1
|
Ohtsuka Y, Shimizu T. Immaturity of gut functions and induction of tolerance during early infancy. Pediatr Int 2025; 67:e15855. [PMID: 39739631 DOI: 10.1111/ped.15855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/02/2025]
Abstract
Tolerance to foreign molecules is primarily induced through three pathways: anergy, active suppression, and clonal deletion. The immaturity of gut functions, including digestion and barrier protection against foreign molecules during early infancy, is closely linked to the induction of tolerance. A significant number of undigested peptides can pass through leaky gut walls during this period, making it an opportune time to introduce active suppression and clonal deletion in the intestine. Increased production and expression of TGF-β and CXCL13 with lymphoid hyperplasia in early infancy can be a convenient way to introduce tolerance and immunoglobulin A (IgA) synthesis against food and other antigens to protect hosts from allergic diseases. In this article we will discuss the immaturity of gut functions and mucosal immunity in infancy and its relationship to the introduction of tolerance.
Collapse
Affiliation(s)
- Yoshikazu Ohtsuka
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
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
|
Gelsomino M, Liotti L, Barni S, Mori F, Giovannini M, Mastrorilli C, Pecoraro L, Saretta F, Castagnoli R, Arasi S, Klain A, del Giudice MM, Novembre E. Elimination Diets in Lactating Mothers of Infants with Food Allergy. Nutrients 2024; 16:2317. [PMID: 39064760 PMCID: PMC11279873 DOI: 10.3390/nu16142317] [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: 06/09/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Breastfeeding is the most important nutrition source for infants. However, managing breastfed infants with signs and symptoms related to food allergy can be difficult. Many studies have shown the presence of different food allergens in breast milk, but the clinical role of these antigens in human milk is still much debated. Milk is the main suspect in exclusively breastfed infants with signs and symptoms attributable to food allergy, even if other foods may be responsible. This narrative review analyzes the recommendations provided by international guidelines to determine the diagnosis and management of IgE-mediated and non-IgE-mediated food allergies in exclusively breastfed infants. Dietary restrictions in lactating mothers of infants with suspected FA are usually not necessary. Only in the very few cases where significant allergy signs and symptoms occur in the infant during exclusive breastfeeding should the lactating mother follow an elimination diet for the suspected food for a short period.
Collapse
Affiliation(s)
- Mariannita Gelsomino
- Pediatric Allergy Unit, Department of Life Sciences and Public Health, University Foundation Policlinico Gemelli IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Lucia Liotti
- Pediatric Unit, Department of Mother and Child Health, Salesi Children’s Hospital, 60123 Ancona, Italy;
| | - Simona Barni
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.)
| | - Francesca Mori
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.)
| | - Mattia Giovannini
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.)
- Department of Health Sciences, University of Florence, 50139 Florence, Italy;
| | - Carla Mastrorilli
- Pediatric Hospital Giovanni XXIII, Pediatric and Emergency Department, AOU Policlinic of Bari, 70126 Bari, Italy;
| | - Luca Pecoraro
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy;
| | - Francesca Saretta
- Pediatric Department, Latisana-Palmanova Hospital, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy;
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stefania Arasi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Angela Klain
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Michele Miraglia del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Elio Novembre
- Department of Health Sciences, University of Florence, 50139 Florence, Italy;
| |
Collapse
|
4
|
Zhan-Dai S, Grases-Pintó B, Lamuela-Raventós RM, Castell M, Pérez-Cano FJ, Vallverdú-Queralt A, Rodríguez-Lagunas MJ. Exploring the Impact of Extra Virgin Olive Oil on Maternal Immune System and Breast Milk Composition in Rats. Nutrients 2024; 16:1785. [PMID: 38892716 PMCID: PMC11174597 DOI: 10.3390/nu16111785] [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: 05/15/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Maternal breast milk plays a key role in providing newborns with passive immunity and stimulating the maturation of an infant's immune system, protecting them from many diseases. It is known that diet can influence the immune system of lactating mothers and the composition of their breast milk. The aim of this study was to establish if a supplementation during the gestation and lactation of Lewis rats with extra virgin olive oil (EVOO), due to the high proportion of antioxidant components in its composition, has an impact on the mother's immune system and on the breast milk's immune composition. For this, 10 mL/kg of either EVOO, refined oil (control oil) or water (REF group) were orally administered once a day to rats during gestation and lactation periods. Immunoglobulin (Ig) concentrations and gene expressions of immune molecules were quantified in several compartments of the mothers. The EVOO group showed higher IgA levels in both the breast milk and the mammary glands than the REF group. In addition, the gene expression of IgA in mammary glands was also boosted by EVOO consumption. Overall, EVOO supplementation during gestation and lactation is safe and does not negatively affect the mother's immune system while improving breast milk immune composition by increasing the presence of IgA, which could be critical for an offspring's immune health.
Collapse
Affiliation(s)
- Sonia Zhan-Dai
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (S.Z.-D.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
| | - Blanca Grases-Pintó
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (S.Z.-D.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
| | - Rosa M. Lamuela-Raventós
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Margarida Castell
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (S.Z.-D.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Francisco J. Pérez-Cano
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (S.Z.-D.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
| | - Anna Vallverdú-Queralt
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Maria José Rodríguez-Lagunas
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (S.Z.-D.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, 08921 Santa Coloma de Gramenet, Spain; (R.M.L.-R.); (A.V.-Q.)
| |
Collapse
|
5
|
Jiang Y, Li X, Zhang W, Ji Y, Yang K, Liu L, Zhang M, Qiao W, Zhao J, Du M, Fan X, Dang X, Chen H, Jiang T, Chen L. Effect of folA gene in human breast milk-derived Limosilactobacillus reuteri on its folate biosynthesis. Front Microbiol 2024; 15:1402654. [PMID: 38812695 PMCID: PMC11133606 DOI: 10.3389/fmicb.2024.1402654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction Folate supplementation is crucial for the human body, and the chemically synthesized folic acid might have undesirable side effects. The use of molecular breeding methods to modify the genes related to the biosynthesis of folate by probiotics to increase folate production is currently a focus of research. Methods In this study, the folate-producing strain of Limosilactobacillus reuteri B1-28 was isolated from human breast milk, and the difference between B1-28 and folA gene deletion strain ΔFolA was investigated by phenotyping, in vitro probiotic evaluation, metabolism and transcriptome analysis. Results The results showed that the folate producted by the ΔFolA was 2-3 folds that of the B1-28. Scanning electron microscope showed that ΔFolA had rougher surface, and the acid-producing capacity (p = 0.0008) and adhesion properties (p = 0.0096) were significantly enhanced than B1-28. Transcriptomic analysis revealed that differentially expressed genes were mainly involved in three pathways, among which the biosynthesis of ribosome and aminoacyl-tRNA occurred in the key metabolic pathways. Metabolomics analysis showed that folA affected 5 metabolic pathways, involving 89 different metabolites. Discussion In conclusion, the editing of a key gene of folA in folate biosynthesis pathway provides a feasible pathway to improve folate biosynthesis in breast milk-derived probiotics.
Collapse
Affiliation(s)
- Yu Jiang
- South Asia Branch of National Engineering Center of Dairy for Maternal and Child Health, Guilin University of Technology, Guilin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Xianping Li
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Wei Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Yadong Ji
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Kai Yang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Lu Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Minghui Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Mengjing Du
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Xiaofei Fan
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Xingfen Dang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Huo Chen
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Tiemin Jiang
- South Asia Branch of National Engineering Center of Dairy for Maternal and Child Health, Guilin University of Technology, Guilin, China
| | - Lijun Chen
- South Asia Branch of National Engineering Center of Dairy for Maternal and Child Health, Guilin University of Technology, Guilin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co., Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| |
Collapse
|
6
|
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
|
7
|
Govers C, Calder PC, Savelkoul HFJ, Albers R, van Neerven RJJ. Ingestion, Immunity, and Infection: Nutrition and Viral Respiratory Tract Infections. Front Immunol 2022; 13:841532. [PMID: 35296080 PMCID: PMC8918570 DOI: 10.3389/fimmu.2022.841532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
Respiratory infections place a heavy burden on the health care system, particularly in the winter months. Individuals with a vulnerable immune system, such as very young children and the elderly, and those with an immune deficiency, are at increased risk of contracting a respiratory infection. Most respiratory infections are relatively mild and affect the upper respiratory tract only, but other infections can be more serious. These can lead to pneumonia and be life-threatening in vulnerable groups. Rather than focus entirely on treating the symptoms of infectious disease, optimizing immune responsiveness to the pathogens causing these infections may help steer towards a more favorable outcome. Nutrition may have a role in such prevention through different immune supporting mechanisms. Nutrition contributes to the normal functioning of the immune system, with various nutrients acting as energy sources and building blocks during the immune response. Many micronutrients (vitamins and minerals) act as regulators of molecular responses of immune cells to infection. It is well described that chronic undernutrition as well as specific micronutrient deficiencies impair many aspects of the immune response and make individuals more susceptible to infectious diseases, especially in the respiratory and gastrointestinal tracts. In addition, other dietary components such as proteins, pre-, pro- and synbiotics, and also animal- and plant-derived bioactive components can further support the immune system. Both the innate and adaptive defense systems contribute to active antiviral respiratory tract immunity. The initial response to viral airway infections is through recognition by the innate immune system of viral components leading to activation of adaptive immune cells in the form of cytotoxic T cells, the production of neutralizing antibodies and the induction of memory T and B cell responses. The aim of this review is to describe the effects of a range different dietary components on anti-infective innate as well as adaptive immune responses and to propose mechanisms by which they may interact with the immune system in the respiratory tract.
