CD27 + . After ConA stimulation, splenocytes from BM- and PB-pups produced more IL-2, IFN-γ and IL-6 and more IL-10 after LPS stimulation (all p > 0.05). CONCLUSION The proportion of lipid soluble forms of choline in the diet during lactation and weaning periods influence the immune system development in rat offspring.
Collapse
Affiliation(s)
- Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Jonathan M Curtis
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, Center for Health Research Innovation, University of Alberta, 4-002G Li Ka Shing, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
16
|
Zhao X, Thijssen S, Chen H, Garssen J, Knippels LMJ, Hogenkamp A. Selenium Modulates the Allergic Response to Whey Protein in a Mouse Model for Cow's Milk Allergy. Nutrients 2021; 13:2479. [PMID: 34444651 PMCID: PMC8400770 DOI: 10.3390/nu13082479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022] Open
Abstract
Cow's milk allergy is a common food allergy in infants, and is associated with an increased risk of developing other allergic diseases. Dietary selenium (Se), one of the essential micronutrients for humans and animals, is an important bioelement which can influence both innate and adaptive immune responses. However, the effects of Se on food allergy are still largely unknown. In the current study it was investigated whether dietary Se supplementation can inhibit whey-induced food allergy in an animal research model. Three-week-old female C3H/HeOuJ mice were intragastrically sensitized with whey protein and cholera toxin and randomly assigned to receive a control, low, medium or high Se diet. Acute allergic symptoms, allergen specific immunoglobulin (Ig) E levels and mast cell degranulation were determined upon whey challenge. Body temperature was significantly higher in mice that received the medium Se diet 60 min after the oral challenge with whey compared to the positive control group, which is indicative of impaired anaphylaxis. This was accompanied by reductions in antigen-specific immunoglobulins and reduced levels of mouse mast cell protease-1 (mMCP-1). This study demonstrates that oral Se supplementation may modulate allergic responses to whey by decreasing specific antibody responses and mMCP-1 release.
Collapse
Affiliation(s)
- Xiaoli Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (X.Z.); (H.C.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (S.T.); (J.G.); (L.M.J.K.)
- School of Food Science Technology, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (S.T.); (J.G.); (L.M.J.K.)
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (X.Z.); (H.C.)
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (S.T.); (J.G.); (L.M.J.K.)
- Danone/Nutricia Research, Global Centre of Excellence Immunology, 3584 CT Utrecht, The Netherlands
| | - Leon M. J. Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (S.T.); (J.G.); (L.M.J.K.)
- Danone/Nutricia Research, Global Centre of Excellence Immunology, 3584 CT Utrecht, The Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (S.T.); (J.G.); (L.M.J.K.)
| |
Collapse
|
17
|
Han X, Ding S, Jiang H, Liu G. Roles of Macrophages in the Development and Treatment of Gut Inflammation. Front Cell Dev Biol 2021; 9:625423. [PMID: 33738283 PMCID: PMC7960654 DOI: 10.3389/fcell.2021.625423] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages, which are functional plasticity cells, have the ability to phagocytize and digest foreign substances and acquire pro-(M1-like) or anti-inflammatory (M2-like) phenotypes according to their microenvironment. The large number of macrophages in the intestinal tract, play a significant role in maintaining the homeostasis of microorganisms on the surface of the intestinal mucosa and in the continuous renewal of intestinal epithelial cells. They are not only responsible for innate immunity, but also participate in the development of intestinal inflammation. A clear understanding of the function of macrophages, as well as their role in pathogens and inflammatory response, will delineate the next steps in the treatment of intestinal inflammatory diseases. In this review, we discuss the origin and development of macrophages and their role in the intestinal inflammatory response or infection. In addition, the effects of macrophages in the occurrence and development of inflammatory bowel disease (IBD), and their role in inducing fibrosis, activating T cells, reducing colitis, and treating intestinal inflammation were also reviewed in this paper.
Collapse
Affiliation(s)
- Xuebing Han
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, China
| | - Hongmei Jiang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, China
| |
Collapse
|
18
|
Yoneyama T, Nakano N, Hara M, Yamada H, Izawa K, Uchida K, Kaitani A, Ando T, Kitaura J, Ohtsuka Y, Ogawa H, Okumura K, Shimizu T. Notch signaling contributes to the establishment of sustained unresponsiveness to food allergens by oral immunotherapy. J Allergy Clin Immunol 2021; 147:1063-1076.e9. [DOI: 10.1016/j.jaci.2020.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 06/02/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
|
19
|
Anvari S, Schuster K, Grimbergen A, Davis CM, Makedonas G. Attenuation of GARP expression on regulatory T cells by protein transport inhibitors. J Immunol Methods 2021; 492:112998. [PMID: 33600819 DOI: 10.1016/j.jim.2021.112998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/02/2020] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
An integrated understanding of the functional capacities of cells in the context of their physical parameters and molecular markers is increasingly demanded in immunologic studies. Regulatory T cells (Tregs) are a subpopulation of T cells involved in immune response modulation and mediating tolerance to self-antigen with their absence leading to a loss of tolerance. Glycoprotein repetitions A predominant (GARP) is a key marker for activated Tregs, but its detection may also be useful in determining the functional capacities of the cell. This study aims to deduce the optimal stimulation period and the impact of protein transport inhibitors (PTIs), commonly used in the detection of intracellular cytokines, on GARP detection. Through flow cytometric analysis we analyzed different cell culture conditions for optimal GARP expression on activated Tregs. Healthy donor PBMCs were stimulated with either Staphylococcal Enterotoxin B (SEB) or PMA/Ionomycin (PMA/Iono), in the presence and absence of PTIs monensin and/or brefeldin A (BFA) and GARP expression was assessed on CD4+ CD25+ FOXP3+ Tregs. The optimal stimulation period for the detection of GARP was highest at 24-h. Furthermore, we determined that GARP expression on Tregs is significantly reduced when cells are treated with the PTIs monensin and/or BFA following PMA/Iono stimulation. This effect was not seen following SEB stimulation. Therefore, due to the effects of PTIs, alternative methods should be considered when performing simultaneous analysis for cytokine expression and GARP expression on Tregs.
Collapse
Affiliation(s)
- Sara Anvari
- Baylor College of Medicine, Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, William T. Shearer Center for Human Immunobiology, 1102 Bates Avenue, Ste. 330, Houston, TX, USA.
| | - Kimberly Schuster
- Baylor College of Medicine, Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, William T. Shearer Center for Human Immunobiology, 1102 Bates Avenue, Ste. 330, Houston, TX, USA.
| | - Andrea Grimbergen
- Baylor College of Medicine, Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, William T. Shearer Center for Human Immunobiology, 1102 Bates Avenue, Ste. 330, Houston, TX, USA.
| | - Carla M Davis
- Baylor College of Medicine, Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, William T. Shearer Center for Human Immunobiology, 1102 Bates Avenue, Ste. 330, Houston, TX, USA.
| | - George Makedonas
- Baylor College of Medicine, Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, William T. Shearer Center for Human Immunobiology, 1102 Bates Avenue, Ste. 330, Houston, TX, USA.
| |
Collapse
|
20
|
Knoop KA, McDonald KG, Hsieh CS, Tarr PI, Newberry RD. Regulatory T Cells Developing Peri-Weaning Are Continually Required to Restrain Th2 Systemic Responses Later in Life. Front Immunol 2021; 11:603059. [PMID: 33613522 PMCID: PMC7891039 DOI: 10.3389/fimmu.2020.603059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
Atopic disorders including allergic rhinitis, asthma, food allergy, and dermatitis, are increasingly prevalent in Western societies. These disorders are largely characterized by T helper type 2 (Th2) immune responses to environmental triggers, particularly inhaled and dietary allergens. Exposure to such stimuli during early childhood reduces the frequency of allergies in at-risk children. These allergic responses can be restrained by regulatory T cells (Tregs), particularly Tregs arising in the gut. The unique attributes of how early life exposure to diet and microbes shape the intestinal Treg population is a topic of significant interest. While imprinting during early life promotes the development of a balanced immune system and protects against immunopathology, it remains unclear if Tregs that develop in early life continue to restrain systemic inflammatory responses throughout adulthood. Here, an inducible deletion strategy was used to label Tregs at specified time points with a targeted mechanism to be deleted later. Deletion of the Tregs labeled peri-weaning at day of life 24, but not before weaning at day of life 14, resulted in increased circulating IgE and IL-13, and abrogated induction of tolerance towards new antigens. Thus, Tregs developing peri-weaning, but not before day of life 14 are continually required to restrain allergic responses into adulthood.
