1
|
Wu Y, Chen B, Wu H, Gao J, Meng X, Chen H. How maternal factors shape the immune system of breastfed infants to alleviate food allergy: A systematic and updated review. Immunology 2025; 174:1-16. [PMID: 39344356 DOI: 10.1111/imm.13864] [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: 10/31/2023] [Accepted: 09/06/2024] [Indexed: 10/01/2024] Open
Abstract
What infants eat early in life may shape the immune system and have long-standing consequences on the health of the host during later life. In the early months post-birth, breast milk serves as the exclusive and optimal nourishment for infants, facilitating crucial molecular exchanges between mother and infant. Recent advances have uncovered that some maternal factors influence breastfed infant outcomes, including the risk of food allergy (FA). To date, accumulated data show that breastfed infants have a lower risk of FA. However, the issue remains disputed, some reported preventive allergy effects, while others did not confirm such effects, or if identified, protective effects were limited to early childhood. The disputed outcomes may be attributed to the maternal status, as it determines the compounds of the breast milk that breastfed infants are exposed to. In this review, we first detail the compounds in breast milk and their roles in infant FA. Then, we present maternal factors resulting in alterations in breast milk compounds, such as maternal health status, maternal diet intake, and maternal food allergen intake, which subsequently impact FA in breastfed infants. Finally, we analyze how these compounds in breast milk alleviated the infant FA by mother-to-infant transmission. Altogether, the mechanisms are primarily linked to the synergetic and direct effects of compounds in breast milk, via promoting the colonization of gut microbiota and the development of the immune system in infants.
Collapse
Affiliation(s)
- Yuhong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Bihua Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Huan Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Verhasselt V, Marchant A, Kollmann TR. Per Os to Protection - Targeting the Oral Route to Enhance Immune-mediated Protection from Disease of the Human Newborn. J Mol Biol 2024; 436:168718. [PMID: 39094783 DOI: 10.1016/j.jmb.2024.168718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Affiliation(s)
- Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, School of Medicine, University of Western Australia, Perth, WA, Australia; Immunology and Breastfeeding Team, Telethon Kids Institute, Perth, WA, Australia
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Tobias R Kollmann
- Dalhousie University, Department of Microbiology & Immunology, Pediatric Infectious Diseases, Canada.
| |
Collapse
|
3
|
Treppiccione L, Maurano F, Luongo D, Rossi M. Intragastric administration of transamidated gliadin interferes with the systemic and intestinal immune responses to wheat gliadin in DQ8 transgenic mice. Cytokine 2024; 182:156722. [PMID: 39116536 DOI: 10.1016/j.cyto.2024.156722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
We have previously shown the ability of transamidated gluten (spf) to modulate both innate and adaptive intestinal immunity elicited by wheat gliadin in HLA-DQ8 transgenic mice (DQ8 mice), a model of gluten sensitivity. Herein, we evaluated the influence of spf when administered intragastrically on the immune response to native gliadin in DQ8 mice. To address the issue, we analysed three regimens of antigen administration: before immunisation (pre-treatment), during immunisation (co-treatment) and through breast milk during the lactating phase (suckling treatment). Mice were immunised mucosally by intranasal delivery of digested wheat gliadin along with cholera toxin in multiple doses. After sacrifice, isolated spleen and mesenteric lymph node (MLN) cells were challenged in vitro and the cytokine profile of culture supernatants assessed by ELISA and multiparametric assay. We found that only pre-treatment with spf was effective in down-regulating the gliadin-specific IFN-γ response and only in spleen cells. Interestingly, spf pre-treatment also induced systemic IL-6, IL-17A and TNF-α. By contrast, we found that spf pre-treatment upregulated INF-γ in MLN but also significantly decreased IL-2. In conclusion, our data provide evidence that the preventive intragastric administration of transamidated gluten is able to interfere with the classical cytokine profile induced by gliadin via mucosal immunisation in a transgenic model expressing one of the HLA molecules associated with coeliac disease.
Collapse
Affiliation(s)
| | | | - Diomira Luongo
- Institute of Food Sciences, CNR, via Roma 64, 83100 Avellino, Italy
| | - Mauro Rossi
- Institute of Food Sciences, CNR, via Roma 64, 83100 Avellino, Italy
| |
Collapse
|
4
|
Miranda-Waldetario MC, Curotto de Lafaille MA. Oral tolerance to dietary antigens and Foxp3 + regulatory T cells. Immunol Rev 2024; 326:8-16. [PMID: 39054615 PMCID: PMC11436310 DOI: 10.1111/imr.13370] [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] [Indexed: 07/27/2024]
Abstract
Immune tolerance to foods develops in the intestine upon food ingestion and is essential to prevent IgE-mediated food allergy and gut inflammation. In homeostasis, the intestine is a tolerogenic environment that favors the formation of food-specific Foxp3+ regulatory T cells. A tolerogenic intestinal environment depends on colonization by diverse microbiota and exposure to solid foods at a critical period in early life. These early immune responses lead to the induction of antigen-specific Foxp3+ regulatory T cells in draining mesenteric lymph nodes. These peripherally induced regulatory cells circulate and seed the lamina propria of the gut, exerting suppressive function systemically and locally in the intestine. Successful establishment of a tolerogenic intestinal environment in early life sets the stage for oral tolerance to new antigens in adult life.
Collapse
Affiliation(s)
- Mariana C.G. Miranda-Waldetario
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A. Curotto de Lafaille
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Verhasselt V. A newborn's perspective on immune responses to food. Immunol Rev 2024; 326:117-129. [PMID: 39162048 DOI: 10.1111/imr.13376] [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] [Indexed: 08/21/2024]
Abstract
In this review, we will highlight infants' immune responses to food, emphasizing the unique aspects of early-life immunity and the critical role of breast milk as a food dedicated to infants. Infants are susceptible to inflammatory responses rather than immune tolerance at the mucosal and skin barriers, necessitating strategies to promote oral tolerance that consider this susceptibility. Breast milk provides nutrients for growth and cell metabolism, including immune cells. The content of breast milk, influenced by maternal genetics and environmental exposures, prepares the infant's immune system for the outside world, including solid foods. To do this, breast milk promotes immune system development through antigen-specific and non-antigen-specific immune education by exposing the newborn to food and respiratory allergens and acting on three key targets for food allergy prevention: the gut microbiota, epithelial cells, and immune cells. Building knowledge of how the maternal exposome and human milk composition influence offspring's healthy immune development will lead to recommendations that meet the specific needs of the developing immune system and increase the chances of promoting an appropriate immune response to food in the long term.
Collapse
Affiliation(s)
- Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Immunology and Breastfeeding team, Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
6
|
Zou M, Pezoldt J, Mohr J, Philipsen L, Leufgen A, Cerovic V, Wiechers C, Pils M, Ortiz D, Hao L, Yang J, Beckstette M, Dupont A, Hornef M, Dersch P, Strowig T, Müller AJ, Raila J, Huehn J. Early-life vitamin A treatment rescues neonatal infection-induced durably impaired tolerogenic properties of celiac lymph nodes. Cell Rep 2024; 43:114153. [PMID: 38687643 DOI: 10.1016/j.celrep.2024.114153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/23/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.
Collapse
Affiliation(s)
- Mangge Zou
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Joern Pezoldt
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Juliane Mohr
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Multi-Parametric Bioimaging and Cytometry (MPBIC) Platform, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Andrea Leufgen
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Carolin Wiechers
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Marina Pils
- Mouse Pathology Platform, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Diego Ortiz
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Lianxu Hao
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Juhao Yang
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Aline Dupont
- Institute of Medical Microbiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Petra Dersch
- Institute for Infectiology, University of Münster, 48149 Münster, Germany; German Center for Infection Research (DZIF), Associated Site University of Münster, 48149 Münster, Germany
| | - Till Strowig
- Department Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Multi-Parametric Bioimaging and Cytometry (MPBIC) Platform, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Intravital Microscopy in Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jens Raila
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
7
|
Bai T, Wang Z, Shao H, Zhang X, Lorenz A, Meng X, Wu Y, Chen H, Li X. Novel Perspective on the Regulation of Offspring Food Allergy by Maternal Diet and Nutrients. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10679-10691. [PMID: 38695770 DOI: 10.1021/acs.jafc.3c09108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
There has been a dramatic surge in the prevalence of food allergy (FA) that cannot be explained solely by genetics, identifying mechanisms of sensitization that are driven by environmental factors has become increasingly important. Diet, gut microbiota, and their metabolites have been shown to play an important role in the development of FA. In this review, we discuss the latest epidemiological evidence on the impact of two major dietary patterns and key nutrients in early life on the risk of offspring developing FA. The Western diet typically includes high sugar and high fat, which may affect the immune system of offspring and increase susceptibility to FA. In contrast, the Mediterranean diet is rich in fiber, which may reduce the risk of FA in offspring. Furthermore, we explore the potential mechanisms by which maternal dietary nutrients during a window of opportunity (pregnancy, birth, and lactation) influences the susceptibility of offspring to FA through multi-interface crosstalk. Finally, we discuss the limitations and gaps in the available evidence regarding the relationship between maternal dietary nutrients and the risk of FA in offspring. This review provides novel perspective on the regulation of offspring FA by maternal diet and nutrients.