Collapse
Affiliation(s)
- Coen Govers
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health Service (NHS) Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | | | - R. J. Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
- Research & Development, FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
8
|
Ghosh P, Rohatgi P, Bose K. Determinants of time-trends in exclusivity and continuation of breastfeeding in India: An investigation from the National Family Health Survey. Soc Sci Med 2021; 292:114604. [PMID: 34864276 DOI: 10.1016/j.socscimed.2021.114604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/06/2021] [Accepted: 11/23/2021] [Indexed: 11/28/2022]
Abstract
Breastfeeding is an essential key factor for reducing child mortality and the risk of disease. Therefore, to ensure health and wellbeing for all (Sustainable Development Goal 03), and to reduce inequalities in child health status (SDG 10), understanding the determinants of breastfeeding is essential. Our research aims to investigate determinants of different breastfeeding practices from 2005-06 (NFHS-3) to 2015-16 (NFHS-4), and changes in different breastfeeding practices among various demographic, social, religious, and economic groups during this decade. We have used a multivariate binary logistic regression model to estimate significant determinants of different breastfeeding practices. The results show an 8.4% hike in Exclusive Breastfeeding (EBF), along with a 3.3% and 1% drop in the Continuation of Breastfeeding (CBF) up to one and two years respectively during this decade in India. Significant variation has been found in EBF, CBF1 and CBF2 among various categories of child's birth interval, place of residence, social groups, economic communities, and geographic regions. Maternal nutritional status and anemia levels had significant influence on CBF1 and CBF2, and gender of the children on EBF and CBF2 from 2005-06 to 2015-16. There was no significant variation in CBF1 and CBF2 among children born in different birth orders, whereas EBF was significantly less among higher birth orders children. A dramatic improvement in EBF was found among first birth ordered children, teenage and/or highly educated mother, rich families, and in north India, and CBF2 was significantly raised only among higher educated mothers. The estimation of determinants throughout the decade is essential for improving government policies related to supporting and encouraging breastfeeding. The study suggests more initiatives and investment for the promotion, support and improvement of CBF for one and two years, and reduction of inequality among various demographic and socio-economic groups and geographic regions.
Collapse
Affiliation(s)
- Pritam Ghosh
- Department of Geography, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; Department of Geography, Ramsaday College, Amta, Howrah 711401, West Bengal, India.
| | - Pratima Rohatgi
- Department of Geography, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| | - Kaushik Bose
- Department of Anthropology, Vidyasagar University, Midnapore, West Bengal, India.
| |
Collapse
|
9
|
Rio-Aige K, Azagra-Boronat I, Castell M, Selma-Royo M, Collado MC, Rodríguez-Lagunas MJ, Pérez-Cano FJ. The Breast Milk Immunoglobulinome. Nutrients 2021; 13:nu13061810. [PMID: 34073540 PMCID: PMC8230140 DOI: 10.3390/nu13061810] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022] Open
Abstract
Breast milk components contribute to the infant’s immune development and protection, and among other immune factors, immunoglobulins (Igs) are the most studied. The presence of IgA in milk has been known for a long time; however, less information is available about the presence of other Igs such as IgM, IgG, and their subtypes (IgG1, IgG2, IgG3, and IgG4) or even IgE or IgD. The total Ig concentration and profile will change during the course of lactation; however, there is a great variability among studies due to several variables that limit establishing a clear pattern. In this context, the aim of this review was firstly to shed light on the Ig concentration in breast milk based on scientific evidence and secondly to study the main factors contributing to such variability. A search strategy provided only 75 studies with the prespecified eligibility criteria. The concentrations and proportions found have been established based on the intrinsic factors of the study—such as the sampling time and quantification technique—as well as participant-dependent factors, such as lifestyle and environment. All these factors contribute to the variability of the immunoglobulinome described in the literature and should be carefully addressed for further well-designed studies and data interpretation.
Collapse
Affiliation(s)
- Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| |
Collapse
|
10
|
Yokanovich LT, Newberry RD, Knoop KA. Regulation of oral antigen delivery early in life: Implications for oral tolerance and food allergy. Clin Exp Allergy 2021; 51:518-526. [PMID: 33403739 PMCID: PMC8743004 DOI: 10.1111/cea.13823] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/19/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022]
Abstract
The increasing incidence of food allergy remains a significant public health concern. Food allergy is partially due to a lack, or loss of tolerance to food allergens. Clinical outcomes surrounding early life practices, such as breastfeeding, antibiotic use and food allergen exposure, indicate the first year of life in children represents a unique time for shaping the immune system to reduce allergic outcomes. Animal models have identified distinctive aspects of when and where dietary antigens are delivered within the intestinal tract to promote oral tolerance prior to weaning. Additionally, animal models have identified contributions from maternal proteins from breast milk and bacterial products from the gut microbiota in regulating dietary antigen exposure and promoting oral tolerance, thus connecting decades of clinical observations on the benefits of breastfeeding, early food allergen introduction and antibiotic avoidance in the first year of life in reducing allergic outcomes. Here, we discuss how exposure to gut luminal antigens, including food allergens, is regulated in early life to generate protective tolerance and the implications of this process for preventing and treating food allergies.
Collapse
Affiliation(s)
| | - Rodney D. Newberry
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathryn A. Knoop
- Department of Immunology, Mayo Clinic, Rochester MN, USA
- Department of Pediatrics, Mayo Clinic, Rochester MN, USA
| |
Collapse
|
11
|
Dawod B, Haidl ID, Azad MB, Marshall JS. Toll-like receptor 2 impacts the development of oral tolerance in mouse pups via a milk-dependent mechanism. J Allergy Clin Immunol 2020; 146:631-641.e8. [DOI: 10.1016/j.jaci.2020.01.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/05/2020] [Accepted: 01/30/2020] [Indexed: 12/31/2022]
|
12
|
Tong L, Hao H, Zhang X, Zhang Z, Lv Y, Zhang L, Yi H. Oral Administration of Bovine Milk-Derived Extracellular Vesicles Alters the Gut Microbiota and Enhances Intestinal Immunity in Mice. Mol Nutr Food Res 2020; 64:e1901251. [PMID: 32180343 DOI: 10.1002/mnfr.201901251] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/01/2020] [Indexed: 12/17/2022]
Abstract
SCOPE Milk-derived extracellular vesicles (mEVs) as nanoparticles are being developed as novel drug vehicles due to their pivotal role in cell-cell communication. As an important bioactive component in milk, little is known about their effect on the gut microbiota and intestinal immunity. Therefore, the effects of mEVs on gut microbiota and intestinal immunity in mice are investigated. METHODS AND RESULTS First, a new method to obtain high-yield mEVs is developed. Afterward, the colonic contents from C57BL/6 mice fed different doses of mEVs (8 weeks) are collected and the microbial composition via 16S rRNA gene sequencing is analyzed. It is found that mEVs could alter the gut microbiota composition and modulate their metabolites-short-chain fatty acids (SCFAs). Furthermore, the effects of mEVs on intestinal immunity are evaluated. It is observed that the expression levels of Muc2, RegIIIγ, Myd88, GATA4 genes, and IgA, sIgA are increased in the intestine, which are significant for the integrity of the mucus layer. CONCLUSION These findings reveal that the genes with critical importance for intestinal barrier function and immune regulation are modified in mice by oral administration mEVs, which also result in the changes of the relative composition of gut microbiome and SCFAs.
Collapse
Affiliation(s)
- Lingjun Tong
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Haining Hao
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Xinyi Zhang
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Zhe Zhang
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Youyou Lv
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Lanwei Zhang
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Huaxi Yi
- Department of Food Science, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, P. R. China
| |
Collapse
|
13
|
Liu W, Chen M, Duo L, Wang J, Guo S, Sun H, Menghe B, Zhang H. Characterization of potentially probiotic lactic acid bacteria and bifidobacteria isolated from human colostrum. J Dairy Sci 2020; 103:4013-4025. [PMID: 32113772 DOI: 10.3168/jds.2019-17602] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/26/2019] [Indexed: 01/02/2023]
Abstract
Breast milk is the main source of nutrition for infants; it contains considerable microflora that can be transmitted to the infant endogenously or by breastfeeding, and it plays an important role in the maturation and development of the immune system. In this study, we isolated and identified lactic acid bacteria (LAB) from human colostrum, and screened 2 strains with probiotic potential. The LAB isolated from 40 human colostrum samples belonged to 5 genera: Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, and Staphylococcus. We also isolated Propionibacterium and Actinomyces. We identified a total of 197 strains of LAB derived from human colostrum based on their morphology and 16S rRNA sequence, among them 8 strains of Bifidobacterium and 10 strains of Lactobacillus, including 3 Bifidobacterium species and 4 Lactobacillus species. The physiological and biochemical characteristics of strains with good probiotic characteristics were evaluated. The tolerances of some of the Bifidobacterium and Lactobacillus strains to gastrointestinal fluid and bile salts were evaluated in vitro, using the probiotic strains Bifidobacterium lactis BB12 and Lactobacillus rhamnosus GG as controls. Among them, B. lactis Probio-M8 and L. rhamnosus Probio-M9 showed survival rates of 97.25 and 78.33% after digestion for 11 h in artificial gastrointestinal juice, and they exhibited growth delays of 0.95 and 1.87 h, respectively, in 0.3% bile salts. These two strains have the potential for application as probiotics and will facilitate functional studies of probiotics in breast milk and the development of human milk-derived probiotics.
Collapse
Affiliation(s)
- Wenjun Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Meixuan Chen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Lana Duo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jicheng Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Shuai Guo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Haotian Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Bilige Menghe
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
14
|
Nunes CF, Nogueira JS, Vianna PHO, Ciambarella BT, Rodrigues PM, Miranda KR, Lobo LA, Domingues RMCP, Busch M, Atella GC, Vale AM, Bellio M, Nóbrega A, Canto FB, Fucs R. Probiotic treatment during neonatal age provides optimal protection against experimental asthma through the modulation of microbiota and T cells. Int Immunol 2019; 30:155-169. [PMID: 29420746 DOI: 10.1093/intimm/dxy011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/02/2018] [Indexed: 02/06/2023] Open
Abstract
The incidence of allergic diseases, which increased to epidemic proportions in developed countries over the last few decades, has been correlated with altered gut microbiota colonization. Although probiotics may play a critical role in the restoration of gut homeostasis, their efficiency in the control of allergy is controversial. Here, we aimed to investigate the effects of probiotic treatment initiated at neonatal or adult ages on the suppression of experimental ovalbumin (OVA)-induced asthma. Neonatal or adult mice were orally treated with probiotic bacteria and subjected to OVA-induced allergy. Asthma-like symptoms, microbiota composition and frequencies of the total CD4+ T lymphocytes and CD4+Foxp3+ regulatory T (Treg) cells were evaluated in both groups. Probiotic administration to neonates, but not to adults, was necessary and sufficient for the absolute prevention of experimental allergen-induced sensitization. The neonatally acquired tolerance, transferrable to probiotic-untreated adult recipients by splenic cells from tolerant donors, was associated with modulation of gut bacterial composition, augmented levels of cecum butyrate and selective accumulation of Treg cells in the airways. Our findings reveal that a cross-talk between a healthy microbiota and qualitative features inherent to neonatal T cells, especially in the Treg cell subset, might support the beneficial effect of perinatal exposure to probiotic bacteria on the development of long-term tolerance to allergens.