Collapse
MESH Headings
- Administration, Oral
- Adoptive Transfer
- Age Factors
- Animals
- Animals, Genetically Modified
- Antigens/administration & dosage
- Antigens/immunology
- Cell Communication
- Colon/immunology
- Colon/metabolism
- Cytokines/blood
- Disease Models, Animal
- Hypersensitivity, Delayed/blood
- Hypersensitivity, Delayed/genetics
- Hypersensitivity, Delayed/immunology
- Immune Tolerance
- Immunoglobulin E/blood
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Ovalbumin
- Phenotype
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Weaning
- Mice
Collapse
Affiliation(s)
- Kathryn A. Knoop
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Keely G. McDonald
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Phillip I. Tarr
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Pediatrics and Molecular Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Rodney D. Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
21
|
Bertolini TB, Biswas M, Terhorst C, Daniell H, Herzog RW, Piñeros AR. Role of orally induced regulatory T cells in immunotherapy and tolerance. Cell Immunol 2020; 359:104251. [PMID: 33248367 DOI: 10.1016/j.cellimm.2020.104251] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/30/2020] [Accepted: 11/01/2020] [Indexed: 12/24/2022]
Abstract
Oral antigen administration to induce regulatory T cells (Treg) takes advantage of regulatory mechanisms that the gastrointestinal tract utilizes to promote unresponsiveness against food antigens or commensal microorganisms. Recently, antigen-based oral immunotherapies (OITs) have shown efficacy as treatment for food allergy and autoimmune diseases. Similarly, OITs appear to prevent anti-drug antibody responses in replacement therapy for genetic diseases. Intestinal epithelial cells and microbiota possibly condition dendritic cells (DC) toward a tolerogenic phenotype that induces Treg via expression of several mediators, e.g. IL-10, transforming growth factor-β, retinoic acid. Several factors, such as metabolites derived from microbiota or diet, impact the stability and expansion of these induced Treg, which include, but are not limited to, FoxP3+ Treg, LAP+ Treg, and/or Tr1 cells. Here, we review various orally induced Treg, their plasticity and cooperation between the Treg subsets, as well as underlying mechanisms controlling their induction and role in oral tolerance.
Collapse
Affiliation(s)
- Thais B Bertolini
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Annie R Piñeros
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
22
|
Mu Q, Edwards MR, Swartwout BK, Cabana Puig X, Mao J, Zhu J, Grieco J, Cecere TE, Prakash M, Reilly CM, Puglisi C, Bachali P, Grammer AC, Lipsky PE, Luo XM. Gut Microbiota and Bacterial DNA Suppress Autoimmunity by Stimulating Regulatory B Cells in a Murine Model of Lupus. Front Immunol 2020; 11:593353. [PMID: 33240280 PMCID: PMC7683516 DOI: 10.3389/fimmu.2020.593353] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022] Open
Abstract
Autoimmune diseases, such as systemic lupus erythematosus, are characterized by excessive inflammation in response to self-antigens. Loss of appropriate immunoregulatory mechanisms contribute to disease exacerbation. We previously showed the suppressive effect of vancomycin treatment during the "active-disease" stage of lupus. In this study, we sought to understand the effect of the same treatment given before disease onset. To develop a model in which to test the regulatory role of the gut microbiota in modifying autoimmunity, we treated lupus-prone mice with vancomycin in the period before disease development (3-8 weeks of age). We found that administration of vancomycin to female MRL/lpr mice early, only during the pre-disease period but not from 3 to 15 weeks of age, led to disease exacerbation. Early vancomycin administration also reduced splenic regulatory B (Breg) cell numbers, as well as reduced circulating IL-10 and IL-35 in 8-week old mice. Further, we found that during the pre-disease period, administration of activated IL-10 producing Breg cells to mice treated with vancomycin suppressed lupus initiation, and that bacterial DNA from the gut microbiota was an inducer of Breg function. Oral gavage of bacterial DNA to mice treated with vancomycin increased Breg cells in the spleen and mesenteric lymph node at 8 weeks of age and reduced autoimmune disease severity at 15 weeks. This work suggests that a form of oral tolerance induced by bacterial DNA-mediated expansion of Breg cells suppress disease onset in the autoimmune-prone MRL/lpr mouse model. Future studies are warranted to further define the mechanism behind bacterial DNA promoting Breg cells.
Collapse
Affiliation(s)
- Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Michael R. Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Brianna K. Swartwout
- Translational Biology, Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, United States
| | - Xavier Cabana Puig
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Jiangdi Mao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Jing Zhu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Joe Grieco
- Translational Biology, Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, United States
| | - Thomas E. Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Meeta Prakash
- Carilion School of Medicine, Virginia Tech, Roanoke, VA, United States
| | | | | | | | | | | | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
23
|
Wróblewska B, Kaliszewska-Suchodoła A, Fuc E, Markiewicz LH, Ogrodowczyk AM, Złotkowska D, Wasilewska E. Effect of Low-Immunogenic Yogurt Drinks and Probiotic Bacteria on Immunoreactivity of Cow's Milk Proteins and Tolerance Induction-In Vitro and In Vivo Studies. Nutrients 2020; 12:E3390. [PMID: 33158132 PMCID: PMC7694189 DOI: 10.3390/nu12113390] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
There is no effective therapy for milk allergy. The role of lactic acid bacteria (LAB) and probiotics in protection against allergy-related outcomes is still under investigation. The aim of the study was to evaluate the immunomodulative and therapeutic potential of yogurt drinks in cow's milk allergy (CMA) management. We compared immunoreactivity of α-casein (α-CN), β-casein (β-CN), κ-casein (κ-CN), α-lactalbumin (α-LA), and β-lactoglobulin (β-LG) in 27 yogurt drinks fermented with different basic yogurt cultures, or yogurt cultures enriched with Lactobacillus plantarum and/or Bifidobacterium lactis strains, by competitive ELISA assay. Drinks with the lowest antigenic potential were used as allergoids for CMA therapy. BALB/c mice were sensitized via intraperitoneal injection of α-CN + β-LG mixture with aluminum adjuvant, and gavaged with increasing doses of selected low-immunogenic drinks (YM-basic, or YM-LB-enriched with L. plantarum and B. lactis) to induce tolerance. Milk- or phosphate-buffered saline (PBS)-dosed mice served as controls. Compared to milk, the immunoreactivity of proteins in drinks increased or decreased, depending on the bacterial sets applied for fermentation. Only a few sets acted synergistically in reducing immunoreactivity. The selected low-immunogenic drinks stimulated allergic mice for profiling Th2 to Th1 response and acquire tolerance, and the effect was greater with YM-LB drink, which during long-lasting interventional feeding strongly increased the secretion of regulatory cytokines, i.e., IL-10 and TGF-β, and IgA and decreased IL-4, IgE, and anti-(α-CN + β-LG) IgG1. The studies revealed variations in the potency of yogurt bacteria to change allergenicity of milk proteins and the need for their strict selection to obtain a safe product for allergy sufferers. The YM-LB drink with reduced antigenic potential may be a source of allergoids used in the immunotherapy of IgE mediated CMA, but further clinical or volunteer studies are required.