Collapse
Affiliation(s)
- Tianliang Bai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Zhongliang Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Huming Shao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Agla Lorenz
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg 5020, Austria
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi 330047, China
| |
Collapse
|
8
|
Roth-Walter F, Berni Canani R, O'Mahony L, Peroni D, Sokolowska M, Vassilopoulou E, Venter C. Nutrition in chronic inflammatory conditions: Bypassing the mucosal block for micronutrients. Allergy 2024; 79:353-383. [PMID: 38084827 DOI: 10.1111/all.15972] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Nutritional Immunity is one of the most ancient innate immune responses, during which the body can restrict nutrients availability to pathogens and restricts their uptake by the gut mucosa (mucosal block). Though this can be a beneficial strategy during infection, it also is associated with non-communicable diseases-where the pathogen is missing; leading to increased morbidity and mortality as micronutritional uptake and distribution in the body is hindered. Here, we discuss the acute immune response in respect to nutrients, the opposing nutritional demands of regulatory and inflammatory cells and particularly focus on some nutrients linked with inflammation such as iron, vitamins A, Bs, C, and other antioxidants. We propose that while the absorption of certain micronutrients is hindered during inflammation, the dietary lymph path remains available. As such, several clinical trials investigated the role of the lymphatic system during protein absorption, following a ketogenic diet and an increased intake of antioxidants, vitamins, and minerals, in reducing inflammation and ameliorating disease.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Liam O'Mahony
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Diego Peroni
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Vassilopoulou
- Pediatric Area, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carina Venter
- Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
9
|
López-Fandiño R, Molina E, Lozano-Ojalvo D. Intestinal factors promoting the development of RORγt + cells and oral tolerance. Front Immunol 2023; 14:1294292. [PMID: 37936708 PMCID: PMC10626553 DOI: 10.3389/fimmu.2023.1294292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
The gastrointestinal tract has to harmonize the two seemingly opposite functions of fulfilling nutritional needs and avoiding the entry of pathogens, toxins and agents that can cause physical damage. This balance requires a constant adjustment of absorptive and defending functions by sensing environmental changes or noxious substances and initiating adaptive or protective mechanisms against them through a complex network of receptors integrated with the central nervous system that communicate with cells of the innate and adaptive immune system. Effective homeostatic processes at barrier sites take the responsibility for oral tolerance, which protects from adverse reactions to food that cause allergic diseases. During a very specific time interval in early life, the establishment of a stable microbiota in the large intestine is sufficient to prevent pathological events in adulthood towards a much larger bacterial community and provide tolerance towards diverse food antigens encountered later in life. The beneficial effects of the microbiome are mainly exerted by innate and adaptive cells that express the transcription factor RORγt, in whose generation, mediated by different bacterial metabolites, retinoic acid signalling plays a predominant role. In addition, recent investigations indicate that food antigens also contribute, analogously to microbial-derived signals, to educating innate immune cells and instructing the development and function of RORγt+ cells in the small intestine, complementing and expanding the tolerogenic effect of the microbiome in the colon. This review addresses the mechanisms through which microbiota-produced metabolites and dietary antigens maintain intestinal homeostasis, highlighting the complementarity and redundancy between their functions.
Collapse
Affiliation(s)
- Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL), CSIC-UAM, Madrid, Spain
| | | | | |
Collapse
|
10
|
Peroni DG, Hufnagl K, Comberiati P, Roth-Walter F. Lack of iron, zinc, and vitamins as a contributor to the etiology of atopic diseases. Front Nutr 2023; 9:1032481. [PMID: 36698466 PMCID: PMC9869175 DOI: 10.3389/fnut.2022.1032481] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023] Open
Abstract
Micronutritional deficiencies are common in atopic children suffering from atopic dermatitis, food allergy, rhinitis, and asthma. A lack of iron, in particular, may impact immune activation with prolonged deficiencies of iron, zinc, vitamin A, and vitamin D associated with a Th2 signature, maturation of macrophages and dendritic cells (DCs), and the generation of IgE antibodies. In contrast, the sufficiency of these micronutrients establishes immune resilience, promotion of regulatory cells, and tolerance induction. As micronutritional deficiencies mimic an infection, the body's innate response is to limit access to these nutrients and also impede their dietary uptake. Here, we summarize our current understanding of the physiological function of iron, zinc, and vitamins A and D in relation to immune cells and the clinical consequences of deficiencies in these important nutrients, especially in the perinatal period. Improved dietary uptake of iron is achieved by vitamin C, vitamin A, and whey compounds, whereas zinc bioavailability improves through citrates and proteins. The addition of oil is essential for the dietary uptake of beta-carotene and vitamin D. As for vitamin D, the major source comes via sun exposure and only a small amount is consumed via diet, which should be factored into clinical nutritional studies. We summarize the prevalence of micronutritional deficiencies of iron, zinc, and vitamins in the pediatric population as well as nutritional intervention studies on atopic diseases with whole food, food components, and micronutrients. Dietary uptake via the lymphatic route seems promising and is associated with a lower atopy risk and symptom amelioration. This review provides useful information for clinical studies and concludes/emphasizes that a healthy, varied diet containing dairy products, fish, nuts, fruits, and vegetables as well as supplementing foods or supplementation with micronutrients as needed is essential to combat the atopic march.
Collapse
Affiliation(s)
- Diego G. Peroni
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Karin Hufnagl
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria,Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Pasquale Comberiati
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria,Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria,*Correspondence: Franziska Roth-Walter, ;
| |
Collapse
|
11
|
Shi J, Wang Y, Cheng L, Wang J, Raghavan V. Gut microbiome modulation by probiotics, prebiotics, synbiotics and postbiotics: a novel strategy in food allergy prevention and treatment. Crit Rev Food Sci Nutr 2022; 64:5984-6000. [PMID: 36576159 DOI: 10.1080/10408398.2022.2160962] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Food allergy has caused lots of global public health issues, particularly in developed countries. Presently, gut microbiota has been widely studied on allergy, while the role of dysbiosis in food allergy remains unknown. Scientists found that changes in gut microbial compositions and functions are strongly associated with a dramatic increase in the prevalence of food allergy. Altering microbial composition is crucial in modulating food antigens' immunogenicity. Thus, the potential roles of probiotics, prebiotics, synbiotics, and postbiotics in affecting gut bacteria communities and the immune system, as innovative strategies against food allergy, begins to attract high attention of scientists. This review briefly summarized the mechanisms of food allergy and discussed the role of the gut microbiota and the use of probiotics, prebiotics, synbiotics, and postbiotics as novel therapies for the prevention and treatment of food allergy. The perspective studies on the development of novel immunotherapy in food allergy were also described. A better understanding of these mechanisms will facilitate the development of preventive and therapeutic strategies for food allergy.
Collapse
Affiliation(s)
- Jialu Shi
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Youfa Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Lei Cheng
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Vijaya Raghavan
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| |
Collapse
|
12
|
Friesen L, Kostlan R, Liu Q, Yu H, Zhu J, Lukacs N, Kim CH. Cutting Edge: The Expression of Transcription Inhibitor GFI1 Is Induced by Retinoic Acid to Rein in Th9 Polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1237-1242. [PMID: 36165199 PMCID: PMC9522314 DOI: 10.4049/jimmunol.2200328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/07/2022] [Indexed: 11/07/2022]
Abstract
IL-9, produced mainly by specialized T cells, mast cells, and group 2 innate lymphoid cells, regulates immune responses, including anti-helminth and allergic responses. Polarization of naive CD4 T cells into IL-9-producing T cells (Th9s) is induced by IL-4 and TGF-β1 or IL-1β. In this article, we report that the transcription factor growth factor-independent 1 transcriptional repressor (GFI1) plays a negative role in mouse Th9 polarization. Moreover, the expression of GFI1 is controlled by liganded RARα, allowing GFI1 to mediate the negative effect of retinoic acid on IL-9 expression. The Gfi1 gene has multiple RARα binding sites in the promoter region for recruiting nuclear coactivator steroid receptor coactivator-3 and p300 for histone epigenetic modifications in a retinoic acid-dependent manner. Retinoic acid-induced GFI1 binds the Il9 gene and suppresses its expression. Thus, GFI1 is a novel negative regulator of Il9 gene expression. The negative GFI1 pathway for IL-9 regulation provides a potential control point for Th9 activity.
Collapse
Affiliation(s)
- Leon Friesen
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Raymond Kostlan
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Qingyang Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Hao Yu
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IL
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | - Nicholas Lukacs
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI;
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
13
|
Danielewicz H. Breastfeeding and Allergy Effect Modified by Genetic, Environmental, Dietary, and Immunological Factors. Nutrients 2022; 14:nu14153011. [PMID: 35893863 PMCID: PMC9331378 DOI: 10.3390/nu14153011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022] Open
Abstract
Breastfeeding (BF) is the most natural mode of nutrition. Its beneficial effect has been revealed in terms of both the neonatal period and those of lifelong effects. However, as for protection against allergy, there is not enough data. In the current narrative review, the literature within the last five years from clinical trials and population-based studies on breastfeeding and allergy from different aspects was explored. The aim of this review was to explain how different factors could contribute to the overall effect of BF. Special consideration was given to accompanying exposure to cow milk, supplement use, the introduction of solid foods, microbiota changes, and the epigenetic function of BF. Those factors seem to be modifying the impact of BF. We also identified studies regarding BF in atopic mothers, with SCFA as a main player explaining differences according to this status. Conclusion: Based on the population-based studies, breastfeeding could be protective against some allergic phenotypes, but the results differ within different study groups. According to the new research in that matter, the effect of BF could be modified by different genetic (HMO composition), environmental (cesarean section, allergen exposure), dietary (SCFA, introduction of solid food), and immunologic factors (IgG, IgE), thus partially explaining the variance.