Collapse
Affiliation(s)
- Caroline Fraga Nunes
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Jeane S Nogueira
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Pedro Henrique Oliveira Vianna
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Karla Rodrigues Miranda
- Faculdade de Farmácia - Universidade Federal do Rio de Janeiro, Rio de Janeiro/Campus Macaé, Macaé - RJ, Brazil
| | - Leandro Araújo Lobo
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil
| | | | - Mileane Busch
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro - RJ, Brazil
| | - Georgia Correa Atella
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro - RJ, Brazil
| | - André Macedo Vale
- Laboratório de Imunorreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil
| | - Maria Bellio
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nóbrega
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio B Canto
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Rita Fucs
- Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| |
Collapse
|
15
|
Berdi M, de Lauzon-Guillain B, Forhan A, Castelli FA, Fenaille F, Charles MA, Heude B, Junot C, Adel-Patient K. Immune components of early breastmilk: Association with maternal factors and with reported food allergy in childhood. Pediatr Allergy Immunol 2019; 30:107-116. [PMID: 30368940 DOI: 10.1111/pai.12998] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/28/2018] [Accepted: 10/06/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Breastmilk (BM) may participate in driving gut barrier function and immunity in the neonate. We analyzed immune and growth factor concentrations in early BM and their association with maternal/environmental characteristics and with food allergy (FA) in childhood. METHODS One BM sample was collected in maternity from some mothers in the EDEN birth cohort (n = 2002 mother-child dyads). A random selection was performed among available samples (subcohort, n = 272), for which all deliveries were full-term, various maternal/environmental characteristics were recorded, and parents answered yearly the question "Has a medical doctor diagnosed a FA in your child?" (26 parent-reported FA cases). Only samples collected between day 2 and day 6 post-partum were considered for descriptive analysis (n = 263). Samples for all other FA cases available were added to the subcohort (46 additional cases; "casecohort" design). Fifty cytokines, antibodies, and growth factor concentrations were determined using multiplexed kits and analyzed using robust statistical procedures. RESULTS BM components exhibited wide concentration ranges and global day-to-day variation. Different clusters of correlated factors appeared, with components from the main cluster related to maternal diet during pregnancy. Primiparity was positively associated with eleven other components, whereas other factors (eg, maternal atopy and smoking) were related to fewer components. Finally, the casecohort design highlighted a positive association between CXCL10, TNFβ, and IL-2 concentrations and reported FA in childhood. CONCLUSION Beyond the unique description of early BM composition, we show that immune information transmitted to the neonate is related to various maternal factors and identified components associated with FA diagnosis in childhood.
Collapse
Affiliation(s)
- Mikaïl Berdi
- UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Immuno-Allergie Alimentaire, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Blandine de Lauzon-Guillain
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center, Early Origin of the Child's Health and Development Team (ORCHAD), Paris, France.,Paris Descartes University, Paris, France
| | - Anne Forhan
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center, Early Origin of the Child's Health and Development Team (ORCHAD), Paris, France.,Paris Descartes University, Paris, France
| | - Florence Anne Castelli
- UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - François Fenaille
- UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Marie-Aline Charles
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center, Early Origin of the Child's Health and Development Team (ORCHAD), Paris, France.,Paris Descartes University, Paris, France
| | - Barbara Heude
- INSERM, UMR1153 Epidemiology and Biostatistics Sorbonne Paris Cité Center, Early Origin of the Child's Health and Development Team (ORCHAD), Paris, France.,Paris Descartes University, Paris, France
| | - Christophe Junot
- UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Immuno-Allergie Alimentaire, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France.,UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Karine Adel-Patient
- UMR Service de Pharmacologie et Immunoanalyse, Laboratoire d'Immuno-Allergie Alimentaire, CEA, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | | |
Collapse
|
16
|
Munblit D, Abrol P, Sheth S, Chow LY, Khaleva E, Asmanov A, Lauriola S, Padovani EM, Comberiati P, Boner AL, Warner JO, Boyle RJ, Peroni DG. Levels of Growth Factors and IgA in the Colostrum of Women from Burundi and Italy. Nutrients 2018; 10:E1216. [PMID: 30177587 PMCID: PMC6164593 DOI: 10.3390/nu10091216] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Colostrum is produced in the first days postpartum. It is a known source of immune mediators for a newborn within the first week of life. Although it is still unclear if colostrum composition varies between populations, recent data suggest differences. Hepatocyte growth factor (HGF); transforming growth factor-β (TGF-β) 1, 2, and 3; and immunoglobulin A (IgA) are key immunological components of colostrum that stimulate neonatal gastrointestinal and immune system development. We aimed to investigate the differences in the concentration between immune markers in the colostrum of mothers living in Burundi and Italy, and to identify the factors associated with differences. In this cross-sectional birth cohort study, a total of 99 colostrum samples from Burundian (n = 23) and Italian (n = 76) women were collected at 0 to 6 days postpartum. A clinical chemistry analyser was used for IgA quantification and electro-chemiluminescence, for HGF and TGFβ1-3 assessment. A univariate analysis and multivariate linear regression model were used for statistical testing. The concentrations of TGF-β2 (p = 0.01) and IgA (p < 0.01) were significantly higher in the colostrum from the women residing in Burundi than in Italy, both in a univariate analysis and upon the adjustment for confounding factors. A similar trend is seen for HGF, reaching statistical significance upon a multivariate analysis. We found a moderate to strong positive correlation between the TGF-β isoforms and IgA concentration in both countries (p < 0.01), with stronger concentration in the colostrum from Burundi. The results of this study are in support of previous data, suggesting that concentration of the immune active molecules is higher in the human milk of women residing in developing countries. However, with a small sample size, caution must be applied, as the findings require further confirmation. Future work should also be focused on other factors (e.g., lipid and microbial composition), as well as the investigation into colostrum and between populations comparison, adjusting for potential confounders.
Collapse
Affiliation(s)
- Daniel Munblit
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
- Faculty of Pediatrics, Sechenov University, 119991 Moscow, Russia.
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), 6010 Park Ave, West New York, NJ 07093, USA.
| | - Priya Abrol
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
| | - Shreya Sheth
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
| | - Li Yan Chow
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
| | - Ekaterina Khaleva
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), 6010 Park Ave, West New York, NJ 07093, USA.
- Department of Paediatrics, Saint-Petersburg State Paediatric Medical University, 194353 Saint-Petersburg, Russia.
| | - Alan Asmanov
- The Research and Clinical Institute for Pediatrics named after Academician Yuri Veltischev of the Pirogov Russian National Research Medical University, 125412 Moscow, Russia.
| | - Silvana Lauriola
- Department of Life and Reproduction Sciences, Section of Paediatrics, University of Verona, 37124 Verona, Italy.
| | - Ezio M Padovani
- Department of Life and Reproduction Sciences, Section of Paediatrics, University of Verona, 37124 Verona, Italy.
| | - Pasquale Comberiati
- Department of Life and Reproduction Sciences, Section of Paediatrics, University of Verona, 37124 Verona, Italy.
| | - Attilio L Boner
- Department of Life and Reproduction Sciences, Section of Paediatrics, University of Verona, 37124 Verona, Italy.
| | - John O Warner
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), 6010 Park Ave, West New York, NJ 07093, USA.
| | - Robert J Boyle
- Department of Paediatrics, Imperial College London, London W2 1NY, UK.
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), 6010 Park Ave, West New York, NJ 07093, USA.
| | - Diego G Peroni
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), 6010 Park Ave, West New York, NJ 07093, USA.
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
17
|
Abstract
The growth and maturity of the peripheral immune system and subsequent development of pulmonary immunity in early life is dictated by host, environmental and microbial factors. Dysregulation during the critical window of immune development in the postnatal years results in disease which impacts on lifelong lung health. Asthma is a common disease in childhood and is often preceded by wheezing illnesses during the preschool years. However, the mechanisms underlying development of wheeze and how and why only some children progress to asthma is unknown. Human studies to date have generally focused on peripheral immune development, with little assessment of local tissue pathology in young children. Moreover, mechanisms underlying the interactions between inflammation and tissue repair at mucosal surfaces in early life remain unknown. Disappointingly, mechanistic studies in mice have predominantly used adult models. This review will consider the aspects of the neonatal immune system which might contribute to the development of early life wheezing disorders and asthma, and discuss the external environmental factors which may influence this process.