Collapse
Affiliation(s)
- Barbara Wróblewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | | | - Ewa Fuc
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Lidia Hanna Markiewicz
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Anna Maria Ogrodowczyk
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Dagmara Złotkowska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Ewa Wasilewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| |
Collapse
|
24
|
Sosa AC, Kariuki B, Gan Q, Knutsen AP, Bellone CJ, Guzmán MA, Barrera LA, Tomatsu S, Chauhan AK, Armbrecht E, Montaño AM. Oral immunotherapy tolerizes mice to enzyme replacement therapy for Morquio A syndrome. J Clin Invest 2020; 130:1288-1300. [PMID: 31743109 DOI: 10.1172/jci125607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Immune response to therapeutic enzymes poses a detriment to patient safety and treatment outcome. Enzyme replacement therapy (ERT) is a standard therapeutic option for some types of mucopolysaccharidoses, including Morquio A syndrome caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency. Current protocols tolerize patients using cytotoxic immunosuppressives, which can cause adverse effects. Here we show development of tolerance in Morquio A mice via oral delivery of peptide or GALNS for 10 days prior to ERT. Our results show that using an immunodominant peptide (I10) or the complete GALNS enzyme to orally induce tolerance to GALNS prior to ERT resulted in several improvements to ERT in mice: (a) decreased splenocyte proliferation after in vitro GALNS stimulation, (b) modulation of the cytokine secretion profile, (c) decrease in GALNS-specific IgG or IgE in plasma, (d) decreased GAG storage in liver, and (e) fewer circulating immune complexes in plasma. This model could be extrapolated to other lysosomal storage disorders in which immune response hinders ERT.
Collapse
Affiliation(s)
- Angela C Sosa
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Barbara Kariuki
- Department of Pediatrics, Division of Allergy and Immunology
| | - Qi Gan
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Alan P Knutsen
- Department of Pediatrics, Division of Allergy and Immunology
| | | | - Miguel A Guzmán
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Luis A Barrera
- Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Rheumatology, School of Medicine
| | | | - Adriana M Montaño
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Iberg CA, Hawiger D. Natural and Induced Tolerogenic Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:733-744. [PMID: 32015076 DOI: 10.4049/jimmunol.1901121] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are highly susceptible to extrinsic signals that modify the functions of these crucial APCs. Maturation of DCs induced by diverse proinflammatory conditions promotes immune responses, but certain signals also induce tolerogenic functions in DCs. These "induced tolerogenic DCs" help to moderate immune responses such as those to commensals present at specific anatomical locations. However, also under steady-state conditions, some DCs are characterized by inherent tolerogenic properties. The immunomodulatory mechanisms constitutively present in such "natural tolerogenic DCs" help to promote tolerance to peripheral Ags. By extending tolerance initially established in the thymus, these functions of DCs help to regulate autoimmune and other immune responses. In this review we will discuss the mechanisms and functions of natural and induced tolerogenic DCs and offer further insight into how their possible manipulations may ultimately lead to more precise treatments for various immune-mediated conditions and diseases.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
26
|
Caër C, Wick MJ. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front Immunol 2020; 11:410. [PMID: 32256490 PMCID: PMC7093381 DOI: 10.3389/fimmu.2020.00410] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex immune-mediated disease of the gastrointestinal tract that increases morbidity and negatively influences the quality of life. Intestinal mononuclear phagocytes (MNPs) have a crucial role in maintaining epithelial barrier integrity while controlling pathogen invasion by activating an appropriate immune response. However, in genetically predisposed individuals, uncontrolled immune activation to intestinal flora is thought to underlie the chronic mucosal inflammation that can ultimately result in IBD. Thus, MNPs are involved in fine-tuning mucosal immune system responsiveness and have a critical role in maintaining homeostasis or, potentially, the emergence of IBD. MNPs include monocytes, macrophages and dendritic cells, which are functionally diverse but highly complementary. Despite their crucial role in maintaining intestinal homeostasis, specific functions of human MNP subsets are poorly understood, especially during diseases such as IBD. Here we review the current understanding of MNP ontogeny, as well as the recently identified human intestinal MNP subsets, and discuss their role in health and IBD.
Collapse
Affiliation(s)
- Charles Caër
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
27
|
van Sadelhoff JHJ, Hogenkamp A, Wiertsema SP, Harthoorn LF, Loonstra R, Hartog A, Garssen J. A free amino acid-based diet partially prevents symptoms of cow's milk allergy in mice after oral sensitization with whey. Immun Inflamm Dis 2020; 8:93-105. [PMID: 32031763 PMCID: PMC7016843 DOI: 10.1002/iid3.288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Amino acid-based formulas (AAFs) are used for the dietary management of cow's milk allergy (CMA). Whether AAFs have the potential to prevent the development and/or symptoms of CMA is not known. OBJECTIVE The present study evaluated the preventive effects of an amino acid (AA)-based diet on allergic sensitization and symptoms of CMA in mice and aimed to provide insight into the underlying mechanism. METHODS C3H/HeOuJ mice were sensitized with whey protein or with phosphate-buffered saline as sham-sensitized control. Starting 2 weeks before sensitization, mice were fed with either a protein-based diet or an AA-based diet with an AA composition based on that of the AAF Neocate, a commercially available AAF prescribed for the dietary management of CMA. Upon challenge, allergic symptoms, mast cell degranulation, whey-specific immunoglobulin levels, and FoxP3+ cell counts in jejunum sections were assessed. RESULTS Compared to mice fed with the protein-based diet, AA-fed mice had significantly lower acute allergic skin responses. Moreover, the AA-based diet prevented the whey-induced symptoms of anaphylaxis and drop in body temperature. Whereas the AA-based diet had no effect on the levels of serum IgE and mucosal mast cell protease-1 (mMCP-1), AA-fed mice had significantly lower serum IgG2a levels and tended to have lower IgG1 levels (P = .076). In addition, the AA-based diet prevented the whey-induced decrease in FoxP3+ cells. In sham-sensitized mice, no differences between the two diets were observed in any of the tested parameters. CONCLUSION This study demonstrates that an AA-based diet can at least partially prevent allergic symptoms of CMA in mice. Differences in FoxP3+ cell counts and serum levels of IgG2a and IgG1 may suggest enhanced anti-inflammatory and tolerizing capacities in AA-fed mice. This, combined with the absence of effects in sham-sensitized mice indicates that AAFs for the prevention of food allergies may be an interesting concept that warrants further research.
Collapse
Affiliation(s)
- Joris H. J. van Sadelhoff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | | | | | | | - Anita Hartog
- CeO ImmunologyDanone Nutricia ResearchUtrechtThe Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
- CeO ImmunologyDanone Nutricia ResearchUtrechtThe Netherlands
| |
Collapse
|
28
|
Abstract
In the past decade, nonalcoholic fatty liver disease (NAFLD) has become a leading cause of chronic liver disease and cirrhosis, as well as an important risk factor for hepatocellular carcinoma (HCC). NAFLD encompasses a spectrum of liver lesions, including simple steatosis, steatohepatitis and fibrosis. Although steatosis is often harmless, the lobular inflammation that characterizes nonalcoholic steatohepatitis (NASH) is considered a driving force in the progression of NAFLD. The current view is that innate immune mechanisms represent a key element in supporting hepatic inflammation in NASH. However, increasing evidence points to the role of adaptive immunity as an additional factor promoting liver inflammation. This Review discusses data regarding the role of B cells and T cells in sustaining the progression of NASH to fibrosis and HCC, along with the findings that antigens originating from oxidative stress act as a trigger for immune responses. We also highlight the mechanisms affecting liver immune tolerance in the setting of steatohepatitis that favour lymphocyte activation. Finally, we analyse emerging evidence concerning the possible application of immune modulating treatments in NASH therapy.