Collapse
Affiliation(s)
- Hanna Danielewicz
- 1st Clinical Department of Pediatrics, Allergology and Cardiology, Wroclaw Medical University, ul. Chałubińskiego 2a, 50-368 Wrocław, Poland
| |
Collapse
|
14
|
Schocker F, Jappe U. Breastfeeding: Maternally Transferred Allergens in Breast Milk: Protective or Sensitizing? Mol Nutr Food Res 2022; 66:e2200066. [PMID: 35619210 DOI: 10.1002/mnfr.202200066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/01/2022] [Indexed: 11/11/2022]
Abstract
According to a thorough literature search, the following allergen sources have been associated with allergy symptoms in the exclusively breastfed child: hen's egg, cow's milk, peanut, trout. Subsequently, several studies use the advantage of molecular allergology and investigate the potential transfer of single allergens into breastmilk. This is shown for caseins, whey proteins, gliadin, ovalbumin, ovomucoid, the peanut allergens Ara h 2 and Ara h 6, as well as the inhalant allergens Der p 1 and Blo t 5. It is still a matter of debate whether or not food allergens transferred via breastfeeding to the baby promote allergic sensitization or induce tolerance and via which mechanisms they may shift the immune response to the one or other side. Noteworthy, some breastfed children are described to be sensitized to foods before being exposed to solid foods, and this exposure may have occurred through breastmilk. In the light of these findings the investigation of food allergens transferred from the mother's diet into breastmilk and their impact on sensitization or allergy prevention remains a current topic in research. This review describes breastmilk in its composition and provides data on the identification of food allergens therein including human and mouse studies.
Collapse
Affiliation(s)
- Frauke Schocker
- Division of Clinical and Molecular Allergology, Priority Research Area Asthma and Allergy, Research Center Borstel, Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research, Germany
| | - Uta Jappe
- Division of Clinical and Molecular Allergology, Priority Research Area Asthma and Allergy, Research Center Borstel, Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research, Germany.,Interdisciplinary Allergy Outpatient Clinic, Department of Pneumology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
15
|
Treppiccione L, Luongo D, Maurano F, Rossi M. Next generation strategies to recover immunological tolerance in celiac disease. Int Rev Immunol 2022; 42:237-245. [PMID: 35225129 DOI: 10.1080/08830185.2022.2044807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Celiac disease (CD) is an autoimmune disease that occurs in genetically predisposed individuals following the ingestion of gluten. Its prevalence is rising worldwide. A gluten-free (GF) diet is mandatory for the management of CD. However, several issues persist regarding the nutritional quality of GF products. Importantly, deep knowledge about the pathogenic mechanisms in CD highlights the central role of CD4+ T cell-mediated immunity in CD. Furthermore, intestinal T regulatory cells are functional in CD, but cytokines such as IL-15, produced under inflammatory conditions, hamper their activity. This paves the way for the development of immunomodulatory strategies to the GF diet. From this perspective, microbiological approaches were considered able to modulate the gluten-specific immune response. Interestingly, gliadin peptide-based immunotherapy to abolish the inflammatory CD4+T cell-mediated response has been explored in CD patients. Furthermore, different biotechnological approaches based on the use of chemically/enzymatically modified gluten molecules have been proved effective in different models of CD. However, the choice of the right age in infants to introduce the antigen and thus induce tolerance still remains an important issue to solve. Addressing all these points should help to design an effective intervention strategy for preventing CD.
Collapse
Affiliation(s)
| | | | | | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy
| |
Collapse
|
16
|
Hornef M, Pabst O, Annesi-Maesano I, Fleddermann M, von Mutius E, Schaubeck M, Fiocchi A. Allergic diseases in infancy II-oral tolerance and its failure. World Allergy Organ J 2021; 14:100586. [PMID: 34868451 PMCID: PMC8609161 DOI: 10.1016/j.waojou.2021.100586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022] Open
Abstract
Objective The early window of opportunity describes the timeframe after birth in which essential interactions of the immune system and the newly developing microbiota take place. The infant's immune system has to be reactive to invading pathogens and at the same time tolerant to dietary antigens. If the mechanisms of defense and tolerance induction are disturbed, the risk of infections or allergies is increased. Method This is a narrative review of the recently published information on the topic of neonatal intestinal development and mechanisms of oral tolerance and summarizes the discussions and conclusions from the 8th Human Milk Workshop. Results The early postnatal period sets the stage for life-long host-microbiome interaction. In this early phase, specific developmental mechanisms ensure physiologic interaction with the developing microbiota. Innate and adaptive immune cells interact in a concerted way to induce and uphold oral tolerance. Factors in human milk can support this induction of tolerance and simultaneously protect against infection and allergy development. Conclusion Understanding the developmental mechanisms in this early phase of immune system development is the first step to develop strategies of pathology prevention. As human milk protects the infant from infections, and aids to develop a tolerogenic immune response, further knowledge on the protective factors in human milk and their effect on the immune system is required.
Collapse
Affiliation(s)
- Mathias Hornef
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Pauwelsstr. 30, Aachen, 52074, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Pauwelsstr. 30, Aachen, 52074, Germany
| | - Isabella Annesi-Maesano
- Epidemiology of Allergic and Respiratory Diseases Department, IPLESP, French Institute of Health and Medical Research and Sorbonne University, Medical School Saint Antoine, 27 Rue Chaligny, Paris, 75012, France
| | - Manja Fleddermann
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Straße 7, Pfaffenhofen, 85276, Germany
| | - Erika von Mutius
- Dr. von Hauner Children's Hospital, University of Munich, Lindwurmstr. 4, Munich, 80337, Germany
| | - Monika Schaubeck
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Straße 7, Pfaffenhofen, 85276, Germany
| | - Alessandro Fiocchi
- Division of Allergy, Pediatric Hospital Bambino Gesú (IRCCS), Piazza di Sant'Onofrio 4, Rome, 00165, Italy
| |
Collapse
|
17
|
Krawiec M, Fisher HR, Du Toit G, Bahnson HT, Lack G. Overview of oral tolerance induction for prevention of food allergy-Where are we now? Allergy 2021; 76:2684-2698. [PMID: 33539570 DOI: 10.1111/all.14758] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 01/10/2023]
Abstract
Oral tolerance induction through early introduction of allergenic food has proven effective in randomized controlled trials. This new approach to weaning has been incorporated into many national and international infants' feeding guidelines. However, there are questions that require further discussion, such as, which foods should be introduced early, should the intervention be targeted to infants at high-risk or to the general population, and what is the ideal timing for early food introduction. This review examines the extent to which recent trials address these critical questions and highlights areas where further research is required.
Collapse
Affiliation(s)
- Marta Krawiec
- Department of Women and Children’s Health (Paediatric Allergy) School of Life Course Sciences Faculty of Life Sciences and Medicine King’s College London London UK
- Children’s Allergy Service Evelina London Guy’s and St Thomas’ Hospital London UK
| | - Helen R. Fisher
- Department of Women and Children’s Health (Paediatric Allergy) School of Life Course Sciences Faculty of Life Sciences and Medicine King’s College London London UK
- Children’s Allergy Service Evelina London Guy’s and St Thomas’ Hospital London UK
| | - George Du Toit
- Department of Women and Children’s Health (Paediatric Allergy) School of Life Course Sciences Faculty of Life Sciences and Medicine King’s College London London UK
- Children’s Allergy Service Evelina London Guy’s and St Thomas’ Hospital London UK
- Asthma UK Centre in Allergic Mechanisms of Asthma London UK
| | - Henry T. Bahnson
- Benaroya Research Institute and the Immune Tolerance Network Seattle USA
| | - Gideon Lack
- Department of Women and Children’s Health (Paediatric Allergy) School of Life Course Sciences Faculty of Life Sciences and Medicine King’s College London London UK
- Children’s Allergy Service Evelina London Guy’s and St Thomas’ Hospital London UK
- Peter Gorer Department of Immunobiology School of Immunology and Microbial Sciences King’s College London London UK
| |
Collapse
|
18
|
Pinheiro-Rosa N, Torres L, Oliveira MDA, Andrade-Oliveira MF, Guimarães MADF, Coelho MM, Alves JDL, Maioli TU, Faria AMC. Oral tolerance as antigen-specific immunotherapy. IMMUNOTHERAPY ADVANCES 2021; 1:ltab017. [PMID: 35919733 PMCID: PMC9327124 DOI: 10.1093/immadv/ltab017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Summary
Oral tolerance is a physiological phenomenon described more than a century ago as a suppressive immune response to antigens that gain access to the body by the oral route. It is a robust and long-lasting event with local and systemic effects in which the generation of mucosally induced regulatory T cells (iTreg) plays an essential role. The idea of using oral tolerance to inhibit autoimmune and allergic diseases by oral administration of target antigens was an important development that was successfully tested in 1980s. Since then, several studies have shown that feeding specific antigens can be used to prevent and control chronic inflammatory diseases in both animal models and clinically. Therefore, oral tolerance can be classified as an antigen-specific form of oral immunotherapy (OIT). In the light of novel findings on mechanisms, sites of induction and factors affecting oral tolerance, this review will focus on specific characteristics of oral tolerance induction and how they impact in its therapeutic application.