Collapse
Affiliation(s)
- Clare M Lloyd
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, Royal Brompton Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
18
|
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
|
19
|
Jeurink PV, Knipping K, Wiens F, Barańska K, Stahl B, Garssen J, Krolak-Olejnik B. Importance of maternal diet in the training of the infant's immune system during gestation and lactation. Crit Rev Food Sci Nutr 2018; 59:1311-1319. [PMID: 29393671 DOI: 10.1080/10408398.2017.1405907] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Latest forecasts predict that half of the European population will be allergic within the coming 15 years, with food allergies contributing substantially to the total burden; preventive measures are urgently needed. Unfortunately, all attempted alimentary strategies for primary prevention of allergic diseases through allergen avoidance so far have failed. This also holds true for the prevention of food allergies in breastfed infants by the common practice of excluding certain foods with allergenic potential from the maternal diet. As a preventive measure, therefore, exclusion diets should be discouraged. They can exhaust nursing mothers and negatively impact both their nutritional status as well as their motivation to breastfeed. A prolonged exclusion diet may be indicated solely in cases of doctor-diagnosed food allergy following rigid medical tests (e.g. double-blind placebo-controlled food challenges). Indicated cases usually involve exclusion of only a few food items. Continued breastfeeding is generally important for many aspects of the infant's health, including the training of the infant's immune responses to foreign compounds and avoidance of overshooting inflammatory responses. Recent studies suggest that the presence of maternal dietary proteins in amniotic fluid, cord blood, and human milk might support the induction of tolerance towards solid foods in infants. These are exactly the same species of proteins or remnants thereof that, in comparatively few cases, trigger allergic responses. However, the insight that the proteins of maternal dietary origin in human milk are more likely to be cure (or, more precise, directing prevention) than curse has still largely evaded the attention of health care professionals consulted by worried breastfeeding mothers. In this paper, we summarize recent literature on the importance of exposure to dietary proteins in the establishment of immunological tolerance and hence prevention of allergic disease. Multiple organizations have used the scientific knowledge to build (local) guidelines (e.g. AAAAI, EAACI, BSACI) that can support health care professionals to provide the best strategy to prevent the onset of allergic diseases. We thus hope to clarify existing confusion about the allergenic propensities of dietary proteins during early life, which has contributed to exaggerated fears around the diet of pregnant and breastfeeding mothers.
Collapse
Affiliation(s)
- P V Jeurink
- a Nutricia Research , Utrecht , the Netherlands.,b Division of Pharmacology, Department of Pharmaceutical Sciences , Faculty of Science, Utrecht University , the Netherlands
| | - K Knipping
- a Nutricia Research , Utrecht , the Netherlands.,b Division of Pharmacology, Department of Pharmaceutical Sciences , Faculty of Science, Utrecht University , the Netherlands
| | - F Wiens
- a Nutricia Research , Utrecht , the Netherlands
| | - K Barańska
- c Department of Neonatology , Wroclaw Medical University , Wroclaw , Poland
| | - B Stahl
- a Nutricia Research , Utrecht , the Netherlands
| | - J Garssen
- a Nutricia Research , Utrecht , the Netherlands.,b Division of Pharmacology, Department of Pharmaceutical Sciences , Faculty of Science, Utrecht University , the Netherlands
| | - B Krolak-Olejnik
- c Department of Neonatology , Wroclaw Medical University , Wroclaw , Poland
| |
Collapse
|
20
|
Anderson RC, MacGibbon AKH, Haggarty N, Armstrong KM, Roy NC. Bovine dairy complex lipids improve in vitro measures of small intestinal epithelial barrier integrity. PLoS One 2018; 13:e0190839. [PMID: 29304106 PMCID: PMC5755888 DOI: 10.1371/journal.pone.0190839] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/20/2017] [Indexed: 11/18/2022] Open
Abstract
Appropriate intestinal barrier maturation is essential for absorbing nutrients and preventing pathogens and toxins from entering the body. Compared to breast-fed infants, formula-fed infants are more susceptible to barrier dysfunction-associated illnesses. In infant formula dairy lipids are usually replaced with plant lipids. We hypothesised that dairy complex lipids improve in vitro intestinal epithelial barrier integrity. We tested milkfat high in conjugated linoleic acid, beta serum (SureStart™Lipid100), beta serum concentrate (BSC) and a ganglioside-rich fraction (G600). Using Caco-2 cells as a model of the human small intestinal epithelium, we analysed the effects of the ingredients on trans-epithelial electrical resistance (TEER), mannitol flux, and tight junction protein co-localisation. BSC induced a dose-dependent improvement in TEER across unchallenged cell layers, maintained the co-localisation of tight junction proteins in TNFα-challenged cells with increased permeability, and mitigated the TEER-reducing effects of lipopolysaccharide (LPS). G600 also increased TEER across healthy and LPS-challenged cells, but it did not alter the co-location of tight junction proteins in TNFα-challenged cells. SureStart™Lipid100 had similar TEER-increasing effects to BSC when added at twice the concentration (similar lipid concentration). Ultimately, this research aims to contribute to the development of infant formulas supplemented with dairy complex lipids that support infant intestinal barrier maturation.
Collapse
Affiliation(s)
- Rachel C. Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Riddet Centre of Research Excellence, Massey University, Palmerston North, New Zealand
- * E-mail:
| | | | - Neill Haggarty
- Fonterra Research and Development Centre, Palmerston North, New Zealand
| | - Kelly M. Armstrong
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Nicole C. Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Riddet Centre of Research Excellence, Massey University, Palmerston North, New Zealand
| |
Collapse
|
21
|
Abstract
Breast feeding has been associated with improved infant outcomes in multiple aspects, including immune outcomes such as infections and potentially atopy and autoimmunity. However associations do not necessarily implicate cause and effect and at this point, exactly how breast feeding and components of breast milk may modulate the infant's immune compartment remains unclear, especially in humans. Some lines of evidence suggest that breastfeeding affects the development of the infant's thymus, a critical organ for T cell development. This may be a direct effect mediated by breast milk components or alternatively, a secondary effect from the impact of breast feeding on the infant's gut microbiome. Here we discuss the potential mechanisms and impact of this association between breast feeding and thymic development.
Collapse
Affiliation(s)
- Peter S Hsu
- Allergy and Immunology, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre Nepean, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Ohsaki A, Venturelli N, Buccigrosso TM, Osganian SK, Lee J, Blumberg RS, Oyoshi MK. Maternal IgG immune complexes induce food allergen-specific tolerance in offspring. J Exp Med 2017; 215:91-113. [PMID: 29158374 PMCID: PMC5748859 DOI: 10.1084/jem.20171163] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/24/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022] Open
Abstract
The role of maternal immune responses in tolerance induction is poorly understood. To study whether maternal allergen sensitization affects offspring susceptibility to food allergy, we epicutaneously sensitized female mice with ovalbumin (OVA) followed by epicutaneous sensitization and oral challenge of their offspring with OVA. Maternal OVA sensitization prevented food anaphylaxis, OVA-specific IgE production, and intestinal mast cell expansion in offspring. This protection was mediated by neonatal crystallizable fragment receptor (FcRn)-dependent transfer of maternal IgG and OVA immune complexes (IgG-IC) via breast milk and induction of allergen-specific regulatory T (T reg) cells in offspring. Breastfeeding by OVA-sensitized mothers or maternal supplementation with IgG-IC was sufficient to induce neonatal tolerance. FcRn-dependent antigen presentation by CD11c+ dendritic cells (DCs) in offspring was required for oral tolerance. Human breast milk containing OVA-IgG-IC induced tolerance in humanized FcRn mice. Collectively, we demonstrate that interactions of maternal IgG-IC and offspring FcRn are critical for induction of T reg cell responses and control of food-specific tolerance in neonates.
Collapse
Affiliation(s)
- Asa Ohsaki
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | | | | | | | - John Lee
- Division of Immunology, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Richard S Blumberg
- Gastroenterology Division, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA.,Harvard Digestive Diseases Center, Boston, MA
| | - Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital, Boston, MA .,Department of Pediatrics, Harvard Medical School, Boston, MA
| |
Collapse
|
23
|
Girbovan A, Sur G, Samasca G, Lupan I. Is the evidence of breast feeding protection against coeliac disease real? Allergol Immunopathol (Madr) 2017; 45:616-618. [PMID: 28410871 DOI: 10.1016/j.aller.2017.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/24/2017] [Indexed: 10/19/2022]
Abstract
Many recent studies discredit breastfeeding protection against coeliac disease. We will try to answer the question: "Is the evidence of breast feeding protection against coeliac disease real?"
Collapse
|
24
|
Vieira Borba V, Sharif K, Shoenfeld Y. Breastfeeding and autoimmunity: Programing health from the beginning. Am J Reprod Immunol 2017; 79. [PMID: 29083070 DOI: 10.1111/aji.12778] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Breast milk is not only a completely adapted nutrition source for the newborn but also an impressive array of immune-active molecules that afford protection against infections and shape mucosal immune responses. Decisive imprinting events might be modulated during the first months of life with potential health long-term effects, enhancing the importance of breastfeeding as a major influence on the immune system correct development and modifying disease susceptibility. The aim of this review was to clarify the link between breastfeeding and autoimmune diseases, inquiring the related mechanisms, based on data available in the literature. Being breastfed was associated with a lower incidence of diabetes, celiac disease, multiple sclerosis and asthma, explained by the protection against early infections, anti-inflammatory properties, antigen-specific tolerance induction, and regulation of infant's microbiome. The protective role of human milk in idiopathic juvenile arthritis, rheumatoid arthritis, and inflammatory bowel diseases remains controversial. On the other hand, the breastfeeding mother faces a health-challenging period in life. High levels of prolactin may lead either to the development of autoimmune diseases in susceptible mothers or exacerbations of current immune-mediated disorders. These features raise the question if mothers with autoimmune diseases, mainly systemic lupus erythematosus, should avoid breastfeeding.
Collapse
Affiliation(s)
- Vânia Vieira Borba
- Department 'A' of Internal Medicine, Coimbra University Hospital Centre, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Kassem Sharif
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Department 'B' of Internal Medicine, Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
25
|
Ashigai H, Komano Y, Wang G, Kawachi Y, Sunaga K, Yamamoto R, Takata R, Miyake M, Yanai T. Polysaccharide from black currant ( Ribes nigrum L.) stimulates dendritic cells through TLR4 signaling. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2017; 36:141-145. [PMID: 29038769 PMCID: PMC5633528 DOI: 10.12938/bmfh.16-029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/20/2017] [Indexed: 11/05/2022]
Abstract
Black currant (Ribes nigrum) has various beneficial properties for human health. In particular, polysaccharide from black currant was found to be an immunostimulating food ingredient and was reported to have antitumor activity in a mouse model. We named it cassis polysaccharide (CAPS). In a previous study, CAPS administration caused tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) production in vitro and in vivo, but the immunological mechanism of CAPS was not demonstrated. In this study, we revealed the CAPS immunostimulating mechanism in vitro. First, we found that CAPS activated dendritic cells (DCs). Second, we investigated whether it depends on Toll-like receptor 4 (TLR4) and myeloid differentiation primary response (Myd). We concluded that CAPS stimulates DCs through Myd88 depending TLR4 signaling and activates Th1-type cytokine release.
