1
|
Rahman RS, Wesemann DR. Whence and wherefore IgE? Immunol Rev 2024; 326:48-65. [PMID: 39041740 PMCID: PMC11436312 DOI: 10.1111/imr.13373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Despite the near ubiquitous presence of Ig-based antibodies in vertebrates, IgE is unique to mammals. How and why it emerged remains mysterious. IgE expression is greatly constrained compared to other IgH isotypes. While other IgH isotypes are relatively abundant, soluble IgE has a truncated half-life, and IgE plasma cells are mostly short-lived. Despite its rarity, IgE is consequential and can trigger life-threatening anaphylaxis. IgE production reflects a dynamic steady state with IgG memory B cells feeding short-lived IgE production. Emerging evidence suggests that IgE may also potentially be produced in longer-lived plasma cells as well, perhaps as an aberrancy stemming from its evolutionary roots from an antibody isotype that likely functioned more like IgG. As a late derivative of an ancient systemic antibody system, the benefits of IgE in mammals likely stems from the antibody system's adaptive recognition and response capability. However, the tendency for massive, systemic, and long-lived production, common to IgH isotypes like IgG, were likely not a good fit for IgE. The evolutionary derivation of IgE from an antibody system that for millions of years was good at antigen de-sensitization to now functioning as a highly specialized antigen-sensitization function required heavy restrictions on antibody production-insufficiency of which may contribute to allergic disease.
Collapse
Affiliation(s)
- Rifat S Rahman
- Department of Internal Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Duane R Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Furiness KN, El Ansari YS, Oettgen HC, Kanagaratham C. Allergen-specific IgA and IgG antibodies as inhibitors of mast cell function in food allergy. FRONTIERS IN ALLERGY 2024; 5:1389669. [PMID: 38919913 PMCID: PMC11196826 DOI: 10.3389/falgy.2024.1389669] [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: 02/21/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Food allergy, a group of adverse immune responses to normally innocuous food protein antigens, is an increasingly prevalent public health issue. The most common form is IgE-mediated food allergy in which food antigen-induced crosslinking of the high-affinity IgE-receptor, FcεRI, on the surface of mast cells triggers the release of inflammatory mediators that contribute to a wide range of clinical manifestations, including systemic anaphylaxis. Mast cells also play a critical function in adaptive immunity to foods, acting as adjuvants for food-antigen driven Th2 cell responses. While the diagnosis and treatment of food allergy has improved in recent years, no curative treatments are currently available. However, there is emerging evidence to suggest that both allergen-specific IgA and IgG antibodies can counter the activating effects of IgE antibodies on mast cells. Most notably, both antigen-specific IgA and IgG antibodies are induced in the course of oral immunotherapy. In this review, we highlight the role of mast cells in food allergy, both as inducers of immediate hypersensitivity reactions and as adjuvants for type 2 adaptive immune responses. Furthermore, we summarize current understanding of the immunomodulatory effects of antigen-specific IgA and IgG antibodies on IgE-induced mast cell activation and effector function. A more comprehensive understanding of the regulatory role of IgA and IgG in food allergy may provide insights into physiologic regulation of immune responses to ingested antigens and could seed novel strategies to treat allergic disease.
Collapse
Affiliation(s)
- Kameryn N. Furiness
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Yasmeen S. El Ansari
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Cynthia Kanagaratham
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Filimonova I, Innocenti G, Vogl T. Phage Immunoprecipitation Sequencing (PhIP-Seq) for Analyzing Antibody Epitope Repertoires Against Food Antigens. Methods Mol Biol 2024; 2717:101-122. [PMID: 37737980 DOI: 10.1007/978-1-0716-3453-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
While thousands of food and environmental allergens have been reported, conventional methods for allergy testing typically rely on measuring immunoglobulin E (IgE) binding against panels of dozens to hundreds of antigens. Beyond IgE, also the specificity of other Ig (sub-)classes such as IgG4, has gained interest because of a potential protective role toward allergy.Phage immunoprecipitation sequencing (PhIP-Seq) allows to study hundreds of thousands of rationally selected peptide antigens and to resolve binding specificities of different Ig classes. This technology combines synthetic DNA libraries encoding antigens, with the display on the surface of T7 bacteriophages and next-generation sequencing (NGS) for quantitative readouts. Thereby binding of entire Ig repertoires can be measured to detect the exact epitopes of food allergens and to study potential cross-reactivity.In this chapter, we provide a summary of both the key experimental steps and various strategies for analyzing PhIP-Seq datasets, as well as comparing the advantages and disadvantages of this methodology for measuring antibody responses against food antigens.