Collapse
Affiliation(s)
- Natália Pinheiro-Rosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lícia Torres
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mariana de Almeida Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcos Felipe Andrade-Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Andrade de Freitas Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monique Macedo Coelho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana de Lima Alves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana M Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
19
|
Macchiaverni P, Rekima A, van den Elsen L, Renz H, Verhasselt V. Allergen shedding in human milk: Could it be key for immune system education and allergy prevention? J Allergy Clin Immunol 2021; 148:679-688. [PMID: 34310930 DOI: 10.1016/j.jaci.2021.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 02/02/2023]
Abstract
In addition to being a source of nutrients for the developing newborn, human milk contains thousands of bioactive compounds, which influence infant health in the short-term as exemplified by its major benefits on infectious disease prevention. Many of the human milk compounds also have the required characteristics to instruct immune development and guide long-term health. Prebiotics, probiotics, and varied antimicrobial molecules all have the potential to shape the composition and function of the establishing gut microbiota, which is known to be a major determinant of immune function. Another and less explored way human milk can instruct long-term immunity is through antigen shedding. Here, we will review the evidence that antigens from maternal environment and more specifically from allergen sources are found in human milk. We will discuss data from rodent models and birth cohorts showing that allergen shedding in breast milk may influence long-term allergy risk. We will uncover the variables that may underlie heterogeneity in oral tolerance induction and allergy prevention in children breast-fed by allergen-exposed mothers. We will focus on the parameters that control antigen transfer to breast milk, on the unique biological characteristics of allergens in breast milk, and on the milk bioactive compounds that were found to influence immune response in offspring. We propose this understanding is fundamental to guide maternal interventions leading to lifelong allergen tolerance.
Collapse
Affiliation(s)
- Patricia Macchiaverni
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Akila Rekima
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Lieke van den Elsen
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany; Member of the Universities of Giessen and Marburg Lung Center (UGMLC), Marburg, Germany; German Center for Lung Research (DZL), Marburg, Germany; In Vivo Planetary Health, Worldwide Universities Network (WUN), West New York
| | - Valerie Verhasselt
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia; In Vivo Planetary Health, Worldwide Universities Network (WUN), West New York.
| |
Collapse
|
20
|
Wagner C, Torow N, Hornef MW, Lelouard H. Spatial and temporal key steps in early-life intestinal immune system development and education. FEBS J 2021; 289:4731-4757. [PMID: 34076962 DOI: 10.1111/febs.16047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Education of our intestinal immune system early in life strongly influences adult health. This education strongly relies on series of events that must occur in well-defined time windows. From initial colonization by maternal-derived microbiota during delivery to dietary changes from mother's milk to solid foods at weaning, these early-life events have indeed long-standing consequences on our immunity, facilitating tolerance to environmental exposures or, on the contrary, increasing the risk of developing noncommunicable diseases such as allergies, asthma, obesity, and inflammatory bowel diseases. In this review, we provide an outline of the recent advances in our understanding of these events and how they are mechanistically related to intestinal immunity development and education. First, we review the susceptibility of neonates to infections and inflammatory diseases, related to their immune system and microbiota changes. Then, we highlight the maternal factors involved in protection and education of the mucosal immune system of the offspring, the role of the microbiota, and the nature of neonatal immune system until weaning. We also present how the development of some immune responses is intertwined in temporal and spatial windows of opportunity. Finally, we discuss pending questions regarding the neonate particular immune status and the activation of the intestinal immune system at weaning.
Collapse
Affiliation(s)
- Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | | |
Collapse
|
21
|
van den Elsen LWJ, Verhasselt V. Human Milk Drives the Intimate Interplay Between Gut Immunity and Adipose Tissue for Healthy Growth. Front Immunol 2021; 12:645415. [PMID: 33912171 PMCID: PMC8071867 DOI: 10.3389/fimmu.2021.645415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
As the physiological food for the developing child, human milk is expected to be the diet that is best adapted for infant growth needs. There is also accumulating evidence that breastfeeding influences long-term metabolic outcomes. This review covers the potential mechanisms by which human milk could regulate healthy growth. We focus on how human milk may act on adipose tissue development and its metabolic homeostasis. We also explore how specific human milk components may influence the interplay between the gut microbiota, gut mucosa immunity and adipose tissue. A deeper understanding of these interactions may lead to new preventative and therapeutic strategies for both undernutrition and other metabolic diseases and deserves further exploration.
Collapse
Affiliation(s)
| | - Valerie Verhasselt
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
22
|
The relationship between umbilical cord blood vitamin A levels and late preterm infant morbidities: a prospective cohort study. Eur J Pediatr 2021; 180:791-797. [PMID: 32851492 DOI: 10.1007/s00431-020-03787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
The aim of this study is to explore the association between umbilical cord blood (UCB) vitamin A levels and late preterm infants morbidities. We conducted a prospective cohort study of 208 late-preterm infants(from 34 0/7 to 36 6/7 weeks gestational age) between January 1, 2014 and June 30, 2015. UCB specimens were collected shortly after birth, and vitamin A levels were determined by enzyme-linked immunosorbent assay. Prevalence of low UCB vitamin A level < 0.7 μmol/L was 37.5% in late preterm infants. In comparison to vaginal delivery, cesarean section was associated with UCB vitamin A level < 0.7 μmol/L (P < 0.001). Nevertheless, UCB vitamin A levels did not correlate with gestational age, birth weight, and gender. UCB vitamin A level < 0.7 μmol/L was not an independent risk factor for hospitalization, oxygen supplementation, hyperbilirubinemia, sepsis, and respiratory distress syndrome.Conclusions: Low umbilical cord blood vitamin A levels are common among late-preterm infants. Cesarean section delivery is associated with low umbilical cord blood vitamin A level. Low umbilical cord blood vitamin A levels at birth do not increase morbidity of late-preterm infants, including hyperbilirubinemia, sepsis, and respiratory distress syndrome. What is Known: • Late preterm infants have a higher morbidity and mortality rates when compared to term infants. • Low plasma vitamin A levels increase the risk of preterm infants' morbidity. What is New: • Late preterm infants commonly have low level of umbilical cord blood vitamin A. • Low umbilical cord blood vitamin A level at birth appears to be not associated with the morbidity of late-preterm infants. • Cesarean section is associated with low umbilical cord blood vitamin A level < 0.7 μmol/L compared with vaginal delivery.
Collapse
|
23
|
The Gut‒Breast Axis: Programming Health for Life. Nutrients 2021; 13:nu13020606. [PMID: 33673254 PMCID: PMC7917897 DOI: 10.3390/nu13020606] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The gut is a pivotal organ in health and disease. The events that take place in the gut during early life contribute to the programming, shaping and tuning of distant organs, having lifelong consequences. In this context, the maternal gut plays a quintessence in programming the mammary gland to face the nutritional, microbiological, immunological, and neuroendocrine requirements of the growing infant. Subsequently, human colostrum and milk provides the infant with an impressive array of nutrients and bioactive components, including microbes, immune cells, and stem cells. Therefore, the axis linking the maternal gut, the breast, and the infant gut seems crucial for a correct infant growth and development. The aim of this article is not to perform a systematic review of the human milk components but to provide an insight of their extremely complex interactions, which render human milk a unique functional food and explain why this biological fluid still truly remains as a scientific enigma.
Collapse
|
24
|
Zhang B, Liu E, Gertie JA, Joseph J, Xu L, Pinker EY, Waizman DA, Catanzaro J, Hamza KH, Lahl K, Gowthaman U, Eisenbarth SC. Divergent T follicular helper cell requirement for IgA and IgE production to peanut during allergic sensitization. Sci Immunol 2020; 5:5/47/eaay2754. [PMID: 32385053 DOI: 10.1126/sciimmunol.aay2754] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Immunoglobulin A (IgA) is the dominant antibody isotype in the gut and has been shown to regulate microbiota. Mucosal IgA is also widely believed to prevent food allergens from penetrating the gut lining. Even though recent work has elucidated how bacteria-reactive IgA is induced, little is known about how IgA to food antigens is regulated. Although IgA is presumed to be induced in a healthy gut at steady state via dietary exposure, our data do not support this premise. We found that daily food exposure only induced low-level, cross-reactive IgA in a minority of mice. In contrast, induction of significant levels of peanut-specific IgA strictly required a mucosal adjuvant. Although induction of peanut-specific IgA required T cells and CD40L, it was T follicular helper (TFH) cell, germinal center, and T follicular regulatory (TFR) cell-independent. In contrast, IgG1 and IgE production to peanut required TFH cells. These data suggest an alternative paradigm in which the cellular mechanism of IgA production to food antigens is distinct from IgE and IgG1. We developed an equivalent assay to study this process in stool samples from healthy, nonallergic humans, which revealed substantial levels of peanut-specific IgA that were stable over time. Similar to mice, patients with loss of CD40L function had impaired titers of gut peanut-specific IgA. This work challenges two widely believed but untested paradigms about antibody production to dietary antigens: (i) the steady state/tolerogenic response to food antigens includes IgA production and (ii) TFH cells drive food-specific gut IgA.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elisha Y Pinker
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Columbia University, New York, NY 10027, USA
| | - Daniel A Waizman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jason Catanzaro
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Pulmonology, Allergy, Immunology and Sleep Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Kedir Hussen Hamza
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden
| | - Katharina Lahl
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden.,Division of Biopharma, Institute for Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
25
|
Ahmad SM, Huda MN, Raqib R, Qadri F, Alam MJ, Afsar MNA, Peerson JM, Tanumihardjo SA, Stephensen CB. High-Dose Neonatal Vitamin A Supplementation to Bangladeshi Infants Increases the Percentage of CCR9-Positive Treg Cells in Infants with Lower Birthweight in Early Infancy, and Decreases Plasma sCD14 Concentration and the Prevalence of Vitamin A Deficiency at Two Years of Age. J Nutr 2020; 150:3005-3012. [PMID: 32939553 PMCID: PMC7675026 DOI: 10.1093/jn/nxaa260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Vitamin A (VA) stores are low in early infancy and may impair development of the immune system. OBJECTIVE This study determined if neonatal VA supplementation (VAS) affects the following: 1) development of regulatory T (Treg) cells; 2) chemokine receptor 9 (CCR9) expression, which directs mucosal targeting of immune cells; and 3) systemic endotoxin exposure as indicated by changed plasma concentrations of soluble CD14 (sCD14). Secondarily, VA status, growth, and systemic inflammation were investigated. METHODS In total, 306 Bangladeshi infants were randomly assigned to receive 50,000 IU VA or placebo (PL) within 48 h of birth, and immune function was assessed at 6 wk, 15 wk, and 2 y. Primary outcomes included the following: 1) peripheral blood Treg cells; 2) percentage of Treg, T, and B cells expressing CCR9; and 3) plasma sCD14. Secondary outcomes included the following: 4) VA status measured using the modified relative dose-response (MRDR) test and plasma retinol; 5) infant growth; and 6) plasma C-reactive protein (CRP). Statistical analysis identified group differences and interactions with sex and birthweight. RESULTS VAS increased (P = 0.004) the percentage of CCR9+ Treg cells (13.2 ± 1.37%) relative to PL (9.17 ± 1.15%) in children below the median birthweight but had the opposite effect (P = 0.04) in those with higher birthweight (VA, 9.13 ± 0.89; PL, 12.1 ± 1.31%) at 6 and 15 wk (values are combined mean ± SE). VAS decreased (P = 0.003) plasma sCD14 (1.56 ± 0.025 mg/L) relative to PL (1.67 ± 0.032 mg/L) and decreased (P = 0.034) the prevalence of VA deficiency (2.3%) relative to PL (9.2%) at 2 y. CONCLUSIONS Neonatal VAS enhanced mucosal targeting of Treg cells in low-birthweight infants. The decreased systemic exposure to endotoxin and improved VA status at 2 y may have been due to VA-mediated improvements in gut development resulting in improved barrier function and nutrient absorption. This trial was registered at clinicaltrials.gov as NCT01583972 and NCT02027610.