Collapse
Affiliation(s)
- Hiroshi Ashigai
- Research Laboratories for Health Science & Food Technologies, Kirin Company, Ltd., 1-17-1 Namamugi, Tsurumi-ku, Yokohama 230-8628, Japan
| | - Yuta Komano
- Research Laboratories for Health Science & Food Technologies, Kirin Company, Ltd., 1-17-1 Namamugi, Tsurumi-ku, Yokohama 230-8628, Japan
| | - Guanying Wang
- Research Laboratories for Health Science & Food Technologies, Kirin Company, Ltd., 1-17-1 Namamugi, Tsurumi-ku, Yokohama 230-8628, Japan
| | - Yasuji Kawachi
- Research Laboratories for Health Science & Food Technologies, Kirin Company, Ltd., 1-17-1 Namamugi, Tsurumi-ku, Yokohama 230-8628, Japan
| | - Kazuko Sunaga
- Marketing Department, Mercian Corporation, 4-10-2 Nakano, Nakano-ku, Tokyo 164-0001, Japan
| | - Reiko Yamamoto
- Research Laboratories for Wine Technologies, Kirin Company, Ltd., 4-9-1 Johnan, Fujisawa 251-0057, Japan
| | - Ryoji Takata
- Research Laboratories for Wine Technologies, Kirin Company, Ltd., 4-9-1 Johnan, Fujisawa 251-0057, Japan
| | - Mika Miyake
- Research Laboratories for Health Science & Food Technologies, Kirin Company, Ltd., 1-17-1 Namamugi, Tsurumi-ku, Yokohama 230-8628, Japan
| | - Takaaki Yanai
- Research Laboratories for Wine Technologies, Kirin Company, Ltd., 4-9-1 Johnan, Fujisawa 251-0057, Japan
| |
Collapse
|
26
|
Indrio F, Riezzo G, Tafuri S, Ficarella M, Carlucci B, Bisceglia M, Polimeno L, Francavilla R. Probiotic Supplementation in Preterm: Feeding Intolerance and Hospital Cost. Nutrients 2017; 9:nu9090965. [PMID: 28858247 PMCID: PMC5622725 DOI: 10.3390/nu9090965] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022] Open
Abstract
We hypothesized that giving the probiotic strain Lactobacillus reuteri (L. reuteri) DSM 17938 to preterm, formula-fed infants would prevent an early traumatic intestinal inflammatory insult modulating intestinal cytokine profile and reducing the onset of feeding intolerance. Newborn were randomly allocated during the first 48 h of life to receive either daily probiotic (108 colony forming units (CFUs) of L. reuteri DSM 17938) or placebo for one month. All the newborns underwent to gastric ultrasound for the measurement of gastric emptying time. Fecal samples were collected for the evaluation of fecal cytokines. Clinical data on feeding intolerance and weight gain were collected. The costs of hospital stays were calculated. The results showed that the newborns receiving L. reuteri DSM 17938 had a significant decrease in the number of days needed to reach full enteral feeding (p < 0.01), days of hospital stay (p < 0.01), and days of antibiotic treatment (p < 0.01). Statistically significant differences were observed in pattern of fecal cytokine profiles. The anti-inflammatory cytokine interleukin (IL)-10, was increased in newborns receiving L. reuteri DSM 17938. Pro-inflammatory cytokines: IL-17, IL-8, and tumor necrosis factor (TNF)-alpha levels were increased in newborns given placebo. Differences in the gastric emptying and fasting antral area (FAA) were also observed. Our study demonstrates an effective role for L. reuteri DSM 17938 supplementation in preventing feeding intolerance and improving gut motor and immune function development in bottle-fed stable preterm newborns. Another benefit from the use of probiotics is the reducing cost for the Health Care service.
Collapse
Affiliation(s)
- Flavia Indrio
- Department of Pediatric, Aldo Moro University of Bari, Ospedale Pediatrico Giovanni XXIII via Amendola 276, 70125 Bari, Italy.
| | - Giuseppe Riezzo
- Laboratory of Nutritional Physiopathology, National Institute for Digestive Diseases, Istituto di Ricerca e Cura a Carattere Scientifico (I.R.C.C.S.), Saverio de Bellis, 70013 Castellana Grotte (BA), Italy.
| | - Silvio Tafuri
- Department of Biomedical Sciences and Human Oncology, Section of Hygiene, Aldo Moro University of Bari, 70125 Bari, Italy.
| | - Maria Ficarella
- Department of Pediatric, Aldo Moro University of Bari, Ospedale Pediatrico Giovanni XXIII via Amendola 276, 70125 Bari, Italy.
| | - Barbara Carlucci
- Division of Neonatology, Ospedale Perrino, 72100 Brindisi, Italy.
| | - Massimo Bisceglia
- Department of Pediatrics, Neonatology Division San Giovanni di Dio Hospital, 88900 Crotone, Italy.
| | - Lorenzo Polimeno
- Department of Organ Transplantation, Gastroenterology Section, University Aldo Moro Bari, 70125 Bari, Italy.
| | - Ruggiero Francavilla
- Department of Pediatric, Aldo Moro University of Bari, Ospedale Pediatrico Giovanni XXIII via Amendola 276, 70125 Bari, Italy.
| |
Collapse
|
27
|
Ruiz L, Espinosa-Martos I, García-Carral C, Manzano S, McGuire MK, Meehan CL, McGuire MA, Williams JE, Foster J, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Kvist LJ, Otoo GE, Lackey KA, Flores K, Pareja RG, Bode L, Rodríguez JM. What's Normal? Immune Profiling of Human Milk from Healthy Women Living in Different Geographical and Socioeconomic Settings. Front Immunol 2017; 8:696. [PMID: 28713365 PMCID: PMC5492702 DOI: 10.3389/fimmu.2017.00696] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022] Open
Abstract
Human milk provides a very wide range of nutrients and bioactive components, including immune factors, human milk oligosaccharides, and a commensal microbiota. These factors are essential for interconnected processes including immunity programming and the development of a normal infant gastrointestinal microbiome. Newborn immune protection mostly relies on maternal immune factors provided through milk. However, studies dealing with an in-depth profiling of the different immune compounds present in human milk and with the assessment of their natural variation in healthy women from different populations are scarce. In this context, the objective of this work was the detection and quantification of a wide array of immune compounds, including innate immunity factors (IL1β, IL6, IL12, INFγ, TNFα), acquired immunity factors (IL2, IL4, IL10, IL13, IL17), chemokines (IL8, Groα, MCP1, MIP1β), growth factors [IL5, IL7, epidermal growth factor (EGF), granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, TGFβ2], and immunoglobulins (IgA, IgG, IgM), in milk produced by healthy women of different ethnicities living in different geographic, dietary, socioeconomic, and environmental settings. Among the analyzed factors, IgA, IgG, IgM, EGF, TGFβ2, IL7, IL8, Groα, and MIP1β were detected in all or most of the samples collected in each population and, therefore, this specific set of compounds might be considered as the "core" soluble immune factors in milk produced by healthy women worldwide. This approach may help define which immune factors are (or are not) common in milk produced by women living in various conditions, and to identify host, lifestyle, and environmental factors that affect the immunological composition of this complex biological fluid. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT02670278.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Irene Espinosa-Martos
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
- Probisearch S.L., C/Santiago Grisolía, Tres Cantos, Spain
| | - Cristina García-Carral
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Susana Manzano
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Michelle K. McGuire
- School of Biological Sciences, Washington State University, Pullman, WA, United States
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Mark A. McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - James Foster
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Daniel W. Sellen
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E. Moore
- Division of Women’s Health, King’s College London, London, United Kingdom
- MRC Unit, Serekunda, Gambia
| | | | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | - Kimberly A. Lackey
- School of Biological Sciences, Washington State University, Pullman, WA, United States
| | - Katherine Flores
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | | | - Lars Bode
- Department of Pediatrics, and Mother Milk Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA, United States
| | - Juan M. Rodríguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
28
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 401] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
29
|
Abstract
The incidence of celiac disease (CD) has increased over the last half-century, resulting in rising interest in identifying risk factors for CD. The necessity of duodenal biopsies in the diagnosis of CD has recently been challenged. Areas covered: This review covers the recent literature regarding the role of infant feeding practices, including breastfeeding and timing of gluten introduction, and the microbiota in the development of CD. Additionally, the application of the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for a non-biopsy approach to the diagnosis of CD is reviewed. Expert commentary: Recent investigations have not revealed any significant effect of breastfeeding or timing of gluten introduction on the risk of CD in at-risk populations. There are alterations in the microbiota of CD patients. However, the role of the microbiome and whether its manipulation has a clinical effect are unknown. Preliminary data suggests a non-biopsy approach to diagnosis of pediatric CD can be applied to several populations, although additional studies are needed. Prospective investigations are underway to examine the interplay of infant feeding practices and the microbiome and to identify particular CD-specific biomarkers that may aid in the diagnosis and ultimately prevention of CD.