Collapse
Affiliation(s)
- Ioanna Filimonova
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria
| | - Gabriel Innocenti
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria
| | - Thomas Vogl
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria.
| |
Collapse
|
4
|
Chen C, Sang Z, Xie Q, Xue W. Effects of hazelnut protein isolate-induced food allergy on the gut microenvironment in a BALB/c mouse model. Food Funct 2023; 14:8761-8774. [PMID: 37718731 DOI: 10.1039/d3fo02324a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Hazelnuts are reported as among the nuts that cause severe allergic reactions. However, few systematic studies exist on the changes in the gut microenvironment following hazelnut allergy. This study focused on the effects of hazelnut allergy on the duodenum, jejunum, ileum and colon microenvironment in vivo. We established a hazelnut protein isolate (HPI)-allergic mouse model, which was distinguished by the visible allergy symptoms, dropped temperatures and enhanced allergic inflammatory factor levels in serum, such as HPI-specific immunoglobulin E (sIgE), sIgG2a, interleukin-4, histamine, mouse mast cell protease-1, TNF-α, monocyte chemotactic protein-1 and lipopolysaccharide. For HPI sensitized mice, aggravated mast cell degranulation, severe morphologic damage and inflammatory cell infiltration were observed in the duodenum, jejunum, ileum, and colon, while goblet cell numbers were reduced in the duodenum, jejunum and ileum. Secretory IgA of the jejunum and tight junctions of the duodenum and jejunum were decreased significantly after HPI sensitization. There was no remarkable difference in the pH values of small intestinal contents, but the pH values of colonic contents were elevated, which was due to the decreased short-chain fatty acids (mainly acetate, propionate and butyrate) in the colon. The antioxidant capacity of both large and small intestinal contents declined after HPI sensitization, as evidenced by the increased malondialdehyde and decreased superoxide dismutase activity. HPI sensitization induced gut microbiota dysbiosis with decreased α diversity and altered β diversity in colonic contents. Spearman correlation analysis indicated that the increased characteristic genera, namely Bacteroides, Lactobacillus, Alloprevotella, Erysipelatoclostridium, Parabacteroides, and Helicobacter, played potentially synergistic roles in promoting allergy and gut microenvironment dysregulation.
Collapse
Affiliation(s)
- Chen Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Ziqing Sang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Qiang Xie
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Wentong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
5
|
Scheurer S, Junker AC, He C, Schülke S, Toda M. The Role of IgA in the Manifestation and Prevention of Allergic Immune Responses. Curr Allergy Asthma Rep 2023; 23:589-600. [PMID: 37610671 PMCID: PMC10506939 DOI: 10.1007/s11882-023-01105-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE OF REVIEW Immunoglobulin A (IgA) mediates immune exclusion of antigens in the gut. Notably, IgA plays also a role in the prevention of IgE-mediated allergies and induction of immune tolerance. The present review addresses the role of IgA in the manifestation of IgE-mediated allergies, including allergen-specific immunotherapy (AIT), the regulation of IgA production, and the mechanism of IgA in immune cell activation. RECENT FINDINGS The majority of studies report an association of IgA with the induction of immune tolerance in IgE-mediated allergies. However, reports on the involvement of humoral and mucosal IgA, IgA subtypes, monomeric and polymeric IgA, and the mechanism of IgA-mediated immune cell activation are confounding. Effects by IgA are likely mediated by alteration of microbiota, IgE-blocking capacity, or activation of inhibitory signaling pathways. However, the precise mechanism of IgA-regulation, the contribution of serum and/or mucosal IgA, and IgA1/2 subtypes, on the manifestation of IgE-mediated allergies, and the underlying immune modulatory mechanism are still elusive.