Collapse
Affiliation(s)
- Shaikh M Ahmad
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - M Nazmul Huda
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
- USDA Western Human Nutrition Research Center at University of California, Davis, CA, USA
- Nutrition Department, University of California, Davis, CA, USA
| | - Rubhana Raqib
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Md Jahangir Alam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Md Nure Alam Afsar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Janet M Peerson
- USDA Western Human Nutrition Research Center at University of California, Davis, CA, USA
| | - Sherry A Tanumihardjo
- University of Wisconsin–Madison, Department of Nutritional Sciences, Madison, WI, USA
| | - Charles B Stephensen
- USDA Western Human Nutrition Research Center at University of California, Davis, CA, USA
- Nutrition Department, University of California, Davis, CA, USA
| |
Collapse
|
26
|
Altered vaginal microbiome and relative co-abundance network in pregnant women with penicillin allergy. Allergy Asthma Clin Immunol 2020; 16:79. [PMID: 32944033 PMCID: PMC7491301 DOI: 10.1186/s13223-020-00475-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022] Open
Abstract
Background Penicillin allergy is frequently reported in adults and children. Recent studies suggest that microbiota plays a key role in the development and progression of allergy. In this study, the relationship between vaginal microbiome and pregnant women with penicillin allergy was investigated. Methods Vaginal samples before labor from 12 pregnant women with penicillin allergy and 15 non-allergic pregnant women were collected. Bacterial community structure of all study subjects and the discrepancies between the two groups were analyzed using 16S rRNA sequencing based on Illumina Hiseq 2500 platform. Results The abundant phyla among all participants were Firmicutes, Actinobacteria and Bacteroidetes. The predominant genus was Lactobacillus. Compared to non-allergic pregnant women, Actinobacteria, Coriobacteriaceae, Lachnospiraceae, Paraprevotella and Anoxybacillus significantly decreased, whereas Deltaproteobacteria, Peptostreptococcaceae, Enterococcus and Megamonas were more abundant in penicillin allergic women. Additionally, obvious discrepancies were observed in the co-abundance network at the genus level between the two groups. Conclusions There were differences in the microbial community structure and composition of reproduction tract between penicillin allergic and non-allergic pregnant women. These shifts may be related to maternal and neonatal health.
Collapse
|
27
|
Sanidad KZ, Zeng MY. Neonatal gut microbiome and immunity. Curr Opin Microbiol 2020; 56:30-37. [PMID: 32634598 PMCID: PMC8729197 DOI: 10.1016/j.mib.2020.05.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022]
Abstract
Early life is a critical time window for the neonatal gut to be progressively populated with different bacterial species that collectively promote gut maturation. A fully developed and healthy gut microbiome in neonates is an important driver for the development of other aspects of health. Unlike the relatively stable gut microbiome in adults, the developing gut microbiome in neonates exhibits higher plasticity and adaptability. This also underscores the unique window of opportunity for intervention or preventive measures to improve long-term health through modulations of the gut microbiome in early life. Better understanding of the neonatal gut microbiome - how it arises and how it impacts immune cell development - will help us appreciate the underpinnings of immune-related diseases. Here, we examine recent findings on the neonatal gut microbiome and discuss their implications for understanding this important driver of the maturation of the immune system and immunity against infections in early life.
Collapse
Affiliation(s)
- Katherine Z Sanidad
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York City, NY, United States
| | - Melody Y Zeng
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York City, NY, United States; Department of Pediatrics, Weill Cornell Medicine, New York City, NY, United States.
| |
Collapse
|
28
|
Adel-Patient K, Bernard H, Fenaille F, Hazebrouck S, Junot C, Verhasselt V. Prevention of Allergy to a Major Cow's Milk Allergen by Breastfeeding in Mice Depends on Maternal Immune Status and Oral Exposure During Lactation. Front Immunol 2020; 11:1545. [PMID: 32849523 PMCID: PMC7396486 DOI: 10.3389/fimmu.2020.01545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background: The high incidence of food allergy in childhood points to the need of elucidating early life factors dictating allergy susceptibility. Here, we aim to address in a mouse model how the exposure to a major cow's milk allergen through breastmilk of mothers with different immune status influences food allergy outcome in offspring. Methods: BALB/cJ future dams were either kept naïve, or sensitized through the oral route using cholera toxin (“orally sensitized”) or through the i.p. route using alum (“i.p. sensitized”), or rendered fully tolerant (oral gavage without any adjuvant) to bovine β-lactoglobulin (BLG). After mating with naïve males and delivery, mothers were orally exposed or not to BLG during the whole lactation. Then, eight groups of lactating mothers were considered: naïve, i.p. sensitized, orally sensitized, or tolerant, each exposed or not during lactation. In order to specifically address breastmilk effects on their allergy susceptibility, pups from naïve-synchronized mothers were cross-fostered by the different groups of treated dams and lactating mothers at delivery. In some experiments, mothers kept their own pups to address a possible in utero effect. BLG antigen, BLG-specific antibodies, and BLG-immune complexes were measured in breastmilk from the different lactating mother groups. Allergic sensitization was monitored in 5-weeks old female offspring (n = 7–8/group of lactating mothers) by determining BLG-specific antibodies in plasma and splenocytes cytokine secretion after i.p. injections of BLG/alum. Allergic reaction to oral BLG challenge was evaluated by measuring mMCP1 in plasma. Results: Offspring was protected from one allergic i.p. sensitization when nursed by i.p. sensitized mothers, independently of BLG exposure during lactation. Orally sensitized dams conferred protection in offspring solely when exposed to BLG during lactation, while naïve mothers did not provide any protection upon BLG exposure. The levels of protection correlated with the levels of BLG-specific antibodies and BLG-immune complex in breastmilk. There was a trend for decreased sensitization in offspring breastfed by tolerant and exposed mothers, which was not associated with transfer of specific antibodies through breastmilk. Protection provided by nursing by treated/exposed mothers was not persistent after a boost i.p. injection of the progeny and then did not protect them from an allergic reaction induced at this time point. No additional in utero effects were evidenced. Conclusion: Our study demonstrates the strong potential of breastmilk to modulate immune response to a major cow's milk allergen in the progeny. It highlights the importance of maternal immune status and of her consumption of the allergen during lactation in dictating the outcomes in offspring. This opens perspectives where modulating maternal immune status might increase the chance of cow's milk allergy prevention in breastfed children.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hervé Bernard
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - François Fenaille
- Laboratoire du Métabolisme des Médicaments, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé, CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Stéphane Hazebrouck
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Christophe Junot
- Laboratoire d'Immuno-Allergie Alimentaire, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France.,Laboratoire du Métabolisme des Médicaments, Service de Pharmacologie et d'Immunoanalyse, Département Médicaments et Technologies pour la Santé, CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Valérie Verhasselt
- Chair of Human Lactology, School of Molecular Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
29
|
Ganal-Vonarburg SC, Hornef MW, Macpherson AJ. Microbial-host molecular exchange and its functional consequences in early mammalian life. Science 2020; 368:604-607. [PMID: 32381716 DOI: 10.1126/science.aba0478] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecules from symbiotic microorganisms pervasively infiltrate almost every organ system of a mammalian host, marking the initiation of microbial-host mutualism in utero, long before the newborn acquires its own microbiota. Starting from in utero development, when maternal microbial molecules can penetrate the placental barrier, we follow the different phases of adaptation through the life events of birth, lactation, and weaning, as the young mammal adapts to the microbes that colonize its body surfaces. The vulnerability of early-life mammals is mitigated by maternal detoxification and excretion mechanisms, the protective effects of maternal milk, and modulation of neonatal receptor systems. Host adaptations to microbial exposure during specific developmental windows are critical to ensure organ function for development, growth, and immunity.