Collapse
Affiliation(s)
- Grace J Lee
- a Division of Pediatric Gastroenterology, Department of Pediatrics and Communicable Diseases , C.S. Mott Children's Hospital, University of Michigan , Ann Arbor , MI , USA
| | - John Y Kao
- b Division of Gastroenterology, Department of Internal Medicine , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
30
|
Immune Components in Human Milk Are Associated with Early Infant Immunological Health Outcomes: A Prospective Three-Country Analysis. Nutrients 2017; 9:nu9060532. [PMID: 28538696 PMCID: PMC5490511 DOI: 10.3390/nu9060532] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 01/22/2023] Open
Abstract
The role of breastfeeding in improving allergy outcomes in early childhood is still unclear. Evidence suggests that immune mediators in human milk (HM) play a critical role in infant immune maturation as well as protection against atopy/allergy development. We investigated relationships between levels of immune mediators in colostrum and mature milk and infant outcomes in the first year of life. In a large prospective study of 398 pregnant/lactating women in the United Kingdom, Russia and Italy, colostrum and mature human milk (HM) samples were analysed for immune active molecules. Statistical analyses used models adjusting for the site of collection, colostrum collection time, parity and maternal atopic status. Preliminary univariate analysis showed detectable interleukin (IL) 2 and IL13 in HM to be associated with less eczema. This finding was further confirmed in multivariate analysis, with detectable HM IL13 showing protective effect OR 0.18 (95% CI 0.04-0.92). In contrast, a higher risk of eczema was associated with higher HM concentrations of transforming growth factor β (TGFβ) 2 OR 1.04 (95% CI 1.01-1.06) per ng/mL. Parental-reported food allergy was reported less often when IL13 was detectable in colostrum OR 0.10 (95% CI 0.01-0.83). HM hepatocyte growth factor (HGF) was protective for common cold incidence at 12 months OR 0.19 (95% CI 0.04-0.92) per ng/mL. Data from this study suggests that differences in the individual immune composition of HM may have an influence on early life infant health outcomes. Increased TGFβ2 levels in HM are associated with a higher incidence of reported eczema, with detectable IL13 in colostrum showing protective effects for food allergy and sensitization. HGF shows some protective effect on common cold incidence at one year of age. Future studies should be focused on maternal genotype, human milk microbiome and diet influence on human milk immune composition and both short- and long-term health outcomes in the infant.
Collapse
|
31
|
Rekima A, Macchiaverni P, Turfkruyer M, Holvoet S, Dupuis L, Baiz N, Annesi-Maesano I, Mercenier A, Nutten S, Verhasselt V. Long-term reduction in food allergy susceptibility in mice by combining breastfeeding-induced tolerance and TGF-β-enriched formula after weaning. Clin Exp Allergy 2017; 47:565-576. [PMID: 27883236 DOI: 10.1111/cea.12864] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Oral tolerance induction in early life is a promising approach for food allergy prevention. Its success requires the identification of factors necessary for its persistence. OBJECTIVES We aimed to assess in mice duration of allergy prevention by breastfeeding-induced oral tolerance and whether oral TGF-β supplementation after weaning would prolong it. METHODS We quantified ovalbumin (OVA) and OVA-specific immunoglobulin levels by ELISA in milk from the EDEN birth cohort. As OVA-specific Ig was found in all samples, we assessed whether OVA-immunized mice exposed to OVA during lactation could prevent allergic diarrhoea in their 6- and 13-week-old progeny. In some experiments, a TGF-β-enriched formula was given after weaning. RESULTS At 6 weeks, only 13% and 34% of mice breastfed by OVA-exposed mothers exhibited diarrhoea after six and seven OVA challenges vs. 44% and 72% in mice breastfed by naïve mothers (P = 0.02 and 0.01). Protection was associated with decreased levels of MMCP1 and OVA-specific IgE (P < 0.0001). At 13 weeks, although OVA-specific IgE remained low (P = 0.001), diarrhoea occurrence increased to 32% and 46% after six and seven OVA challenges in mice breastfed by OVA-exposed mothers. MMCP1 levels were not significantly inhibited. Supplementation with TGF-β after weaning induced a strong protection in 13-week-old mice breastfed by OVA-exposed mothers compared with mice breastfed by naive mothers (0%, 13% and 32% of diarrhoea at the fifth, sixth and seventh challenges vs. 17, 42 and 78%; P = 0.05, 0.0043 and 0.0017). MMCP1 levels decreased by half compared with control mice (P = 0.02). Prolonged protection was only observed in mice rendered tolerant by breastfeeding and was associated with an improved gut barrier. CONCLUSIONS In mice, prevention of food allergy by breastfeeding-induced tolerance is of limited duration. Nutritional intervention by TGF-β supplementation after weaning could prolong beneficial effects of breast milk on food allergy prevention.
Collapse
Affiliation(s)
- A Rekima
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France
| | - P Macchiaverni
- Institute of Biomedical Sciences - University of São Paulo, São Paulo, Brazil
| | - M Turfkruyer
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France
| | - S Holvoet
- Nestle Research Center, Lausanne, Switzerland
| | - L Dupuis
- Nestle Research Center, Lausanne, Switzerland
| | - N Baiz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | - I Annesi-Maesano
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | - A Mercenier
- Nestle Research Center, Lausanne, Switzerland
| | - S Nutten
- Nestle Research Center, Lausanne, Switzerland
| | - V Verhasselt
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France.,The International Inflammation 'in-FLAME' Network, Worldwide Universities Network, Perth, Australia
| |
Collapse
|
32
|
Lima-Ojeda JM, Rupprecht R, Baghai TC. "I Am I and My Bacterial Circumstances": Linking Gut Microbiome, Neurodevelopment, and Depression. Front Psychiatry 2017; 8:153. [PMID: 28878696 PMCID: PMC5572414 DOI: 10.3389/fpsyt.2017.00153] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/02/2017] [Indexed: 01/01/2023] Open
Abstract
Recently, there has been renewed interest in the role played by microbiome in both human health and human disease. A correct equilibrium between the human host and their microorganisms is important for an appropriate physiological function. Extensive research has shown that microbes that inhabit the gastrointestinal tract-or gut microbiota-are involved not only in both nutritive and digestive activities but also in immunological processes. Moreover, the gut microbiome influences both central nervous system and energy homeostasis. An altered gut microbiome has been associated with the pathophysiology of different diseases, including neuropsychiatric disorders. Apparently, both environmental-diet, exposition to antibiotics, and infections-and host-genetic factors have a strong influence on gut microbiome, modulating the risk for neuropsychiatric illness. Also, early life disruption of the microbiome-gut-brain (MGB) axis has been associated with an increased risk of developing depression later in life, suggesting a link between gut microbiome, neurodevelopment, and depression. This review aims to contribute to this growing area of research by exploring the role played by the gut microbiome in neurodevelopment and in the etiology of the depressive syndrome, including nutritional, immunological, and energy homeostasis approaches.
Collapse
Affiliation(s)
- Juan M Lima-Ojeda
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Thomas C Baghai
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
33
|
Carlier FM, Sibille Y, Pilette C. The epithelial barrier and immunoglobulin A system in allergy. Clin Exp Allergy 2016; 46:1372-1388. [PMID: 27684559 DOI: 10.1111/cea.12830] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway and intestinal epithelial layers represent first-line physical barriers, playing a key role in mucosal immunity. Barrier dysfunction, characterized by alterations such as disruption of cell-cell apical junctions and aberrant epithelial responses, probably constitutes early and key events for chronic immune responses to environmental antigens in the skin and in the gut. For instance, barrier dysfunction drives Th2 responses in atopic disorders or eosinophilic esophagitis. Such epithelial impairment is also a salient feature of allergic asthma and growing evidence indicates that barrier alterations probably play a driving role in this disease. IgA has been identified as the most abundant immunoglobulin in mucosa, where it acts as an active barrier through immune exclusion of inhaled or ingested antigens or pathogens. Historically, it has been thought to represent the serum factor underlying reaginic activity before IgE was discovered. Despite several studies about regulation and major functions of IgA at mucosal surfaces, its role in allergy remains largely unclear. This review aims at summarizing findings about epithelial functions and IgA biology that are relevant to allergy, and to integrate the emerging concepts and the recent developments in mucosal immunology, and how these could translate to clinical observations in allergy.
Collapse
Affiliation(s)
- F M Carlier
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium.
| | - Y Sibille
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium
| | - C Pilette
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.,Walloon Excellence in Lifesciences and Biotechnology, Wavre, Belgium
| |
Collapse
|
34
|
Baïz N, Macchiaverni P, Tulic MK, Rekima A, Annesi-Maesano I, Verhasselt V. Early oral exposure to house dust mite allergen through breast milk: A potential risk factor for allergic sensitization and respiratory allergies in children. J Allergy Clin Immunol 2016; 139:369-372.e10. [PMID: 27566456 DOI: 10.1016/j.jaci.2016.07.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/20/2016] [Accepted: 07/04/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Nour Baïz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | | | - Meri K Tulic
- EA 6302 University Nice Sophia Antipolis, TIM, Nice, France; International Inflammation 'in-FLAME' Network, Worldwide Universities Network, Perth, Australia
| | - Akila Rekima
- EA 6302 University Nice Sophia Antipolis, TIM, Nice, France
| | - Isabella Annesi-Maesano
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | - Valérie Verhasselt
- EA 6302 University Nice Sophia Antipolis, TIM, Nice, France; International Inflammation 'in-FLAME' Network, Worldwide Universities Network, Perth, Australia.
| | | |
Collapse
|
35
|
Verhasselt V. Is infant immunization by breastfeeding possible? Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0139. [PMID: 25964452 DOI: 10.1098/rstb.2014.0139] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Breastfeeding is known as the most efficient way to prevent infectious disease in early life. Maternal anti-microbial immunoglobulins transfer through milk confers passive immunity to the breastfed child while his immune system is maturing. Maternal milk also contains bioactive factors that will stimulate this maturation. From the literature on breastfeeding prevention of immune-mediated disease and more specifically from our experiments conducted in the field of allergic disease prevention, we propose that breastfeeding may also induce antigen-specific immune responses in the breastfed child. We found that early oral antigen exposure through breast milk leads to tolerance or immune priming depending on the nature of the antigen transferred and accompanying maternal milk cofactors. Here, we will discuss our data in the light of prevention of infectious disease and will propose that possible milk transfer of microbial antigen could affect actively the immune response in breastfed children and thereby their long-term susceptibility to infectious disease. Further research in this direction may lead to novel strategies of early life vaccination, taking advantage of the possibility to stimulate antigen-specific immune responses through breast milk.