Collapse
Affiliation(s)
- Stephan Scheurer
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany.
| | - Ann-Christine Junker
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
| | - Chaoqi He
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Stefan Schülke
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
- Division of Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
6
|
Seppo AE, Jackson CM, Järvinen KM. The role of IgA in food allergy remains elusive-timing is everything? J Allergy Clin Immunol 2023; 151:1481-1483. [PMID: 36905964 DOI: 10.1016/j.jaci.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023]
Affiliation(s)
- Antti E Seppo
- Division of Pediatric Allergy and Immunology and Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Courtney M Jackson
- Division of Pediatric Allergy and Immunology and Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Kirsi M Järvinen
- Division of Pediatric Allergy and Immunology and Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
7
|
Ma B, Gavzy SJ, Saxena V, Song Y, Piao W, Lwin HW, Lakhan R, Iyyathurai J, Li L, France M, Paluskievicz C, Shirkey MW, Hittle L, Munawwar A, Mongodin EF, Bromberg JS. Strain-specific alterations in gut microbiome and host immune responses elicited by tolerogenic Bifidobacterium pseudolongum. Sci Rep 2023; 13:1023. [PMID: 36658194 PMCID: PMC9852428 DOI: 10.1038/s41598-023-27706-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
The beneficial effects attributed to Bifidobacterium are largely attributed to their immunomodulatory capabilities, which are likely to be species- and even strain-specific. However, their strain-specificity in direct and indirect immune modulation remain largely uncharacterized. We have shown that B. pseudolongum UMB-MBP-01, a murine isolate strain, is capable of suppressing inflammation and reducing fibrosis in vivo. To ascertain the mechanism driving this activity and to determine if it is specific to UMB-MBP-01, we compared it to a porcine tropic strain B. pseudolongum ATCC25526 using a combination of cell culture and in vivo experimentation and comparative genomics approaches. Despite many shared features, we demonstrate that these two strains possess distinct genetic repertoires in carbohydrate assimilation, differential activation signatures and cytokine responses signatures in innate immune cells, and differential effects on lymph node morphology with unique local and systemic leukocyte distribution. Importantly, the administration of each B. pseudolongum strain resulted in major divergence in the structure, composition, and function of gut microbiota. This was accompanied by markedly different changes in intestinal transcriptional activities, suggesting strain-specific modulation of the endogenous gut microbiota as a key to immune modulatory host responses. Our study demonstrated a single probiotic strain can influence local, regional, and systemic immunity through both innate and adaptive pathways in a strain-specific manner. It highlights the importance to investigate both the endogenous gut microbiome and the intestinal responses in response to probiotic supplementation, which underpins the mechanisms through which the probiotic strains drive the strain-specific effect to impact health outcomes.
Collapse
Affiliation(s)
- Bing Ma
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Samuel J Gavzy
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yang Song
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hnin Wai Lwin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ram Lakhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michael France
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christina Paluskievicz
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marina W Shirkey
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lauren Hittle
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Emmanuel F Mongodin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Lung Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jonathan S Bromberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
The Crosstalk between the Gut Microbiota Composition and the Clinical Course of Allergic Rhinitis: The Use of Probiotics, Prebiotics and Bacterial Lysates in the Treatment of Allergic Rhinitis. Nutrients 2022; 14:nu14204328. [PMID: 36297012 PMCID: PMC9607052 DOI: 10.3390/nu14204328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Although massive progress in discovering allergic rhinitis (AR) aetiology has been made in recent years, its prevalence is still rising and it significantly impacts patients' lives. That is why further and non-conventional research elucidating the role of new factors in AR pathogenesis is needed, facilitating discoveries of new treatment approaches. One of these factors is the gut microbiota, with its specific roles in health and disease. This review presents the process of gut microbiota development, especially in early life, focusing on its impact on the immune system. It emphasizes the link between the gut microbiota composition and immune changes involved in AR development. Specifically, it elucidates the significant link between bacteria colonizing the gut and the Th1/Th2 imbalance. Probiotics, prebiotics and bacterial lysates, which are medications that restore the composition of intestinal bacteria and indirectly affect the clinical course of AR, are also discussed.