Collapse
Affiliation(s)
- Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Mathias W Hornef
- Institute for Medical Microbiology, RWTH University Hospital, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Andrew J Macpherson
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland.
| |
Collapse
|
30
|
Rekima A, Bonnart C, Macchiaverni P, Metcalfe J, Tulic MK, Halloin N, Rekima S, Genuneit J, Zanelli S, Medeiros S, Palmer DJ, Prescott S, Verhasselt V. A role for early oral exposure to house dust mite allergens through breast milk in IgE-mediated food allergy susceptibility. J Allergy Clin Immunol 2020; 145:1416-1429.e11. [PMID: 31954775 DOI: 10.1016/j.jaci.2019.12.912] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Successful prevention of food allergy requires the identification of the factors adversely affecting the capacity to develop oral tolerance to food antigen in early life. OBJECTIVES This study sought to determine whether oral exposure to Dermatophagoides pteronyssinus through breast milk affects gut mucosal immunity with long-term effects on IgE-mediated food allergy susceptibility. METHODS Gut immunity was explored in 2-week-old mice breast-fed by mothers exposed to D pteronyssinus, protease-inactivated D pteronyssinus, or to PBS during lactation. We further analyzed oral tolerance to a bystander food allergen, ovalbumin (OVA). In a proof-of-concept study, Der p 1 and OVA levels were determined in 100 human breast milk samples and the association with prevalence of IgE-mediated egg allergy at 1 year was assessed. RESULTS Increased permeability, IL-33 levels, type 2 innate lymphoid cell activation, and Th2 cell differentiation were found in gut mucosa of mice nursed by mothers exposed to D pteronyssinus compared with PBS. This pro-Th2 gut mucosal environment inhibited the induction of antigen-specific FoxP3 regulatory T cells and the prevention of food allergy by OVA exposure through breast milk. In contrast, protease-inactivated D pteronyssinus had no effect on offspring gut mucosal immunity. Based on the presence of Der p 1 and/or OVA in human breast milk, we identified groups of lactating mothers, which mirror the ones found in mice to be responsible for different egg allergy risk. CONCLUSIONS This study highlights an unpredicted potential risk factor for the development of food allergy, that is, D pteronyssinus allergens in breast milk, which disrupt gut immune homeostasis and prevents oral tolerance induction to bystander food antigen through their protease activity.
Collapse
Affiliation(s)
- Akila Rekima
- School of Molecular Sciences, University of Western Australia, Perth, Australia
| | - Chrystelle Bonnart
- Institut National de la Santé et de la Recherche Médicale, U1220, Toulouse, France
| | | | - Jessica Metcalfe
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Meri K Tulic
- EA6302 Immune Tolerance, Université de Nice Sophia-Antipolis, Nice, France; Institut National de la Santé et de la Recherche Médicale, U1065, Mediterranean Centre for Molecular Medicine, Team 12, Nice, France; inVIVO Global Network, Research Group of the Worldwide Universities Network, West New York, NJ
| | - Nicolas Halloin
- EA6302 Immune Tolerance, Université de Nice Sophia-Antipolis, Nice, France
| | - Samah Rekima
- Institut Biologie Valrose, Université Côte d'Azur, Institut National de la Santé et de la Recherche Medicale, Centre National de la Recherche Scientifique, Nice, France
| | - Jon Genuneit
- inVIVO Global Network, Research Group of the Worldwide Universities Network, West New York, NJ; Pediatric Epidemiology, Department of Pediatrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Samantha Zanelli
- EA6302 Immune Tolerance, Université de Nice Sophia-Antipolis, Nice, France
| | - Samara Medeiros
- EA6302 Immune Tolerance, Université de Nice Sophia-Antipolis, Nice, France; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Debra J Palmer
- Telethon Kids Institute, University of Western Australia, Perth, Australia; inVIVO Global Network, Research Group of the Worldwide Universities Network, West New York, NJ
| | - Susan Prescott
- Telethon Kids Institute, University of Western Australia, Perth, Australia; inVIVO Global Network, Research Group of the Worldwide Universities Network, West New York, NJ; Perth Childrens Hospital, Perth, Australia; School of Medicine, University of Western Australia, Crawley, Australia
| | - Valerie Verhasselt
- School of Molecular Sciences, University of Western Australia, Perth, Australia; inVIVO Global Network, Research Group of the Worldwide Universities Network, West New York, NJ.
| |
Collapse
|
31
|
Nagata Y, Yamamoto T, Kadowaki M. Ginger Increases ALDH1A1 Expression and Enhances Retinoic Acid Signaling in a Human Colonic Epithelial Cell Line. J Nutr Sci Vitaminol (Tokyo) 2020; 66:462-467. [PMID: 33132350 DOI: 10.3177/jnsv.66.462] [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: 11/27/2022]
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) in intestinal epithelial cells (IECs) plays a critical role in regulating immune responses through the production of retinoic acid (RA). However, little is known about its regulation by dietary components. We previously demonstrated that kakkonto, a Japanese traditional herbal medicine, and its constituent puerarin induce the expression of ALDH1A1 mRNA in colonic IECs and thereby attenuate food allergy symptoms in mice. This study aims to investigate the cellular responses of IECs to ALDH1A1 expression as a result of natural food components. The seven medicinal herbs that compose kakkonto were used to treat cultured an IEC line: Caco-2 cells. Expressions levels of ALDH1A1 were analyzed in Caco-2 cells by quantitative RT-PCR, immunocytochemistry and western blotting. Ginger increased the expression levels of ALDH1A1 mRNA and protein in Caco-2 cells. In addition, ginger significantly upregulated the gene expression of retinoic acid receptor (RAR) alpha (RARA), thereby enhancing RA signaling. Furthermore, ginger downregulated the expression of histone deacetylase (HDAC)2 (HDAC2) and HDAC3 in Caco-2 cells. The present study suggests the possibility that food ingredients such as a ginger modulate vitamin A metabolism in the gut through the regulation of RA synthesis, which may contribute to RA-mediated regulation of immune responses and the regulation of allergic inflammation.
Collapse
Affiliation(s)
- Yuka Nagata
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Takeshi Yamamoto
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama
| | - Makoto Kadowaki
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama
| |
Collapse
|
32
|
Hornef MW, Torow N. 'Layered immunity' and the 'neonatal window of opportunity' - timed succession of non-redundant phases to establish mucosal host-microbial homeostasis after birth. Immunology 2019; 159:15-25. [PMID: 31777069 PMCID: PMC6904599 DOI: 10.1111/imm.13149] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
The intricate host–microbial interaction and the overwhelming complexity of the mucosal immune system in the adult host raise the question of how this system is initially established. Here, we propose the implementation of the concept of the ‘postnatal window of opportunity’ into the model of a ‘layered immunity’ to explain how the newborn's mucosal immune system matures and how host–microbial immune homeostasis is established after birth. We outline the concept of a timed succession of non‐redundant phases during postnatal immune development and discuss the possible influence of external factors and conditions.
Collapse
Affiliation(s)
- Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
33
|
Lysosome-Rich Enterocytes Mediate Protein Absorption in the Vertebrate Gut. Dev Cell 2019; 51:7-20.e6. [PMID: 31474562 PMCID: PMC6783362 DOI: 10.1016/j.devcel.2019.08.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/06/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
The guts of neonatal mammals and stomachless fish have a limited capacity for luminal protein digestion, which allows oral acquisition of antibodies and antigens. However, how dietary protein is absorbed during critical developmental stages when the gut is still immature is unknown. Here, we show that specialized intestinal cells, which we call lysosome-rich enterocytes (LREs), internalize dietary protein via receptor-mediated and fluid-phase endocytosis for intracellular digestion and trans-cellular transport. In LREs, we identify a conserved endocytic machinery, composed of the scavenger receptor complex Cubilin/Amnionless and Dab2, that is required for protein uptake by LREs and for growth and survival of larval zebrafish. Moreover, impairing LRE function in suckling mice, via conditional deletion of Dab2, leads to stunted growth and severe protein malnutrition reminiscent of kwashiorkor, a devastating human malnutrition syndrome. These findings identify digestive functions and conserved molecular mechanisms in LREs that are crucial for vertebrate growth and survival.
Collapse
|
34
|
Fujimura T, Lum SZC, Nagata Y, Kawamoto S, Oyoshi MK. Influences of Maternal Factors Over Offspring Allergies and the Application for Food Allergy. Front Immunol 2019; 10:1933. [PMID: 31507589 PMCID: PMC6716146 DOI: 10.3389/fimmu.2019.01933] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
The prevalence of food allergy has been steadily rising worldwide with the highest incidence noted among younger children, and increasingly recognized as a growing public concern. The first known ingestion of foods often causes allergic reaction, suggesting that sensitization of offspring with food allergens may occur during pregnancy and/or through breastfeeding. This creates a milieu that shapes the neonatal immune responses to these allergens. However, the effects of maternal allergen exposure and maternal sensitization with allergens on development of allergies in offspring remain controversial. This review discusses recent advances from human data in our understanding of how maternal factors, namely, food allergens, allergen-specific immunoglobulins, cytokines, genetics, and environmental factors transferred during pregnancy or breastfeeding influence offspring allergies and how such effects may be applicable to food allergy. Based on information obtained from mouse models of asthma and food allergy, the review also dissects the mechanisms by which maternal factors, including the impact of immune complexes, transforming growth factor-β, vitamin A, and regulatory T-cell responses, contribute to the induction of neonatal tolerance vs. development of allergic responses to maternally transferred allergens.
Collapse
Affiliation(s)
- Takashi Fujimura
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | | | - Yuka Nagata
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Seiji Kawamoto
- Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | - Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
35
|
Torow N, Hornef MW. The Timed Pathway to Homeostasis. Immunity 2019; 50:1127-1129. [PMID: 31117008 DOI: 10.1016/j.immuni.2019.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Al Nabhani et al. (2019) describe the weaning reaction, a transient, microbiota-induced innate immune stimulation during the third and fourth weeks after birth that is associated with protection from immune-mediated enteric diseases in adulthood. This strictly timed, non-redundant process highlights the cooperative action of dietary, microbial, and developmental factors in the establishment of immune homeostasis.