Collapse
Affiliation(s)
- Valerie Verhasselt
- EA 6302 University Nice Sophia Antipolis, Tolerance Immunitaire Team (TIM), Hôpital de l'Archet, 1-Route Saint Antoine de Ginestière, BP3079, 06202 Nice Cedex 03, France
| |
Collapse
|
36
|
Hua MC, Chen CC, Yao TC, Tsai MH, Liao SL, Lai SH, Chiu CY, Yeh KW, Huang JL. Role of Maternal Allergy on Immune Markers in Colostrum and Secretory Immunoglobulin A in Stools of Breastfed Infants. J Hum Lact 2016; 32:160-7. [PMID: 26243755 DOI: 10.1177/0890334415598783] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 07/07/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Although protection against infectious diseases has been observed among breastfed infants as compared to formula-fed infants, possible benefits of breastfeeding by allergic mothers for allergy prevention remain controversial. OBJECTIVES The aims of this study were to determine whether maternal allergy would influence immune markers (secretory immunoglobulin A [sIgA], interleukin-8 [IL-8], soluble CD14 [sCD14]) in colostrum and the associations between maternal allergy and fecal sIgA levels in breastfed infants. METHODS Study subjects were enrolled from the Prediction of Allergies in Taiwanese Children (PATCH) birth cohort study. Colostrum samples were obtained from 98 lactating mothers. Stool samples were collected from 108 infants within 5 days after birth and at 2 and 4 months of age. We compared concentrations of sIgA, IL-8, and sCD14 in colostrum between mothers with and without a history of allergic disease and allergic sensitization. We also compared fecal sIgA levels between breastfed and formula-fed infants and between infants with allergic and nonallergic mothers. RESULTS The sIgA concentrations were significantly higher in colostrum from allergic mothers than from nonallergic mothers (P = .01) and from allergic mothers who were immunoglobulin E (IgE) sensitized compared to nonallergic mothers who were not IgE sensitized (P = .023). Breastfed infants had significantly higher fecal sIgA levels as compared to formula-fed infants, regardless of whether their lactating mothers had an allergy (P < .05). CONCLUSION We found that breastfeeding is associated with increased infants' fecal sIgA levels and may have potential protective effects to the infants during the first 4 months of life, regardless of whether their lactating mothers have allergies.
Collapse
Affiliation(s)
- Man-Chin Hua
- Department of Pediatrics, Chang Gung Memorial Hospital, Keelung, Taiwan Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Chang Chen
- Chang Gung University College of Medicine, Taoyuan, Taiwan Division of Gastroenterology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tsung-Chieh Yao
- Chang Gung University College of Medicine, Taoyuan, Taiwan Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ming-Han Tsai
- Department of Pediatrics, Chang Gung Memorial Hospital, Keelung, Taiwan Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Sui-Ling Liao
- Department of Pediatrics, Chang Gung Memorial Hospital, Keelung, Taiwan Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shen-Hao Lai
- Chang Gung University College of Medicine, Taoyuan, Taiwan Division of Chest, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Yung Chiu
- Department of Pediatrics, Chang Gung Memorial Hospital, Keelung, Taiwan Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Kuo-Wei Yeh
- Chang Gung University College of Medicine, Taoyuan, Taiwan Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jing-Long Huang
- Chang Gung University College of Medicine, Taoyuan, Taiwan Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW This article aims to review the evidence that breast milk can actively shape neonate gut immune system development toward a mature immune system capable of responding appropriately to encountered antigens. RECENT FINDINGS Recent findings in the adult have demonstrated the critical role of the interaction between diet, gut microbiota, gut epithelial cells and gut-associated lymphoid tissue in the development of immune responses. Here, we will review what is known in this field in the neonate, compare these data to those obtained in the adult and review how milk factors impact gut immune function in the short and long term. SUMMARY We propose that the neonate immune system and maternal milk represent an entity necessary to ensure not only appropriate function in early life but also long term immune homeostasis.
Collapse
|
38
|
Kim DC, Seo AD, Yang SI, Lee HR, Lee SY. Breastfeeding increases the risk of food sensitization but not affect food allergy symptoms in young children with atopic dermatitis. ALLERGY ASTHMA & RESPIRATORY DISEASE 2016. [DOI: 10.4168/aard.2016.4.3.188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Dong Chan Kim
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - An Deok Seo
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Song I Yang
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Hae Ran Lee
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| |
Collapse
|
39
|
TGF-β-mediated airway tolerance to allergens induced by peptide-based immunomodulatory mucosal vaccination. Mucosal Immunol 2015; 8:1248-61. [PMID: 25783968 DOI: 10.1038/mi.2015.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 01/30/2015] [Indexed: 02/07/2023]
Abstract
We sought to modulate mucosal immune responses using neonatal vaccination to avert the development of allergic airways disease (AAD). Pulmonary pathology in AAD is driven by T helper (TH)2 cytokines, in particular interleukin (IL)4 and IL13, the expression and actions of which are regulated by the transcription factor STAT6. We developed a peptide homolog of STAT6, STAT6-IP. Neonatal mice given, intranasally, STAT6-IP, in an effort to modulate de novo airways immune responses, developed tolerance following subsequent allergen sensitization, with either ovalbumin or ragweed allergens, as demonstrated by reduced TH2 cytokines and specific immunoglobulin (Ig)E and the significant increases in the latency-associated peptide (LAP)(+) T-regulatory (Treg) cell subset and expression of transforming growth factor (TGF)-β. This regulatory phenotype was transferrable by CD4(+) T cells or CD11c(+) dendritic cells (DCs) derived from STAT6-IP-vaccinated mice. Anti-TGF-β treatment during allergen sensitization, however, re-established the pro-inflammatory TH2 response. Thus, neonatal STAT6-IP vaccination induces prospective TGF-β-dependent tolerance to allergen and constitutes a novel highly effective immunomodulatory allergy prevention strategy.
Collapse
|
40
|
Shen N, Clemente JC. Engineering the Microbiome: a Novel Approach to Immunotherapy for Allergic and Immune Diseases. Curr Allergy Asthma Rep 2015; 15:39. [PMID: 26143390 DOI: 10.1007/s11882-015-0538-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The incidence of immune disorders is growing parallel with practices associated with westernization, such as dietary changes, increased use of antibiotics, or elevated rates of Cesarean section. These practices can significantly impact the gut microbiota, the collection of bacteria residing in the human gastrointestinal tract, and subsequently disrupt the delicate balance existing between commensal flora and host immune responses. Restoring this balance by modifying the microbiota has thus emerged as a promising therapeutic approach. Here, we discuss the interaction between gut commensals and immunity, along with the potential of different interventions on the microbiota as treatment for inflammatory and allergic diseases.
Collapse
Affiliation(s)
- Nan Shen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | |
Collapse
|
41
|
Walker M. Why Infant Formula Samples Pose a Risk to Health Care Providers, Hospitals, and Patients. J Obstet Gynecol Neonatal Nurs 2015; 44:618-23. [DOI: 10.1111/1552-6909.12733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
42
|
Abstract
The incidence of allergic diseases is increasing, both in developed and developing countries, concomitantly with the rise in living standards and the adoption of a 'western lifestyle'. For two decades, the hygiene hypothesis - which proposes that the lack of early childhood exposure to infectious agents increases susceptibility to allergic diseases in later life - provided the conceptual framework for unravelling the mechanisms that could account for the increased incidence of allergic diseases. In this Review, we discuss recent evidence that highlights the role of diet as a key factor influencing immune homeostasis and the development of allergic diseases through a complex interplay between nutrients, their metabolites and immune cell populations. Although further investigations are still required to understand these complex relationships, recent data have established a possible connection between metabolic homeostasis and allergic diseases.
Collapse
|
43
|
The Interaction among Microbiota, Immunity, and Genetic and Dietary Factors Is the Condicio Sine Qua Non Celiac Disease Can Develop. J Immunol Res 2015; 2015:123653. [PMID: 26090475 PMCID: PMC4451297 DOI: 10.1155/2015/123653] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/14/2014] [Indexed: 12/24/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy, triggered by dietary wheat gluten and similar proteins of barley and rye in genetically susceptible individuals. This is a complex disorder involving both environmental and immune-genetic factors. The major genetic risk factor for CD is determined by HLA-DQ genes. Dysfunction of the innate and adaptive immune systems can conceivably cause impairment of mucosal barrier function and development of localized or systemic inflammatory and autoimmune processes. Exposure to gluten is the main environmental trigger responsible for the signs and symptoms of the disease, but exposure to gluten does not fully explain the manifestation of CD. Thus, both genetic determination and environmental exposure to gluten are necessary for the full manifestation of CD; neither of them is sufficient alone. Epidemiological and clinical data suggest that other environmental factors, including infections, alterations in the intestinal microbiota composition, and early feeding practices, might also play a role in disease development. Thus, this interaction is the condicio sine qua non celiac disease can develop. The breakdown of the interaction among microbiota, innate immunity, and genetic and dietary factors leads to disruption of homeostasis and inflammation; and tissue damage occurs. Focusing attention on this interaction and its breakdown may allow a better understanding of the CD pathogenesis and lead to novel translational avenues for preventing and treating this widespread disease.
Collapse
|
44
|
Koning N, Kessen SFM, Van Der Voorn JP, Appelmelk BJ, Jeurink PV, Knippels LMJ, Garssen J, Van Kooyk Y. Human Milk Blocks DC-SIGN-Pathogen Interaction via MUC1. Front Immunol 2015; 6:112. [PMID: 25821450 PMCID: PMC4358221 DOI: 10.3389/fimmu.2015.00112] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 02/26/2015] [Indexed: 12/23/2022] Open
Abstract
Beneficial effects of breastfeeding are well-recognized and include both immediate neonatal protection against pathogens and long-term protection against allergies and autoimmune diseases. Although several proteins have been identified to have anti-viral or anti-bacterial effects like secretory IgA or lactoferrin, the mechanisms of immune modulation are not fully understood. Recent studies identified important beneficial effects of glycans in human milk, such as those expressed in oligosaccharides or on glycoproteins. Glycans are recognized by the carbohydrate receptors C-type lectins on dendritic cell (DC) and specific tissue macrophages, which exert important functions in immune modulation and immune homeostasis. A well-characterized C-type lectin is dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), which binds terminal fucose. The present study shows that in human milk, MUC1 is the major milk glycoprotein that binds to the lectin domain of DC-SIGN and prevents pathogen interaction through the presence of Lewis x-type oligosaccharides. Surprisingly, this was specific for human milk, as formula, bovine or camel milk did not show any presence of proteins that interacted with DC-SIGN. The expression of DC-SIGN is found in young infants along the entire gastrointestinal tract. Our data thus suggest the importance of human milk glycoproteins for blocking pathogen interaction to DC in young children. Moreover, a potential benefit of human milk later in life in shaping the infants immune system through DC-SIGN cannot be ruled out.