Collapse
|
9
|
El Ansari YS, Kanagaratham C, Burton OT, Santos JV, Hollister BMA, Lewis OL, Renz H, Oettgen HC. Allergen-Specific IgA Antibodies Block IgE-Mediated Activation of Mast Cells and Basophils. Front Immunol 2022; 13:881655. [PMID: 35865546 PMCID: PMC9294179 DOI: 10.3389/fimmu.2022.881655] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mast cells and basophils have long been implicated in the pathogenesis of IgE-mediated hypersensitivity reactions. They express the high-affinity IgE receptor, FcϵRI, on their surface. Antigen-induced crosslinking of IgE antibodies bound to that receptor triggers a signaling cascade that results in activation, leading to the release of an array of preformed vasoactive mediators and rapidly synthesized lipids, as well as the de novo production of inflammatory cytokines. In addition to bearing activating receptors like FcεRI, these effector cells of allergy express inhibitory ones including FcγR2b, an IgG Fc receptor with a cytosolic inhibitory motif that activates protein tyrosine phosphatases that suppress IgE-mediated activation. We and others have shown that food allergen-specific IgG antibodies strongly induced during the course of oral immunotherapy (OIT), signal via FcγR2b to suppress IgE-mediated mast cell and basophil activation triggered by food allergen challenge. However, the potential inhibitory effects of IgA antibodies, which are also produced in response to OIT and are present at high levels at mucosal sites, including the intestine where food allergens are encountered, have not been well studied. Here we uncover an inhibitory function for IgA. We observe that IgA binds mouse bone marrow-derived mast cells (BMMCs) and peritoneal mast cells. Binding to BMMCs is dependent on calcium and sialic acid. We also found that IgA antibodies inhibit IgE-mediated mast cell degranulation in an allergen-specific fashion. Antigen-specific IgA inhibits IgE-mediated mast cell activation early in the signaling cascade, suppressing the phosphorylation of Syk, the proximal protein kinase mediating FcεRI signaling, and suppresses mast cell production of cytokines. Furthermore, using basophils from a peanut allergic donor we found that IgA binds to basophils and that activation by exposure to peanuts is effectively suppressed by IgA. We conclude that IgA serves as a regulator of mast cell and basophil degranulation, suggesting a physiologic role for IgA in the maintenance of immune homeostasis at mucosal sites.
Collapse
Affiliation(s)
- Yasmeen S. El Ansari
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Oliver T. Burton
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Jenna V. Santos
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | | | - Owen L. Lewis
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Harald Renz
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- *Correspondence: Hans C. Oettgen,
| |
Collapse
|
10
|
Abstract
Secretory immunoglobulin A (SIgA) in human milk plays a central role in complex maternal-infant interactions that influence long-term health outcomes. Governed by genetics and maternal microbial exposure, human milk SIgA shapes both the microbiota and immune system of infants. Historically, SIgA-microbe interactions have been challenging to unravel due to their dynamic and personalized nature, particularly during early life. Recent advances have helped to clarify how SIgA acts beyond simple pathogen clearance to help guide and constrain a healthy microbiota, promote tolerance, and influence immune system development. In this review, we highlight these new findings in the context of the critical early-life window and propose outstanding areas of study that will be key to harnessing the benefits of SIgA to support healthy immune development during infancy.