Collapse
Affiliation(s)
- Natalia Torow
- Institute of Medical Microbiology, University Clinic RWTH Aachen, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, University Clinic RWTH Aachen, Aachen, Germany.
| |
Collapse
|
36
|
Järvinen KM, Martin H, Oyoshi MK. Immunomodulatory effects of breast milk on food allergy. Ann Allergy Asthma Immunol 2019; 123:133-143. [PMID: 31048004 DOI: 10.1016/j.anai.2019.04.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To summarize the literature on immunomodulatory effects of breast milk on sensitization and possible mechanisms of action. DATA SOURCES Animal and human studies in PubMed that assessed breastfeeding or breast milk composition in food allergy. STUDY SELECTIONS All recent studies and some older key publications focusing on this topic. RESULTS Human milk composition is highly variable among mothers, which can affect the developing infant immune system. Human milk also affects the infant gut microbiome, which is associated with food allergy. High levels of human milk immune factors (IgA, cytokines, oligosaccharides) are associated with reduced risk of food allergy in the infant; it remains uncertain whether these are directly protective or biomarkers of transferred protection. Animal studies highlight potential mechanisms of protection provided by antigens, transforming growth factor β, and immunocomplexes, yet their relevance is poorly understood in humans. The role of food antigens in human milk in initial sensitization or tolerance induction is unclear. CONCLUSION The protection against allergy development provided by human milk may be attributable to the effect on the infant gut microbiome or direct effects on immune system. Studies evaluating the effect of breastfeeding and human milk composition on food allergy are needed.
Collapse
Affiliation(s)
- Kirsi M Järvinen
- Division of Pediatric Allergy and Immunology & Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| | - Hayley Martin
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital and the Departments of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
37
|
Chen X, Mayne CG. The Role of Micronutrients in Graft-VS.-Host Disease: Immunomodulatory Effects of Vitamins A and D. Front Immunol 2018; 9:2853. [PMID: 30574143 PMCID: PMC6291446 DOI: 10.3389/fimmu.2018.02853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 01/15/2023] Open
Abstract
Graft-vs.-host disease (GVHD) remains a major obstacle to the success of allogeneic hematopoietic stem cell transplantation (HSCT). GVHD occurs because donor T cells in the allograft recognize the genetically disparate host as foreign and attack the transplant recipient's tissues. While genetic incompatibility between donor and recipient is the primary determinant for the extent of alloimmune response, GVHD incidence and severity are also influenced by non-genetic factors. Recent advances in immunology establish that environmental factors, including dietary micronutrients, contribute significantly to modulating various immune responses and may influence the susceptibility to autoimmune and inflammatory diseases of experimental animals and humans. Emerging clinical and preclinical evidence indicates that certain micronutrients may participate in regulating GVHD risk after allogeneic HSCT. In this review, we summarize recent advances in our understanding with respect to the potential role of micronutrients in the pathogenesis of acute and chronic GVHD, focusing on vitamins A and D.
Collapse
Affiliation(s)
- Xiao Chen
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|
38
|
Dietary and Nutritional Influences on Allergy Prevention. CURRENT TREATMENT OPTIONS IN ALLERGY 2018. [DOI: 10.1007/s40521-018-0182-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
39
|
McCoy KD, Ignacio A, Geuking MB. Microbiota and Type 2 immune responses. Curr Opin Immunol 2018; 54:20-27. [DOI: 10.1016/j.coi.2018.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
|
40
|
Clausen M, Jonasson K, Keil T, Beyer K, Sigurdardottir ST. Fish oil in infancy protects against food allergy in Iceland-Results from a birth cohort study. Allergy 2018; 73:1305-1312. [PMID: 29318622 PMCID: PMC6032905 DOI: 10.1111/all.13385] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Consumption of oily fish or fish oil during pregnancy, lactation and infancy has been linked to a reduction in the development of allergic diseases in childhood. METHODS In an observational study, Icelandic children (n = 1304) were prospectively followed from birth to 2.5 years with detailed questionnaires administered at birth and at 1 and 2 years of age, including questions about fish oil supplementation. Children with suspected food allergy were invited for physical examinations, allergic sensitization tests, and a double-blind, placebo-controlled food challenge if the allergy testing or clinical history indicated food allergy. The study investigated the development of sensitization to food and confirmed food allergy according to age and frequency of postnatal fish oil supplementation using proportional hazards modelling. RESULTS The incidence of diagnosed food sensitization was significantly lower in children who received regular fish oil supplementation (relative risk: 0.51, 95% confidence interval: 0.32-0.82). The incidence of challenge-confirmed food allergy was also reduced, although not statistically significant (0.57, 0.30-1.12). Children who began to receive fish oil in their first half year of life were significantly more protected than those who began later (P = .045 for sensitization, P = .018 for allergy). Indicators of allergy severity decreased with increased fish oil consumption (P = .013). Adjusting for parent education and allergic family history did not change the results. CONCLUSION Postnatal fish oil consumption is associated with decreased food sensitization and food allergies in infants and may provide an intervention strategy for allergy prevention.
Collapse
Affiliation(s)
- M. Clausen
- Children's HospitalLandspitali University HospitalReykjavikIceland
| | - K. Jonasson
- School of Engineering and Natural SciencesUniversity of IcelandReykjavíkIceland
| | - T. Keil
- Institute of Social Medicine, Epidemiology and Health EconomicsCharité ‐ Universitätsmedizin BerlinBerlinGermany
| | - K. Beyer
- Department of Paediatric Pneumology and ImmunologyCharité ‐ Universitätsmedizin BerlinBerlinGermany
| | - S. T. Sigurdardottir
- Department of ImmunologyLandspitali University HospitalReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
41
|
Hufnagl K, Jensen-Jarolim E. Vitamin A and D in allergy: from experimental animal models and cellular studies to human disease. ALLERGO JOURNAL 2018. [DOI: 10.1007/s15007-018-1579-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
42
|
McKenzie C, Tan J, Macia L, Mackay CR. The nutrition-gut microbiome-physiology axis and allergic diseases. Immunol Rev 2018; 278:277-295. [PMID: 28658542 DOI: 10.1111/imr.12556] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Indexed: 02/06/2023]
Abstract
Dietary and bacterial metabolites influence immune responses. This raises the question whether the increased incidence of allergies, asthma, some autoimmune diseases, cardiovascular disease, and others might relate to intake of unhealthy foods, and the decreased intake of dietary fiber. In recent years, new knowledge on the molecular mechanisms underpinning a 'diet-gut microbiota-physiology axis' has emerged to substantiate this idea. Fiber is fermented to short chain fatty acids (SCFAs), particularly acetate, butyrate, and propionate. These metabolites bind 'metabolite-sensing' G-protein-coupled receptors such as GPR43, GPR41, and GPR109A. These receptors play fundamental roles in the promotion of gut homeostasis and the regulation of inflammatory responses. For instance, these receptors and their metabolites influence Treg biology, epithelial integrity, gut homeostasis, DC biology, and IgA antibody responses. The SCFAs also influence gene transcription in many cells and tissues, through their inhibition of histone deacetylase expression or function. Contained in this mix is the gut microbiome, as commensal bacteria in the gut have the necessary enzymes to digest dietary fiber to SCFAs, and dysbiosis in the gut may affect the production of SCFAs and their distribution to tissues throughout the body. SCFAs can epigenetically modify DNA, and so may be one mechanism to account for diseases with a 'developmental origin', whereby in utero or post-natal exposure to environmental factors (such as nutrition of the mother) may account for disease later in life. If the nutrition-gut microbiome-physiology axis does underpin at least some of the Western lifestyle influence on asthma and allergies, then there is tremendous scope to correct this with healthy foodstuffs, probiotics, and prebiotics.
Collapse
Affiliation(s)
- Craig McKenzie
- Infection and Immunity Program, Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Jian Tan
- Infection and Immunity Program, Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Laurence Macia
- Nutritional Immunometabolism Node Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Charles R Mackay
- Infection and Immunity Program, Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| |
Collapse
|
43
|
Sakaguchi K, Koyanagi A, Kamachi F, Harauma A, Chiba A, Hisata K, Moriguchi T, Shimizu T, Miyake S. Breast-feeding regulates immune system development via transforming growth factor-β in mice pups. Pediatr Int 2018; 60:224-231. [PMID: 29290091 DOI: 10.1111/ped.13507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/17/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Breast milk contains important nutrients and immunoregulatory factors that are essential for newborn infants. Recently, epidemiological studies suggested that breast-feeding prevents a wide range of infectious diseases and lowers the incidence of infant allergic diseases. METHODS To examine the effects of breast milk on immunological development in infancy, we established an artificial rearing system for hand-feeding mice and compared mouse pups fed with either breast milk or milk substitute. All mice were killed at 14 days of age and immune cells in the thymus, spleen, and small intestine were examined on flow cytometry. RESULTS The number of thymocytes was higher whereas that of total immune cells of peripheral lymphoid tissues was lower in mice fed breast milk compared with milk substitute-fed mice. In peripheral lymphoid tissues, the proportion of B cells was higher and that of CD8+ T cells, macrophages, dendritic cells, and granulocytes was significantly lower in breast milk-fed mice. The same alteration in immune cells of the thymus and peripheral lymphoid tissues in milk substitute-fed mice was also observed in pups reared by mother mice treated with anti-transforming growth factor-β (anti-TGF-β) monoclonal antibody. CONCLUSIONS Breast milk regulates the differentiation and expansion of innate and adaptive immune cells partly due to TGF-β. Hence, TGF-β in breast milk may be a new therapeutic target for innate immune system-mediated diseases of infancy.