Collapse
Affiliation(s)
- Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy.
| |
Collapse
|
29
|
Salazar A, Nieto JE, Velazquez-Soto H, Jiménez-Martínez MC. Activation of IL-10+ B cells: A novel immunomodulatory mechanism for therapeutic bacterial suspensions. SAGE Open Med 2020; 8:2050312120901547. [PMID: 32002185 PMCID: PMC6963315 DOI: 10.1177/2050312120901547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/23/2019] [Indexed: 01/22/2023] Open
Abstract
Objectives: Bacterial components are used to improve immune responses in patients with respiratory infections. Pharmacological formulations of bacterial components include a mixture of bacterial antigens, some of which are complete inactivated bacteria, that is, named bacterial suspensions; while others are fragments of bacteria, which are presented as bacterial lysates. Although bacterial lysates have been broadly used as immune-stimulators, the biological support for the therapeutic effectiveness of bacterial suspension has not yet been studied. Thus, the aim of our study was to investigate the immunological activity induced by bacterial suspension. Methods: This work was an exploratory translational study. Peripheral blood mononuclear cells were obtained from healthy donors and cultured in time–dose dependent assays with a commercial bacterial suspension. Flow cytometry was used for phenotypic analysis and for determining soluble cytokines in culture supernatants. Results: We observed that bacterial suspension activates B cells in a dose-dependent manner. Peripheral blood mononuclear cells were able to secrete IL-6 and IL-10 after 24 h of bacterial suspension stimulation. TLR2 expression was observed mainly on CD19+ CD38Lo B cells after 72 h of culture; remarkably, most of the TLR2+ CD19+ cells were also IL-10+. Conclusion: Our findings suggest that bacterial suspension induces the activation of B cell subsets as well as the secretion of IL-6 and IL-10. Expression of TLR2 on CD19+ cells could act as an activation loop of IL-10+ B regulatory cells. The clinical implications of these findings are discussed at the end of this article.
Collapse
Affiliation(s)
- Alberto Salazar
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.,Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Jane E Nieto
- Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Henry Velazquez-Soto
- Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Maria C Jiménez-Martínez
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.,Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| |
Collapse
|
30
|
Kim BG, Kim JN, Jang AS, Shin M. Combined Effects of Lactobacillus rhamnosus and Egg Oral Immunotherapy in a Mouse Model of Egg Allergy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:701-711. [PMID: 32400134 PMCID: PMC7225004 DOI: 10.4168/aair.2020.12.4.701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 01/22/2023]
Abstract
Purpose Recent clinical trials have successfully used oral immunotherapy (OIT) to treat food allergies. Probiotics have immunomodulatory effects by balancing Th1/Th2 immunity and enhancing regulatory T-cell activity. In this study, we analyzed the effects of OIT, probiotics alone, and probiotics administered simultaneously with OIT in a mouse model of egg allergy. Methods C3H/HeJ mice were sensitized by intragastric administration of ovomucoid (OM) with cholera toxin. For the OIT regime, increasing doses of OM were administered orally to sensitized mice. Lactobacillus casei variety ramnosus (Lcr35) was also administered. The mice were divided into 4 groups: control (no OIT), OIT, Lcr35, and OIT plus Lcr35 (OIT + Lcr35). The effects of OIT and Lcr35 treatment were estimated based on the symptom score, rectal temperature, serum levels of OM-specific immunoglobulin (Ig)E, IgA, IgG1, and IgG2a immediately after and 2 weeks after ceasing treatment and histological staining of the small intestine. Results The severity of anaphylaxis decreased in all treatment groups. Simultaneous administration of Lcr35 and OIT decreased the severity of anaphylaxis compared to controls and the OIT group. The protective effects were sustained 2 weeks after ceasing treatment in all treatment groups. A significant decrease in OM-specific IgA, IgG1, and IgG2a levels was observed in both the OIT and OIT plus Lcr35 groups. However, a significant decrease in the OM-specific IgE level was observed only in OIT plus Lcr35 treated mice and was sustained 2 weeks after ceasing treatment. Mucin amounts in the small intestine decreased after OIT, OIT plus Lcr35, and Lcr35 treatment with the lowest in the OIT plus Lcr35 group. Conclusions Lcr35 treatment during OIT had some synergic effect for protection against anaphylaxis in a mice model of egg allergy. These findings should be confirmed in future animal studies including more detailed immunological profiles and human studies.
Collapse
Affiliation(s)
- Byeong Gon Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - Ji Na Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - An Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - Meeyong Shin
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea.
| |
Collapse
|
31
|
Martin K, Touil R, Kolb Y, Cvijetic G, Murakami K, Israel L, Duraes F, Buffet D, Glück A, Niwa S, Bigaud M, Junt T, Zamurovic N, Smith P, McCoy KD, Ohashi PS, Bornancin F, Calzascia T. Malt1 Protease Deficiency in Mice Disrupts Immune Homeostasis at Environmental Barriers and Drives Systemic T Cell-Mediated Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2019; 203:2791-2806. [PMID: 31659015 DOI: 10.4049/jimmunol.1900327] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
The paracaspase Malt1 is a key regulator of canonical NF-κB activation downstream of multiple receptors in both immune and nonimmune cells. Genetic disruption of Malt1 protease function in mice and MALT1 mutations in humans results in reduced regulatory T cells and a progressive multiorgan inflammatory pathology. In this study, we evaluated the altered immune homeostasis and autoimmune disease in Malt1 protease-deficient (Malt1PD) mice and the Ags driving disease manifestations. Our data indicate that B cell activation and IgG1/IgE production is triggered by microbial and dietary Ags preferentially in lymphoid organs draining mucosal barriers, likely as a result of dysregulated mucosal immune homeostasis. Conversely, the disease was driven by a polyclonal T cell population directed against self-antigens. Characterization of the Malt1PD T cell compartment revealed expansion of T effector memory cells and concomitant loss of a CD4+ T cell population that phenotypically resembles anergic T cells. Therefore, we propose that the compromised regulatory T cell compartment in Malt1PD animals prevents the efficient maintenance of anergy and supports the progressive expansion of pathogenic, IFN-γ-producing T cells. Overall, our data revealed a crucial role of the Malt1 protease for the maintenance of intestinal and systemic immune homeostasis, which might provide insights into the mechanisms underlying IPEX-related diseases associated with mutations in MALT1.
Collapse
Affiliation(s)
- Kea Martin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Ratiba Touil
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Yeter Kolb
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Grozdan Cvijetic
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Kiichi Murakami
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Laura Israel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Fernanda Duraes
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - David Buffet
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Anton Glück
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Satoru Niwa
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Marc Bigaud
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Natasa Zamurovic
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Philip Smith
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Kathy D McCoy
- Department of Clinical Research, University Clinic for Visceral Surgery and Medicine, University Hospital, 3010 Bern, Switzerland; and
| | - Pamela S Ohashi
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2M9, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5G 2C1, Canada
| | - Frédéric Bornancin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Thomas Calzascia
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland;
| |
Collapse
|
32
|
Untersmayr E, Bax HJ, Bergmann C, Bianchini R, Cozen W, Gould HJ, Hartmann K, Josephs DH, Levi‐Schaffer F, Penichet ML, O'Mahony L, Poli A, Redegeld FA, Roth‐Walter F, Turner MC, Vangelista L, Karagiannis SN, Jensen‐Jarolim E. AllergoOncology: Microbiota in allergy and cancer-A European Academy for Allergy and Clinical Immunology position paper. Allergy 2019; 74:1037-1051. [PMID: 30636005 PMCID: PMC6563061 DOI: 10.1111/all.13718] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
The microbiota can play important roles in the development of human immunity and the establishment of immune homeostasis. Lifestyle factors including diet, hygiene, and exposure to viruses or bacteria, and medical interventions with antibiotics or anti-ulcer medications, regulate phylogenetic variability and the quality of cross talk between innate and adaptive immune cells via mucosal and skin epithelia. More recently, microbiota and their composition have been linked to protective effects for health. Imbalance, however, has been linked to immune-related diseases such as allergy and cancer, characterized by impaired, or exaggerated immune tolerance, respectively. In this AllergoOncology position paper, we focus on the increasing evidence defining the microbiota composition as a key determinant of immunity and immune tolerance, linked to the risk for the development of allergic and malignant diseases. We discuss novel insights into the role of microbiota in disease and patient responses to treatments in cancer and in allergy. These may highlight opportunities to improve patient outcomes with medical interventions supported through a restored microbiome.