Collapse
|
11
|
Sun Y, Xie R, Li L, Jin G, Zhou B, Huang H, Li M, Yang Y, Liu X, Cao X, Wang B, Liu W, Jiang K, Cao H. Prenatal Maternal Stress Exacerbates Experimental Colitis of Offspring in Adulthood. Front Immunol 2021; 12:700995. [PMID: 34804005 PMCID: PMC8595204 DOI: 10.3389/fimmu.2021.700995] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/13/2021] [Indexed: 01/14/2023] Open
Abstract
The prevalence of inflammatory bowel disease (IBD) is increasing worldwide and correlates with dysregulated immune response because of gut microbiota dysbiosis. Some adverse early life events influence the establishment of the gut microbiota and act as risk factors for IBD. Prenatal maternal stress (PNMS) induces gut dysbiosis and perturbs the neuroimmune network of offspring. In this study, we aimed to investigate whether PNMS increases the susceptibility of offspring to colitis in adulthood. The related index was assessed during the weaning period and adulthood. We found that PNMS impaired the intestinal epithelial cell proliferation, goblet cell and Paneth cell differentiation, and mucosal barrier function in 3-week-old offspring. PNMS induced low-grade intestinal inflammation, but no signs of microscopic inflammatory changes were observed. Although there was no pronounced difference between the PNMS and control offspring in terms of their overall measures of alpha diversity for the gut microbiota, distinct microbial community changes characterized by increases in Desulfovibrio, Streptococcus, and Enterococcus and decreases in Bifidobacterium and Blautia were induced in the 3-week-old PNMS offspring. Notably, the overgrowth of Desulfovibrio persisted from the weaning period to adulthood, consistent with the results observed using fluorescence in situ hybridization in the colon mucosa. Mechanistically, the fecal microbiota transplantation experiment showed that the gut microbiota from the PNMS group impaired the intestinal barrier function and induced low-grade inflammation. The fecal bacterial solution from the PNMS group was more potent than that from the control group in inducing inflammation and gut barrier disruption in CaCo-2 cells. After treatment with a TNF-α inhibitor (adalimumab), no statistical difference in the indicators of inflammation and intestinal barrier function was observed between the two groups. Finally, exposure to PNMS remarkably increased the values of the histopathological parameters and the inflammatory cytokine production in a mouse model of experimental colitis in adulthood. These findings suggest that PNMS can inhibit intestinal development, impair the barrier function, and cause gut dysbiosis characterized by the persistent overgrowth of Desulfovibrio in the offspring, resulting in exacerbated experimental colitis in adulthood.
Collapse
Affiliation(s)
- Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Lu Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bingqian Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Huan Huang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yunwei Yang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
12
|
Ley D, Beghin L, Morcel J, Flamein F, Garabedian C, Accart B, Drumez E, Labreuche J, Gottrand F, Hermann E. Impact of early life nutrition on gut health in children: a prospective clinical study. BMJ Open 2021; 11:e050432. [PMID: 34489289 PMCID: PMC8422494 DOI: 10.1136/bmjopen-2021-050432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/03/2021] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION The first 1000 days of life could contribute to individual susceptibility to the later development of chronic non-communicable diseases. Nutrition in early life appears to be an important determinant factor for a sustainable child's health. In this study, we propose to investigate the impact of exclusive breast feeding on gut health in children. METHODS AND ANALYSIS A prospective cohort of newborns (n=350) will be recruited at birth and followed up to 4 years of age. The main objective is to evaluate the link between exclusive breast feeding for at least 3 months and the gut health of the child at 4 years. The primary endpoint of assessment of gut health will be based on the non-invasive measurement of faecal secretory IgA (sIgA) as a sensitive biomarker of the intestinal ecosystem. The presence of gastrointestinal disorders will be defined according to the clinical criteria of Rome IV. Information on parent's nutritional habits and life style, breastfeeding duration and child's complementary feeding will be collected along the follow-up. Cord blood cells and plasma at birth will be purified for further analysis. The meconium and stools collected at birth, 6 months, 2 years and 4 years of age will allow sIgA analysis. ETHICS AND DISSEMINATION This clinical study has obtained the approval from the national ethical committee. We plan to publish the results of the study in peer-review journals and by means of national and international conference. TRIAL REGISTRATION NUMBER NCT04195425.