Collapse
Affiliation(s)
- Keita Sakaguchi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akemi Koyanagi
- Laboratory of Cell Biology, Juntendo University Graduate School of Medicine Research Center, Tokyo, Japan
| | - Fumitaka Kamachi
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akiko Harauma
- School of Life and Environmental Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Asako Chiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Hisata
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toru Moriguchi
- School of Life and Environmental Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
44
|
Vitamin A and D in allergy: from experimental animal models and cellular studies to human disease. ACTA ACUST UNITED AC 2018; 27:72-78. [PMID: 29707474 PMCID: PMC5910477 DOI: 10.1007/s40629-018-0054-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/22/2018] [Indexed: 12/28/2022]
Abstract
Introduction Vitamins A and D are able to modulate innate and adaptive immune responses and may therefore influence the development and the course of allergic diseases. Materials and methods This article reviews the current evidence for the experimental effects of vitamins A and D in vivo in animal models and on immune cells in vitro, and discusses their translational implication. A systematic literature search over the last 10 years was performed using MEDLINE and PubMed databases. Results Deficiencies of vitamin A or vitamin D in mouse models of allergic asthma seem to exacerbate allergic symptoms along with enhanced lung inflammation and Th2 cytokine production. In contrast, supplementation regimes especially with vitamin D were able to attenuate symptoms in therapeutic mouse models. The active metabolites retinoic acid (RA) and 1,25-dihydroxyvitamin D3 (VD3) induced tolerogenic dendritic cells (DCs) and up-regulated T‑regulatory cells in the allergic sensitization phase, which likely contributes to tolerance induction. Additionally, RA and VD3 maintained the stability of eosinophils and mast cells in the effector phase, thereby reducing allergic mediator release. Thus, both active vitamin metabolites RA and VD3 are able to influence allergic immune responses at several immunological sites. Conclusion Animal studies predict that vitamin A and D may also be attractive players in the control of allergy in humans. Whether these experimental observations can be translated to the human situation remains open, as results from clinical trials are controversial.
Collapse
|
45
|
Renz H, Adkins BD, Bartfeld S, Blumberg RS, Farber DL, Garssen J, Ghazal P, Hackam DJ, Marsland BJ, McCoy KD, Penders J, Prinz I, Verhasselt V, von Mutius E, Weiser JN, Wesemann DR, Hornef MW. The neonatal window of opportunity-early priming for life. J Allergy Clin Immunol 2017; 141:1212-1214. [PMID: 29247715 DOI: 10.1016/j.jaci.2017.11.019] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/23/2017] [Accepted: 11/15/2017] [Indexed: 01/05/2023]
Affiliation(s)
| | | | | | | | | | - Johan Garssen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Peter Ghazal
- University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - John Penders
- Care and Public Health Research Institute (Caphri) & NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Maastricht University, Maastricht, The Netherlands
| | - Immo Prinz
- Hannover Medical School, Hannover, Germany
| | - Valerie Verhasselt
- Family Larsson-Rosenquist Foundation Chair in Human Lactology, School of Molecular Science, University of Western Australia, Perth, Australia
| | - Erika von Mutius
- Dr von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany; Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich, Germany
| | | | | | | |
Collapse
|
46
|
Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, Macia L, Mackay CR. Dietary Fiber and Bacterial SCFA Enhance Oral Tolerance and Protect against Food Allergy through Diverse Cellular Pathways. Cell Rep 2017; 15:2809-24. [PMID: 27332875 DOI: 10.1016/j.celrep.2016.05.047] [Citation(s) in RCA: 443] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/02/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023] Open
Abstract
The incidence of food allergies in western countries has increased dramatically in recent decades. Tolerance to food antigens relies on mucosal CD103(+) dendritic cells (DCs), which promote differentiation of regulatory T (Treg) cells. We show that high-fiber feeding in mice improved oral tolerance and protected from food allergy. High-fiber feeding reshaped gut microbial ecology and increased the release of short-chain fatty acids (SCFAs), particularly acetate and butyrate. High-fiber feeding enhanced oral tolerance and protected against food allergy by enhancing retinal dehydrogenase activity in CD103(+) DC. This protection depended on vitamin A in the diet. This feeding regimen also boosted IgA production and enhanced T follicular helper and mucosal germinal center responses. Mice lacking GPR43 or GPR109A, receptors for SCFAs, showed exacerbated food allergy and fewer CD103(+) DCs. Dietary elements, including fiber and vitamin A, therefore regulate numerous protective pathways in the gastrointestinal tract, necessary for immune non-responsiveness to food antigens.
Collapse
Affiliation(s)
- Jian Tan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Craig McKenzie
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Gera Goverse
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Carola G Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, ACT 0200, Australia
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Laurence Macia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Department of Physiology, Faculty of Medicine, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Department of Physiology, Faculty of Medicine, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
47
|
Allergy prevention by breastfeeding: possible mechanisms and evidence from human cohorts. Curr Opin Allergy Clin Immunol 2017; 16:427-33. [PMID: 27518839 DOI: 10.1097/aci.0000000000000303] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Allergy is a modern disease which does not seem to benefit from breast milk preventive effects. We propose that maternal milk composition has not adapted to the needs of allergy prevention because of the recent and rapid increase of allergy. Modulation of breast milk composition may be the best strategy to counteract allergy development. We will review recent advances in understanding of allergy physiopathology and how breast milk factors may be specifically appropriate to interfere with allergy development in early life. RECENT FINDINGS There is strong evidence both from rodent and human studies that breast milk factors may impact on parameters which are now recognized to be essential for allergy physiopathology: infant gut barrier function, microbiota metabolites production, and oral tolerance induction. Data from human cohorts support the possibility to modify breast milk composition by selected interventions and to impact health outcomes in offspring. SUMMARY Nutritional intervention in lactating mothers should endow breast milk with the capacity to combat allergy epidemics in addition to infectious disease.
Collapse
|
48
|
Human Milk and Allergic Diseases: An Unsolved Puzzle. Nutrients 2017; 9:nu9080894. [PMID: 28817095 PMCID: PMC5579687 DOI: 10.3390/nu9080894] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to the development of allergic sensitisation and allergic disease. Studies vary in methodology and definition of outcomes, which lead to considerable heterogeneity. Human milk composition varies both within and between individuals, which may partially explain conflicting data. It is known that human milk composition is very complex and contains variable levels of immune active molecules, oligosaccharides, metabolites, vitamins and other nutrients and microbial content. Existing evidence suggests that modulation of human breast milk composition has potential for preventing allergic diseases in early life. In this review, we discuss associations between breastfeeding/human milk composition and allergy development.
Collapse
|
49
|
Torow N, Hornef MW. The Neonatal Window of Opportunity: Setting the Stage for Life-Long Host-Microbial Interaction and Immune Homeostasis. THE JOURNAL OF IMMUNOLOGY 2017; 198:557-563. [PMID: 28069750 DOI: 10.4049/jimmunol.1601253] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023]
Abstract
The existence of a neonatal window was first highlighted by epidemiological studies that revealed the particular importance of this early time in life for the susceptibility to immune-mediated diseases in humans. Recently, the first animal studies emerged that present examples of early-life exposure-triggered persisting immune events, allowing a detailed analysis of the factors that define this particular time period. The enteric microbiota and the innate and adaptive immune system represent prime candidates that impact on the pathogenesis of immune-mediated diseases and are known to reach a lasting homeostatic equilibrium following a dynamic priming period after birth. In this review, we outline the postnatal establishment of the microbiota and maturation of the innate and adaptive immune system and discuss examples of early-life exposure-triggered immune-mediated diseases that start to shed light on the critical importance of the early postnatal period for life-long immune homeostasis.
Collapse
Affiliation(s)
- Natalia Torow
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
50
|
Jensen-Jarolim E, Pali-Schöll I, Roth-Walter F. Outstanding animal studies in allergy I. From asthma to food allergy and anaphylaxis. Curr Opin Allergy Clin Immunol 2017; 17:169-179. [PMID: 28346234 PMCID: PMC5424575 DOI: 10.1097/aci.0000000000000363] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Animal models published within the past 18 months on asthma, food allergy and anaphylaxis, all conditions of rising public health concern, were reviewed. RECENT FINDINGS While domestic animals spontaneously develop asthma, food allergy and anaphylaxis, in animal models, divergent sensitization and challenge routes, dosages, intervals and antigens are used to induce asthmatic, food allergic or anaphylactic phenotypes. This must be considered in the interpretation of results. Instead of model antigens, gradually relevant allergens such as house dust mite in asthma, and food allergens like peanut, apple and peach in food allergy research were used. Novel engineered mouse models such as a mouse with a T-cell receptor for house dust mite allergen Der p 1, or with transgenic human hFcγR genes, facilitated the investigation of single molecules of interest. Whole-body plethysmography has become a state-of-the-art in-vivo readout in asthma research. In food allergy and anaphylaxis research, novel techniques were developed allowing real-time monitoring of in-vivo effects following allergen challenge. Networks to share tissues were established as an effort to reduce animal experiments in allergy which cannot be replaced by in-vitro measures. SUMMARY Natural and artificial animal models were used to explore the pathophysiology of asthma, food allergy and anaphylaxis and to improve prophylactic and therapeutic measures. Especially the novel mouse models mimicking molecular aspects of the complex immune network in asthma, food allergy and anaphylaxis will facilitate proof-of-concept studies under controlled conditions.
Collapse
Affiliation(s)
- Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| |
Collapse
|