Collapse
Affiliation(s)
- Eva Untersmayr
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
| | - Heather J. Bax
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | | | - Rodolfo Bianchini
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Wendy Cozen
- Center for Genetic EpidemiologyDepartment of Preventive MedicineKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of PathologyKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterKeck School of Medicine of Los AngelesLos AngelesCaliforniaUSA
| | - Hannah J. Gould
- Randall Centre for Cell and Molecular BiophysicsSchool of Basic & Medical BiosciencesKing's College LondonNew Hunt's HouseLondonUK
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Karin Hartmann
- Department of DermatologyUniversity of LuebeckLuebeckGermany
| | - Debra H. Josephs
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | - Francesca Levi‐Schaffer
- Pharmacology and Experimental Therapeutics UnitSchool of PharmacyFaculty of MedicineThe Institute for Drug ResearchThe Hebrew University of JerusalemJerusalemIsrael
| | - Manuel L. Penichet
- Division of Surgical OncologyDepartment of SurgeryDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Department of Microbiology, Immunology and Molecular GeneticsDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCaliforniaUSA
- The Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
- UCLA AIDS InstituteLos AngelesCaliforniaUSA
| | - Liam O'Mahony
- Departments of Medicine and MicrobiologyAPC Microbiome IrelandNational University of IrelandCorkIreland
| | - Aurelie Poli
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Frank A. Redegeld
- Division of PharmacologyFaculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Franziska Roth‐Walter
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Michelle C. Turner
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- McLaughlin Centre for Population Health Risk AssessmentUniversity of OttawaOttawaOntarioCanada
| | - Luca Vangelista
- Department of Biomedical SciencesNazarbayev University School of MedicineAstanaKazakhstan
| | - Sophia N. Karagiannis
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
| | - Erika Jensen‐Jarolim
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| |
Collapse
|
33
|
Chikhi A, Elmecherfi KE, Bernard H, Cortes-Perez N, Kheroua O, Saidi D, Adel-Patient K. Evaluation of the efficiency of hydrolyzed whey formula to prevent cow's milk allergy in the BALB/c mouse model. Pediatr Allergy Immunol 2019; 30:370-377. [PMID: 30672606 DOI: 10.1111/pai.13017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Partially hydrolyzed milk formulas have been proposed for primary prevention in at-risk infants, but evidence of their efficiency and elucidation of the underlying mechanisms are still lacking. Thanks to a Th2-biased mouse model mimicking at-risk patients, we aimed to assess the potency of a partially hydrolyzed whey formula (pHWF) to induce oral tolerance thus preventing further cow's milk (CM) allergy. METHODS BALB/c mice were gavaged with pHWF, standard milk formula (SF), or vehicle only (PBS+). All mice were then orally sensitized to CM using cholera toxin and further chronically exposed to CM. Humoral (IgE, IgG1, IgG2a) and cellular (Th2/Th1/Th17 cytokine secretion; frequency of CD4+GATA3+ and CD4+CD25+Foxp3+ T cells in the spleen) responses against β-lactoglobulin (BLG) and whole caseins (CAS) were assessed, as well as a marker of elicitation of allergic reaction (mMCP-1) released after an oral challenge with CM. RESULTS All markers of sensitization and of allergic reaction were evidenced in the PBS+ mice and were significantly enhanced upon chronic exposure. Gavage with SF totally and durably prevented sensitization and elicitation of the allergic reaction. Conversely, pre-treatment with pHWF only reduced BLG-specific sensitization (IgE, Th2 cytokines), with no significant effect on sensitization to caseins. However, pHWF pre-treatment significantly reduced mMCP-1 concentration in plasma after CM challenges. CD4+CD25+Foxp3+ Treg cell frequency could not be correlated with tolerance efficiency. CONCLUSION Partially hydrolyzed whey formula only partially prevents the further development of CM allergy in this Th2-biased model. A hydrolysate from both whey and casein fractions may be more efficient.
Collapse
Affiliation(s)
- Amina Chikhi
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Kamel-Eddine Elmecherfi
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Hervé Bernard
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Naima Cortes-Perez
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Omar Kheroua
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Djamel Saidi
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université Oran 1 Ahmed Ben Bella, Oran, Algérie
| | - Karine Adel-Patient
- Laboratoire d'Immuno-Allergie Alimentaire, UMR Service de Pharmacologie et d'Immunoanalyse, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
34
|
Delayed-Type Hypersensitivity Underlying Casein Allergy Is Suppressed by Extracellular Vesicles Carrying miRNA-150. Nutrients 2019; 11:nu11040907. [PMID: 31018604 PMCID: PMC6521277 DOI: 10.3390/nu11040907] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022] Open
Abstract
In patients with non-IgE-mediated milk allergy, a cellular mechanism of delayed-type hypersensitivity (DTH) is considered. Recent findings prove that cell-mediated reactions can be antigen-specifically inhibited by extracellular vesicles (EVs) carrying miRNA-150. We sought to establish a new mouse model of DTH to casein and test the possibility of antigen-specific suppression of the inflammatory reaction. To produce soluble antigenic peptides, casein was subjected to alkaline hydrolysis. DTH reaction to casein was induced in CBA, C57BL/6, and BALB/c mice by intradermal (id) injection of the antigen. Cells collected from spleens and lymph nodes were positively or negatively selected and transferred to naive recipients intravenously (iv). CBA mice were tolerized by iv injection of mouse erythrocytes conjugated with casein antigen and following id immunization with the same antigen. Suppressive EVs were harvested from cell cultures and serum of tolerized donors by means of ultrafiltration and ultracentrifugation for further therapeutic utilization. The newly established mouse model of DTH to casein was mediated by CD4+ Th1 cells and macrophages, while EVs produced by casein-tolerized animals effectively suppressed effector cell response, in an miRNA-150-dependent manner. Altogether, our observations contribute to the current understanding of non-IgE-mediated allergy to casein and of the possibilities to downregulate this reaction.
Collapse
|
35
|
Rodríguez-Bolaños M, Perez-Montfort R. Medical and Veterinary Importance of the Moonlighting Functions of Triosephosphate Isomerase. Curr Protein Pept Sci 2019; 20:304-315. [DOI: 10.2174/1389203719666181026170751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Abstract
Triosephosphate isomerase is the fifth enzyme in glycolysis and its canonical function is the
reversible isomerization of glyceraldehyde-3-phosphate and dihydroxyacetone phosphate. Within the
last decade multiple other functions, that may not necessarily always involve catalysis, have been described.
These include variations in the degree of its expression in many types of cancer and participation
in the regulation of the cell cycle. Triosephosphate isomerase may function as an auto-antigen and
in the evasion of the immune response, as a factor of virulence of some organisms, and also as an important
allergen, mainly in a variety of seafoods. It is an important factor to consider in the cryopreservation
of semen and seems to play a major role in some aspects of the development of Alzheimer's disease. It
also seems to be responsible for neurodegenerative alterations in a few cases of human triosephosphate
isomerase deficiency. Thus, triosephosphate isomerase is an excellent example of a moonlighting protein.
Collapse
Affiliation(s)
- Mónica Rodríguez-Bolaños
- Departamento de Bioquimica y Biologia Estructural, Instituto de Fisiologia Celular, Universidad Nacional Autonoma de Mexico, Av. Universidad 3000, Coyoacan, 04510 Mexico DF, Mexico
| | - Ruy Perez-Montfort
- Departamento de Bioquimica y Biologia Estructural, Instituto de Fisiologia Celular, Universidad Nacional Autonoma de Mexico, Av. Universidad 3000, Coyoacan, 04510 Mexico DF, Mexico
| |
Collapse
|
36
|
Introduction of fish and other foods during infancy and risk of asthma in the All Babies In Southeast Sweden cohort study. Eur J Pediatr 2019; 178:395-402. [PMID: 30617650 PMCID: PMC6437126 DOI: 10.1007/s00431-018-03312-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 01/15/2023]
Abstract
The etiology of asthma includes lifestyle factors. Breastfeeding and introduction of complementary foods have been suggested to affect asthma risk, but the scientific foundation is not solid. Children from the birth cohort All Babies In Southeast Sweden study were included (n = 9727). Breastfeeding duration and timing of introduction of infant formula and food were collected prospectively during the first year. Through linkage to the Swedish Patient Register, 948 children were identified with any asthma until age 15-17 years, of which 450 cases were atopic. Breastfeeding duration was not associated to risk of asthma. Introduction of infant formula earlier than at 14 weeks of age was associated with higher risk of non-atopic asthma. Introduction of fish before 43 weeks of age, as compared to later, was associated with a lower risk of asthma, irrespective of atopic classification. Reverse causation was accounted for but did not explain the results.Conclusion: Introduction of infant formula and timing of introduction of fish seem to impact the long-term risk of doctor-diagnosed asthma. Emphasis on the growing body of evidence that early introduction of allergens offers protection against atopic disease should be considered in future recommendations. What is Known: • Breastfeeding and introduction of complementary foods might influence the risk of atopic diseases. Recently, a review stated that more studies are needed to clarify the role for asthma development. What is New: • Introduction of infant formula earlier than at 14 weeks of age was associated with a higher risk of developing non-atopic asthma. The risk was pronounced in children introduced to infant formula before 4 weeks of age while being breastfed less than 4 weeks. • Early fish introduction, before 43 weeks of age, was associated with a lower risk of asthma, independent of atopic classification of asthma.