Collapse
Affiliation(s)
- Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Florence Flamein
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Charles Garabedian
- Univ. Lille, CHU Lille, Department of Obstetrics & Gynecology, F-59000 Lille, France
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
| | | | - Elodie Drumez
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
- CHU Lille, Department of Biostatistics, F-59000 Lille, France
| | - Julien Labreuche
- Univ. Lille, CHU Lille, ULR 2694-METRICS: évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
- CHU Lille, Department of Biostatistics, F-59000 Lille, France
| | - Frederic Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
13
|
Rodriguez N, Tun HM, Field CJ, Mandhane PJ, Scott JA, Kozyrskyj AL. Prenatal Depression, Breastfeeding, and Infant Gut Microbiota. Front Microbiol 2021; 12:664257. [PMID: 34394021 PMCID: PMC8363245 DOI: 10.3389/fmicb.2021.664257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/23/2021] [Indexed: 02/01/2023] Open
Abstract
Depressive symptoms are common during pregnancy and are estimated to affect 7-20% of pregnant women, with higher prevalence found in those with a prior history of depression, in ethnic minorities, and those with increased exposure to stressful life events. Maternal depression often remains undiagnosed, and its symptoms can increase adverse health risks to the infant, including impaired cognitive development, behavioral problems, and higher susceptibility to physical illnesses. Accumulating research evidence supports the association between maternal physical health elements to infant gut health, including factors such as mode of delivery, medication, feeding status, and antibiotic use. However, specific maternal prenatal psychosocial factors and their effect on infant gut microbiota and immunity remains an area that is not well understood. This article reviews the literature and supplements it with new findings to show that prenatal depression alters: (i) gut microbial composition in partially and fully formula-fed infants at 3-4 months of age, and (ii) gut immunity (i.e., secretory Immunoglobulin A) in all infants independent of breastfeeding status. Understanding the implications of maternal depression on the infant gut microbiome is important to enhance both maternal and child health and to better inform disease outcomes and management.
Collapse
Affiliation(s)
- Nicole Rodriguez
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Hein M Tun
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | | | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Li L, Fang Z, Lee YK, Zhao J, Zhang H, Lu W, Chen W. Prophylactic effects of oral administration of Lactobacillus casei on house dust mite-induced asthma in mice. Food Funct 2021; 11:9272-9284. [PMID: 33047743 DOI: 10.1039/d0fo01363c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study investigated the prophylactic effects of five Lactobacillus casei strains on house dust mite (HDM)-induced asthma in mice. BALB/c mice were orally administered with L. casei strains for one week before HDM treatment. Ketotifen and Lactobacillus rhamnosus GG were used as positive controls. All L. casei strains decreased the number of granulocytes and the levels of Th2 and Th17 inflammatory cytokines in the lungs, L. casei3 significantly decreased the airway inflammation score. Further studies showed that L. casei3, L. casei4, and L. casei5 decreased the chemokine levels, L. casei2, L. casei4, and L. casei5 promoted the secretion of secretory immunoglobulin A (sIgA), L. casei2 upregulated the interleukin (IL)-10 levels, and L. casei1 had no effect on these immune indices. L. casei1 and L. casei4 decreased the serum levels of total IgE and HDM-specific IgG1, respectively. L. casei3 and L. casei5 decreased both HDM-specific IgG1 and total IgE levels. L. casei2 did not affect the levels of these immunoglobulins. The gut microbiota analysis revealed that all five L. casei strains enhanced the richness of the gut microbiota mainly by increasing the abundance of Firmicutes, while there were differences at the genus level.Thus, the prophylactic effects of L. casei on HDM-induced mixed chronic airway inflammatory asthma exerted as they differentially affected the immune responses and gut microbiota composition. L. casei3, which exhibited the highest prophylactic effect, increased the acetate and propionate contents in a strain-dependent manner.
Collapse
Affiliation(s)
- Lingzhi Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhifeng Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China and Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| |
Collapse
|
15
|
Iwamoto A, Inoue Y, Tachibana H, Kawahara H. Immunomodulatory effect of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in allergic conditions in vitro and in vivo. Cytotechnology 2021; 73:333-342. [PMID: 34149169 PMCID: PMC8166990 DOI: 10.1007/s10616-020-00438-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
We found that strawberry extract suppressed immunoglobulin (Ig) E production in vitro and in vivo, and identified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as one of the IgE suppressor in the extract. We report here the effect of GAPDH on various Ig productions in vitro and in vivo. GAPDH suppressed IgE and enhanced IgA, IgG and IgM productions in ovalbumin (OVA)-stimulated human peripheral blood mononuclear cells. Oral administration of GAPDH at 10 mg/kg/day to OVA-induced allergy model mice tended to decrease total IgE level and increase total IgA and IgG levels in sera, and also decreased OVA-specific IgE and IgG levels. It is known that the increase of total IgA as well as the decrease of total and specific IgE is important for alleviating allergic symptoms. In addition, GAPDH accelerated IgA production and increased some cytokine secretions such as IL-4, TGF-β1 and IFN-γ in the OVA-immunized mice spleen lymphocytes. These cytokines involved in the class-switching, IgA enhancement, and IgE suppression, respectively, supporting above results. Our study suggests a possibility that oral administration of GAPDH may induce the immunomodulation in allergic responses.