Collapse
Affiliation(s)
- Nathalie Koning
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Sabine F M Kessen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | | | - Ben J Appelmelk
- Department of Medical Microbiology and Infection Control, VU University Medical Center , Amsterdam , Netherlands
| | - Prescilla V Jeurink
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Leon M J Knippels
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Johan Garssen
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Yvette Van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| |
Collapse
|
45
|
Effect of a protein-free diet in the development of food allergy and oral tolerance in BALB/c mice. Br J Nutr 2015; 113:935-43. [PMID: 25759975 DOI: 10.1017/s0007114515000173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to investigate the effect of a protein-free diet in the induction of food allergy and oral tolerance in BALB/c mice. The experimental model used was mice that were fed, since weaning up to adulthood, a balanced diet in which all dietary proteins were replaced by amino acid diet (Aa). The absence of dietary proteins did not prevent the development of food allergy to ovalbumin (OVA) in these mice. However, Aa-fed mice produced lower levels of IgE, secretory IgA and cytokines. In addition, when compared with mice from control group, Aa-fed mice had a milder aversive reaction to the allergen measured by consumption of OVA-containing solution and weight loss during food allergy development. In addition, mice that did not have dietary proteins in their diets were less susceptible to induction of oral tolerance. One single oral administration was not enough to suppress specific serum Ig and IgG1 levels in the Aa-fed group, although it was efficient to induce suppression in the control group. The present results indicate that the stimulation by dietary proteins alters both inflammatory reactivity and regulatory immune reactivity in mice probably due to their effect in the maturation of the immune system.
Collapse
|
46
|
Savilahti EM, Kukkonen AK, Kuitunen M, Savilahti E. Soluble CD14, α-and β-defensins in breast milk: association with the emergence of allergy in a high-risk population. Innate Immun 2014; 21:332-7. [PMID: 25432966 DOI: 10.1177/1753425914541560] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As innate immunity factors in breast milk (BM) modulate infants' immune responses, we investigated whether soluble CD14 (sCD14) and defensin levels in BM are associated with the emergence of allergy in childhood. The randomly selected group of 260 mother-child pairs belonged to a randomized, double-blind placebo-controlled trial where 1223 mothers with fetuses at high risk for allergy received for the 4 last wk of pregnancy a mixture of probiotics, or placebo; after birth, the child received the treatment for 6 mo. Children were followed for the emergence of sensitization and allergic symptoms for 5 yr. IgE-mediated allergic disorder was diagnosed in 80 children by the age of 5 yr. Levels of sCD14, human neutrophil peptide (HNP) 1-3 and β-defensin 2 (HBD2) in colostrum and in BM 3 mo post-partum were measured with ELISA. BM sCD14 levels decreased from 0 to 3 mo. HNP1-3 and HBD2 were detected in colostrum, but not in BM 3 mo post-partum. High sCD14 levels in BM 3 mo post-partum were associated with children developing an IgE-mediated allergic disorder by the age of 5 yr. BM HNP1-3, HBD2 or sCD14 levels were not associated with probiotics treatment. Our results suggest that sCD14 in BM influences the emergence of allergy in children with atopic heredity.
Collapse
Affiliation(s)
- Emma M Savilahti
- The Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Anna K Kukkonen
- The Skin and Allergy Hospital, University of Helsinki, Helsinki, Finland
| | - Mikael Kuitunen
- The Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Erkki Savilahti
- The Children's Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Biesbroek G, Bosch AATM, Wang X, Keijser BJF, Veenhoven RH, Sanders EAM, Bogaert D. The impact of breastfeeding on nasopharyngeal microbial communities in infants. Am J Respir Crit Care Med 2014; 190:298-308. [PMID: 24921688 DOI: 10.1164/rccm.201401-0073oc] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Breastfeeding elicits significant protection against respiratory tract infections in infancy. Modulation of respiratory microbiota might be part of the natural mechanisms of protection against respiratory diseases induced by breastfeeding. OBJECTIVES To study the association between breastfeeding and nasopharyngeal microbial communities, including all cultivable and noncultivable bacteria. METHODS In this observational study, we analyzed the microbiota of infants that had received exclusive breastfeeding (n = 101) and exclusive formula feeding (n = 101) at age 6 weeks and 6 months by 16S-based GS-FLX-titanium-pyrosequencing. MEASUREMENTS AND MAIN RESULTS At 6 weeks of age the overall bacterial community composition was significantly different between breastfed and formula-fed children (nonmetric multidimensional scaling, P = 0.001). Breastfed children showed increased presence and abundance of the lactic acid bacterium Dolosigranulum (relative effect size [RES], 2.61; P = 0.005) and Corynebacterium (RES, 1.98; P = 0.039) and decreased abundance of Staphylococcus (RES, 0.48; P 0.03) and anaerobic bacteria, such as Prevotella (RES, 0.25; P < 0.001) and Veillonella (RES, 0.33; P < 0.001). Predominance (>50% of the microbial profile) of Corynebacterium and Dolosigranulum was observed in 45 (44.6%) breastfed infants compared with 19 (18.8%) formula-fed infants (relative risk, 2.37; P = 0.006). Dolosigranulum abundance was inversely associated with consecutive symptoms of wheezing and number of mild respiratory tract infections experienced. At 6 months of age associations between breastfeeding and nasopharyngeal microbiota composition had disappeared. CONCLUSIONS Our data suggest a strong association between breastfeeding and microbial community composition in the upper respiratory tract of 6-week-old infants. Observed differences in microbial community profile may contribute to the protective effect of breastfeeding on respiratory infections and wheezing in early infancy. Clinical trial registered with www.clinicaltrials.gov (NCT 00189020).
Collapse
Affiliation(s)
- Giske Biesbroek
- 1 Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Orivuori L, Mustonen K, Roduit C, Braun-Fahrländer C, Dalphin JC, Genuneit J, Lauener R, Pfefferle P, Riedler J, Weber J, von Mutius E, Pekkanen J, Vaarala O. Immunoglobulin A and immunoglobulin G antibodies against β-lactoglobulin and gliadin at age 1 associate with immunoglobulin E sensitization at age 6. Pediatr Allergy Immunol 2014; 25:329-37. [PMID: 24953294 DOI: 10.1111/pai.12246] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Serum immunoglobulin A (IgA) and immunoglobulin G (IgG) antibodies against wheat gliadin and cow's milk β-lactoglobulin (BLG) are considered markers of gut permeability and inflammation which modulate the development of mucosal tolerance. Living on a farm has been shown to decrease allergies in children. Our aim was to study whether farm environment affected mucosal tolerance, immunoglobulin E (IgE) sensitization, or allergic diseases. METHODS The PASTURE birth cohort study was conducted in Finland, France, Germany, Austria, and Switzerland. At age 1, we measured serum IgA and IgG against wheat gliadin (N = 636) and cow's milk BLG (N = 639) using ELISA. Serum-specific IgE was measured at ages 1 and 6 (N = 459). Data on environmental factors and allergic diseases were collected by questionnaires. Discrete time hazard and multivariate logistic regression models were used for analyses. RESULTS Increased IgA or IgG antibodies against BLG at age 1 increased the risk of sensitization to at least one of the measured allergens or food allergens at age 6. Increased IgG against gliadin at age 1 increased the risk of sensitization to any, at least one inhalant, or at least one food allergen at age 6. Early exposure to cow's milk formula associated with increased IgA or IgG against BLG. No association with farming or clinical allergy was found. CONCLUSIONS Increased IgA or IgG against BLG or gliadin at age 1 was associated with IgE sensitization at age 6. We suggest that an enhanced antibody response to food antigens reflects mucosal tolerance aberrancies, e.g., altered microbiota and/or increased gut permeability, which is later seen as sensitization to allergens.
Collapse
Affiliation(s)
- Laura Orivuori
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
van Neerven R. The effects of milk and colostrum on allergy and infection: Mechanisms and implications. Anim Front 2014. [DOI: 10.2527/af.2014-0010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- R.J.J. van Neerven
- FrieslandCampina, Amersfoort, The Netherlands
- Cell Biology and Immunology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
50
|
Quality indicators for human milk use in very low-birthweight infants: are we measuring what we should be measuring? J Perinatol 2014; 34:287-91. [PMID: 24526005 PMCID: PMC3969767 DOI: 10.1038/jp.2014.5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 12/26/2013] [Accepted: 01/06/2014] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The objective of this study was to compare the currently used human milk (HM) quality indicators that measure whether very low-birthweight (VLBW; <1500 g birthweight) infants 'ever' received HM and whether they were still receiving HM at discharge from the neonatal intensive care unit (NICU) to the actual amount and timing of HM received. STUDY DESIGN This study used data from a large NIH-funded cohort study and calculated whether VLBW infants ever received HM (HM-Ever) and of these infants, the percentage who were still receiving HM at NICU discharge (HM-DC). Then, the HM-DC indicator (exclusive, partial and none) was compared with the amount and timing of HM feedings received by these same infants. RESULT Of the 291 VLBW infants who met inclusion criteria, 285 received some HM (HM-Ever=98%). At NICU discharge (HM-DC), 24.2, 15.1 and 60.7% were receiving exclusive, partial and no HM, respectively. Of the 60.7% infants with no HM-DC, some had received higher amounts of HM during the NICU hospitalization than infants categorized as exclusive and partial for HM-DC. Of the infants with no HM-DC, 76.8 and 59.7% had received exclusive HM during the days 1-14 and days 1-28 exposure periods, respectively. CONCLUSION The average daily dose (HM-DD; in ml kg(-1) d(-1)) and cumulative percentage (HM-PCT; as % of cumulative enteral intake) of HM feedings were sufficient to significantly reduce the risk of multiple morbidities, including late-onset sepsis, necrotizing enterocolitis, neurocognitive delay and rehospitalization, in the majority of the VLBW infants who were discharged with no HM-DC. Quality indicators that focus on the amount and timing of HM feedings in the NICU should be added to the HM-Ever and HM-DC measures.
Collapse
|