Collapse
|
37
|
Xian M, Wang K, Lou H, Wang Y, Zhang L, Wang C. Short-term Haze Exposure Predisposes Healthy Volunteers to Nasal Inflammation. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2019; 11:632-643. [PMID: 31332975 PMCID: PMC6658405 DOI: 10.4168/aair.2019.11.5.632] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/10/2019] [Accepted: 03/26/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE This study aimed to investigate the impact of short-term haze exposure on nasal inflammation in healthy volunteers. METHODS Thirty-three healthy university students were assessed for nasal symptoms, nasal patency, upper and lower respiratory tract nitric oxide (NO) as well as inflammatory mediators and neuropeptides in nasal secretions before and after a 5-day haze episode. Peripheral blood mononuclear cells (PBMCs) were stimulated with particulate matter with an aerodynamic diameter of less than 2.5 μm (PM2.5), and cytokines in the supernatants were examined. RESULTS Mild nasal symptoms were reported by some participants during the haze episode. Objective measures of nasal patency demonstrated that nasal airway resistance was significantly increased from baseline levels, while nasal cavity volume and minimum cross-sectional area were significantly decreased. Similarly, the levels of nasal and exhaled NO, eotaxin, interleukin (IL)-5, chemokine (C-C motif) ligand 17, IL-8, substance P, nerve growth factor and vasoactive intestinal peptides in nasal secretions were significantly increased from baseline values following the haze episode. In contrast, the levels of interferon-γ, IL-10, transforming growth factor-β and neuropeptide Y were significantly decreased. Incubation with 0.1-10 μg/mL PM2.5 significantly increased release of IL-1β, IL-4, IL-5, IL-8 and IL-10 from PBMCs. CONCLUSIONS Short-term haze exposure may lead to nasal inflammation and hypersensitivity in healthy subjects predominantly by Th2 cytokine-mediated immune responses.
Collapse
Affiliation(s)
- Mu Xian
- Beijing Institute of Otolaryngology, Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Kuiji Wang
- Beijing Institute of Otolaryngology, Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Hongfei Lou
- Beijing Institute of Otolaryngology, Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Yang Wang
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Luo Zhang
- Beijing Institute of Otolaryngology, Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.
| | - Chengshuo Wang
- Beijing Institute of Otolaryngology, Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.
| |
Collapse
|
38
|
Bernaldo de Quiros E, Seoane-Reula E, Alonso-Lebrero E, Pion M, Correa-Rocha R. The role of regulatory T cells in the acquisition of tolerance to food allergens in children. Allergol Immunopathol (Madr) 2018; 46:612-618. [PMID: 29739687 DOI: 10.1016/j.aller.2018.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 01/03/2023]
Abstract
Food allergy is a pathological immune reaction that identifies certain harmless food proteins, usually tolerated by the majority of the people, as a threat. The prevalence of these food allergies is increasing worldwide and currently affects 8% of children. Exacerbated reactions to milk, egg and peanut are the most frequent in the pediatric population. It is well known that allergic diseases are a type 2 T-helper (Th2) immune response, characterized by the elevated production of IgE antibodies. However, little is known about the immune mechanisms responsible for the development of clinical tolerance toward food allergens. Recent studies have suggested the key role of regulatory T cells (Tregs) in controlling allergic inflammation. In this review, we discuss the importance of Tregs in the pathogenesis of food allergy and the acquisition of oral tolerance in children. Further investigation in this area will be crucial for the identification of predictive markers and the development of new therapies, which will represent a clinical and social benefit for these allergic diseases.
Collapse
|
39
|
Gonzaga TA, Alves FA, Cheik MFA, de Barros CP, Rezende ERMA, Segundo GRS. Low efficacy of atopy patch test in predicting tolerance development in non-IgE-mediated cow's milk allergy. Allergol Immunopathol (Madr) 2018; 46:241-246. [PMID: 29031891 DOI: 10.1016/j.aller.2017.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/05/2017] [Accepted: 07/25/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND The food atopy patch (APT) test has been used in previous studies to help the diagnosis of non-IgE mediated food allergies (FA). The aim of this study was to evaluate the efficacy of different cow's milk APT preparations to predict oral tolerance in children with previous non-IgE-mediated cow's milk allergy (CMA) diagnosis. METHODS Thirty-two patients non-IgE-mediated CMA diagnosed by oral food challenge (OFC) were enrolled to perform APT with three different cow's milk preparations (fresh, 2% in saline solution, 2% in petrolatum) and comparing with a new OFC after at least three months of diet exclusion. RESULTS Only six (18.7%) subjects presented positive OFC to cow's milk. No differences in gender, onset symptoms age, OFC age, Z-score, and exclusion period were found between positive and negative OFC patients. Preparations using fresh milk and powdered milk in petrolatum presented sensitivity equal to zero and specificity 92.3% and 96.1%. The preparation using powdered milk in saline solution showed sensitivity and specificity of 33.3% and 96.1%. Two patients presented typical IgE symptoms after OFC. CONCLUSION Cow's milk APT presented a low efficacy to predict tolerance in patients with previous non-IgE-mediated CMA and should not be used in clinical routine. The presence of typical IgE reactions after OFC hallmark the necessity of previous IgE-mediated investigation for this patient group.
Collapse
Affiliation(s)
- T A Gonzaga
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil
| | - F A Alves
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil
| | - M F A Cheik
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil
| | - C P de Barros
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil
| | - E R M A Rezende
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil
| | - G R S Segundo
- Food Allergy Research Group, Pediatrics Department, Universidade Federal de Uberlandia, Brazil.
| |
Collapse
|
40
|
Huang X, Tsilochristou O, Perna S, Hofmaier S, Cappella A, Bauer CP, Hoffman U, Forster J, Zepp F, Schuster A, D'Amelio R, Wahn U, Keil T, Lau S, Matricardi PM. Evolution of the IgE and IgG repertoire to a comprehensive array of allergen molecules in the first decade of life. Allergy 2018; 73:421-430. [PMID: 28791748 DOI: 10.1111/all.13269] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND In early childhood, the allergen-specific IgG repertoire is mainly directed to animal and vegetable food molecules and infrequently to airborne molecules. It is unknown whether this early pattern is maintained throughout childhood. OBJECTIVE To investigate the evolution of IgG and IgE responses to a broad panel of allergenic molecules from birth to age 10 years. METHODS We examined the sera collected between birth and age 10 years from participants in the German Multicentre Allergy Study, a birth cohort born in 1990. The IgE (cutoff ≥0.30 ISU) and IgG (cutoff ≥0.10 ISU) responses to 35 genuine allergenic molecules were measured with a multiplex microarray approach (ImmunoCAP ISAC™). RESULTS IgE responses were mostly directed against a restricted group of airborne molecules, with a sequence and prevalence hierarchy (Phl p 1> Bet v 1> Fel d 1> Phl p 5> Der p 2> Der p 1) largely maintained over time. Conversely, the IgG repertoire was much broader, starting with animal foodborne, then spreading to vegetable foodborne and finally to airborne molecules. A strong and persistent IgG response to a given airborne molecule almost invariably preceded or accompanied an IgE response to that molecule. CONCLUSIONS The evolution of IgG and IgE responses throughout childhood differs widely at population level. IgG responses are mostly directed to animal food allergens, while IgE responses are dominated by airborne allergens. However, a strong IgG response almost invariably precedes or accompanies the appearance of IgE to the same molecule in specifically sensitized subjects.
Collapse
Affiliation(s)
- X. Huang
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
- Department of Pediatrics; Shengzhou People's Hospital; Shengzhou China
| | - O. Tsilochristou
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| | - S. Perna
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| | - S. Hofmaier
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| | - A. Cappella
- Department of Clinical and Molecular Medicine; S. Andrea University Hospital; Sapienza University of Rome; Rome Italy
| | - C.-P. Bauer
- Department of Pediatrics; Technical University of Munich; Munich Germany
| | - U. Hoffman
- Department of Pediatrics; Technical University of Munich; Munich Germany
| | - J. Forster
- Department of Pediatrics St. Hedwig; St. Josefs Hospital; Freiburg Germany
| | - F. Zepp
- Department of Pediatrics and Adolescent Medicine; University Medicine Mainz; Mainz Germany
| | - A. Schuster
- Department of Pediatrics; Heinrich-Heine-University; Düsseldorf Germany
| | - R. D'Amelio
- Department of Clinical and Molecular Medicine; S. Andrea University Hospital; Sapienza University of Rome; Rome Italy
| | - U. Wahn
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| | - T. Keil
- Institute for Social Medicine, Epidemiology and Health Economics; Charité-Universitätsmedizin Berlin; Berlin Germany
- Institute of Clinical Epidemiology and Biometry; University of Würzburg; Würzburg Germany
| | - S. Lau
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| | - P. M. Matricardi
- Department of Pediatrics, Pneumology & Immunology; Charité-Universitätsmedizin Berlin; Berlin Germany
| |
Collapse
|
41
|
Jensen-Jarolim E, Bax HJ, Bianchini R, Crescioli S, Daniels-Wells TR, Dombrowicz D, Fiebiger E, Gould HJ, Irshad S, Janda J, Josephs DH, Levi-Schaffer F, O'Mahony L, Pellizzari G, Penichet ML, Redegeld F, Roth-Walter F, Singer J, Untersmayr E, Vangelista L, Karagiannis SN. AllergoOncology: Opposite outcomes of immune tolerance in allergy and cancer. Allergy 2018; 73:328-340. [PMID: 28921585 PMCID: PMC6038916 DOI: 10.1111/all.13311] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2017] [Indexed: 12/11/2022]
Abstract
While desired for the cure of allergy, regulatory immune cell subsets and nonclassical Th2-biased inflammatory mediators in the tumour microenvironment can contribute to immune suppression and escape of tumours from immunological detection and clearance. A key aim in the cancer field is therefore to design interventions that can break immunological tolerance and halt cancer progression, whereas on the contrary allergen immunotherapy exactly aims to induce tolerance. In this position paper, we review insights on immune tolerance derived from allergy and from cancer inflammation, focusing on what is known about the roles of key immune cells and mediators. We propose that research in the field of AllergoOncology that aims to delineate these immunological mechanisms with juxtaposed clinical consequences in allergy and cancer may point to novel avenues for therapeutic interventions that stand to benefit both disciplines.
Collapse
Affiliation(s)
- E Jensen-Jarolim
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - H J Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - R Bianchini
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
| | - S Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - T R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - D Dombrowicz
- INSERM, CHU Lille, European Genomic Institute of Diabetes, Institut Pasteur de Lille, U1011 - Recepteurs Nucleaires, Maladies Cardiovasculaires et Diabete, Universite de Lille, Lille, France
| | - E Fiebiger
- Division of Gastroenterology, Hepatology and Nutrition Research, Department Medicine Research, Childrens' University Hospital Boston, Boston, MA, USA
| | - H J Gould
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - S Irshad
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - J Janda
- Faculty of Science, Charles University, Prague, Czech Republic
| | - D H Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - F Levi-Schaffer
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - L O'Mahony
- Molecular Immunology, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - G Pellizzari
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - M L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, CA, USA
| | - F Redegeld
- Faculty of Science, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - F Roth-Walter
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - J Singer
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - E Untersmayr
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - L Vangelista
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - S N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| |
Collapse
|
42
|
Mitthamsiri W, Pradubpongsa P, Sangasapaviliya A, Boonpiyathad T. Decreased CRTH2 Expression and Response to Allergen Re-stimulation on Innate Lymphoid Cells in Patients With Allergen-Specific Immunotherapy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:662-674. [PMID: 30306748 PMCID: PMC6182198 DOI: 10.4168/aair.2018.10.6.662] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/02/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Purpose Group 2 innate lymphoid cells (ILC2s) have been implicated in the pathogenesis of allergic disease. However, the effect of allergen-specific immunotherapy (AIT) on ILCs remains to be clarified. The aim of this study was to evaluate the levels of ILC subsets in allergic rhinitis (AR) patients in response to house dust mite (HDM)-specific immunotherapy. Methods We enrolled 37 AR patients undergoing AIT (16 responders and 11 non-responders) for 2 years, 35 HDM AR patients and 28 healthy subjects. Peripheral blood mononuclear cells (PBMCs) were analyzed by flow cytometry to identify ILC subsets. Stimulation of ILC2s with recombinant allergen-specific protein was used to determine ILC2's activation (CD69 expression). Results Responder AIT patients and healthy subjects had a decreased frequency of circulating ILC2s compared to non-responder AIT and AR patients. Conversely, ILC1s from responder AIT patients and healthy subjects showed increased frequency compared to non-responder AIT and AR patients. The frequency of ILC3s natural cytotoxicity receptor (NCR)+ and NCR− in responder AIT patients was significantly lower compared to AR patients and healthy subjects. The ILC1: ILC2 proportion in responder AIT patients was similar to that of healthy subjects. PBMCs from patients who were responders to AIT had a significantly lower expression of the activation marker CD69 on ILC2s in response to allergen re-stimulation compared to AR patients, but no difference compared to non-responder AIT patients and healthy subjects. Conclusions We propose that AIT might affect ILC responses. The activation of ILC2s was reduced in AR patients treated with AIT. Our results indicate that a relative ILC1/ILC2 skewed response is a possible key to successful AIT.
Collapse
Affiliation(s)
- Wat Mitthamsiri
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Panitan Pradubpongsa
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Atik Sangasapaviliya
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Tadech Boonpiyathad
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand.
| |
Collapse
|
43
|
Feuille E, Nowak-Wegrzyn A. Allergen-Specific Immunotherapies for Food Allergy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:189-206. [PMID: 29676066 PMCID: PMC5911438 DOI: 10.4168/aair.2018.10.3.189] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/27/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
With rising prevalence of food allergy (FA), allergen-specific immunotherapy (AIT) for FA has become an active area of research in recent years. In AIT, incrementally increasing doses of inciting allergen are given with the goal to increase tolerance, initially through desensitization, which relies on regular exposure to allergen. With prolonged therapy in some subjects, AIT may induce sustained unresponsiveness, in which tolerance is retained after a period of allergen avoidance. Methods of AIT currently under study in humans include oral, sublingual, epicutaneous, and subcutaneous delivery of modified allergenic protein, as well as via DNA-based vaccines encoding allergen with lysosomal-associated membrane protein I. The balance of safety and efficacy varies by type of AIT, as well as by targeted allergen. Age, degree of sensitization, and other comorbidities may affect this balance within an individual patient. More recently, AIT with modified proteins or combined with immunomodulatory therapies has shown promise in making AIT safer and/or more effective. Though methods of AIT are neither currently advised by experts (oral immunotherapy [OIT]) nor widely available, AIT is likely to become a part of recommended management of FA in the coming years. Here, we review and compare methods of AIT currently under study in humans to prepare the practitioner for an exciting new phase in the care of food allergic patients in which improved tolerance to inciting foods will be a real possibility.
Collapse
Affiliation(s)
- Elizabeth Feuille
- Division of Pediatric Pulmonology, Allergy, and Immunology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Anna Nowak-Wegrzyn
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
44
|
Ahluwalia B, Magnusson MK, Öhman L. Mucosal immune system of the gastrointestinal tract: maintaining balance between the good and the bad. Scand J Gastroenterol 2017; 52:1185-1193. [PMID: 28697651 DOI: 10.1080/00365521.2017.1349173] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gastrointestinal tract (GI tract) is a unique organ inhabited by a range of commensal microbes, while also being exposed to an overwhelming load of antigens in the form of dietary antigens on a daily basis. The GI tract has dual roles in the body, in that it performs digestion and uptake of nutrients while also carrying out the complex and important task of maintaining immune homeostasis, i.e., keeping the balance between the good and the bad. It is equally important that we protect ourselves from reacting against the good, meaning that we stay tolerant to harmless food, commensal bacteria and self-antigens, as well as react with force against the bad, meaning induction of immune responses against harmful microorganisms. This complex task is achieved through the presence of a highly efficient mucosal barrier and a specialized multifaceted immune system, made up of a large population of scattered immune cells and organized lymphoid tissues termed the gut-associated lymphoid tissue (GALT). This review provides an overview of the primary components of the human mucosal immune system and how the immune responses in the GI tract are coordinated and induced.
Collapse
Affiliation(s)
- Bani Ahluwalia
- a Department of Microbiology and Immunology , Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden.,b Research Unit , Calmino Group AB , Gothenburg , Sweden
| | - Maria K Magnusson
- a Department of Microbiology and Immunology , Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Lena Öhman
- a Department of Microbiology and Immunology , Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden.,c Department of Internal Medicine and Clinical Nutrition , Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
45
|
Abstract
The gastrointestinal tract has an abundant mucosal immune system to develop and maintain oral tolerance. The oral route of administration takes advantage of the unique set of immune cells and pathways involved in the induction of oral tolerance. Food allergy results from a loss of oral tolerance toward ingested antigens. Oral immunotherapy is thought to initiate desensitization through interaction of an allergen with mucosal dendritic cells that initiate downstream immune system modulation through regulatory T cells and effector T cells.
Collapse
Affiliation(s)
- Erik Wambre
- Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA.
| | - David Jeong
- Virginia Mason Medical Center, 1201 Terry Avenue, Seattle, WA 98101, USA
| |
Collapse
|
46
|
Kostadinova AI, Pablos-Tanarro A, Diks MAP, van Esch BCAM, Garssen J, Knippels LMJ, Willemsen LEM. Dietary Intervention with β-Lactoglobulin-Derived Peptides and a Specific Mixture of Fructo-Oligosaccharides and Bifidobacterium breve M-16V Facilitates the Prevention of Whey-Induced Allergy in Mice by Supporting a Tolerance-Prone Immune Environment. Front Immunol 2017; 8:1303. [PMID: 29123515 PMCID: PMC5662887 DOI: 10.3389/fimmu.2017.01303] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/27/2017] [Indexed: 12/27/2022] Open
Abstract
Cow's milk allergy (CMA) prevails in infants and brings increased risk of developing other allergic diseases. Oral administration of specific β-lactoglobulin (BLG)-derived peptides (PepMix) and a specific blend of short- and long-chain fructo-oligosaccharides and Bifidobacterium breve M-16V (FF/Bb) was found to partially prevent CMA development in mice. In this study, we aimed to expand the knowledge on the preventive potential and the underlying mechanisms of this approach. Three-week-old female C3H/HeOuJ mice were orally exposed to PepMix±FF/Bb prior to a 5-week oral sensitization with whole whey and cholera toxin as an adjuvant. The acute allergic skin response was determined after an intradermal challenge with whole whey protein. Following an oral challenge with whey, regulatory T cells (Tregs) in the small intestine lamina propria (SI-LP) and mRNA expression of immune markers in the Peyer's patches (PP) were investigated. The early impact of PepMix and FF/Bb interventions on the immune system during the oral tolerance (OT) induction phase was investigated after the last OT administration. Pre-exposing mice to PepMix+FF/Bb partially prevented the acute allergic skin response compared to PBS and increased Tregs and activated T cells in the SI-LP compared to sham-sensitized mice. It also increased the mRNA expression of Tbet over GATA3 in the PP of whey-sensitized mice. Directly upon the 6-day OT phase, FF/Bb intervention enhanced cecal content levels of propionic and butyric acid in PepMix-fed mice and the former was positively correlated with Foxp3+ cell numbers in the colon. In the PP of PepMix+FF/Bb-exposed mice, IL-22 mRNA expression increased and IL-10 followed the same tendency, while the Foxp3 expression was increased over GATA3 and RorγT. In the colon, the Tbet mRNA expression increased over GATA3, while IL-22 decreased. In addition, the Foxp3+/GATA3+ and regulatory/effector T cell ratios in the mesenteric lymph nodes and the CD11b+/CD11b- conventional dendritic cells ratio in the SI-LP were increased. In conclusion, the FF/Bb diet facilitates the capacity of the specific BLG-peptides to partially prevent the allergic response after sensitization to whole whey protein, possibly by creating a tolerance-prone environment during the OT phase. Such a dietary intervention might contribute to tailoring successful strategies for CMA prevention.
Collapse
Affiliation(s)
- Atanaska I Kostadinova
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Immunology, Nutricia Research, Utrecht, Netherlands
| | - Alba Pablos-Tanarro
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - Mara A P Diks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Betty C A M van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Immunology, Nutricia Research, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Immunology, Nutricia Research, Utrecht, Netherlands
| | - Léon M J Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Immunology, Nutricia Research, Utrecht, Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
47
|
Allergens with Protease Activity from House Dust Mites. Int J Mol Sci 2017; 18:ijms18071368. [PMID: 28653989 PMCID: PMC5535861 DOI: 10.3390/ijms18071368] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 11/29/2022] Open
Abstract
Globally, house dust mites (HDM) are one of the main sources of allergens causing Type I allergy, which has a high risk of progressing into a severe disabling disease manifestation such as allergic asthma. The strong protease activities of a number of these allergens are thought to be involved in several steps of the pathophysiology of this allergic disease. It has been a common notion that protease activity may be one of the properties that confers allergenicity to proteins. In this review we summarize and discuss the roles of the different HDM proteases in the development of Type I allergy.
Collapse
|
48
|
Freeland DMH, Manohar M, Andorf S, Hobson BD, Zhang W, Nadeau KC. Oral immunotherapy for food allergy. Semin Immunol 2017; 30:36-44. [PMID: 28865877 PMCID: PMC5776738 DOI: 10.1016/j.smim.2017.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 01/20/2023]
Abstract
Food allergy is a pathological, potentially deadly cascade of immune responses to molecules or molecular fragments that are normally innocuous when encountered in foods, such as milk, egg, or peanut. As the incidence and prevalence of food allergy rise, the standard of care is poised to advance beyond food allergen avoidance coupled with injectable epinephrine treatment of allergen-induced systemic reactions. Recent studies provide evidence that oral immunotherapy may effectively redirect the atopic immune responses of food allergy patients as they ingest small but gradually increasing allergen doses over many months, eliciting safer immune responses to these antigens. Research into the molecular and cellular bases of pathological and therapeutic immune responses, and into the possibilities for their safe and effective modulation, is generating tremendous interest in basic and clinical immunology. We synthesize developments, innovations, and key challenges in our understanding of the immune mechanisms associated with atopy and oral immunotherapy for food allergy.
Collapse
Affiliation(s)
- Deborah M Hussey Freeland
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Stanford, CA 94305, USA; Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Monali Manohar
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Stanford, CA 94305, USA; Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sandra Andorf
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Stanford, CA 94305, USA; Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Benjamin D Hobson
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wenming Zhang
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Stanford, CA 94305, USA; Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Stanford, CA 94305, USA; Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA; Division of Allergy, Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|