Collapse
Affiliation(s)
- Akira Iwamoto
- Division of Applied Biological Chemistry, Department of Bioscience and Biochemistry, Faculty of Agriculture, Kyushu University, 744 Motooka Nishi-ku, Fukuoka, 819-0395 Japan
| | - Yuichi Inoue
- Department of Creative Engineering, National Institute of Technology, Kitakyushu College, 5-20-1 Shii, Kokuraminami-ku, Kitakyushu, Fukuoka, 802-0985 Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biochemistry, Faculty of Agriculture, Kyushu University, 744 Motooka Nishi-ku, Fukuoka, 819-0395 Japan
| | - Hiroharu Kawahara
- Department of Creative Engineering, National Institute of Technology, Kitakyushu College, 5-20-1 Shii, Kokuraminami-ku, Kitakyushu, Fukuoka, 802-0985 Japan
| |
Collapse
|
16
|
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: 12] [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
|
17
|
Layhadi JA, Shamji MH. Uncovering the immunological properties of isolated lymphoid follicles. Allergy 2021; 76:1292-1293. [PMID: 32969038 DOI: 10.1111/all.14598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/28/2020] [Accepted: 09/13/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Janice A. Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology National Heart and Lung InstituteImperial College London London UK
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology National Heart and Lung InstituteImperial College London London UK
| |
Collapse
|
18
|
Baumann R, Untersmayr E, Zissler UM, Eyerich S, Adcock IM, Brockow K, Biedermann T, Ollert M, Chaker AM, Pfaar O, Garn H, Thwaites RS, Togias A, Kowalski ML, Hansel TT, Jakwerth CA, Schmidt‐Weber CB. Noninvasive and minimally invasive techniques for the diagnosis and management of allergic diseases. Allergy 2021; 76:1010-1023. [PMID: 33128851 DOI: 10.1111/all.14645] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/13/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022]
Abstract
Allergic diseases of the (upper and lower) airways, the skin and the gastrointestinal tract, are on the rise, resulting in impaired quality of life, decreased productivity, and increased healthcare costs. As allergic diseases are mostly tissue-specific, local sampling methods for respective biomarkers offer the potential for increased sensitivity and specificity. Additionally, local sampling using noninvasive or minimally invasive methods can be cost-effective and well tolerated, which may even be suitable for primary or home care sampling. Non- or minimally invasive local sampling and diagnostics may enable a more thorough endotyping, may help to avoid under- or overdiagnosis, and may provide the possibility to approach precision prevention, due to early diagnosis of these local diseases even before they get systemically manifested and detectable. At the same time, dried blood samples may help to facilitate minimal-invasive primary or home care sampling for classical systemic diagnostic approaches. This EAACI position paper contains a thorough review of the various technologies in allergy diagnosis available on the market, which analytes or biomarkers are employed, and which samples or matrices can be used. Based on this assessment, EAACI position is to drive these developments to efficiently identify allergy and possibly later also viral epidemics and take advantage of comprehensive knowledge to initiate preventions and treatments.
Collapse
Affiliation(s)
- Ralf Baumann
- Medical Faculty Institute for Molecular Medicine Medical School Hamburg (MSH) – Medical University Hamburg Germany
- RWTH Aachen University Hospital Institute for Occupational, Social and Environmental Medicine Aachen Germany
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM) Technical University and Helmholtz Zentrum München München Germany
- Member of the German Center of Lung Research (DZL) and the Helmholtz I&I Initiative Munich Germany
| | - Stefanie Eyerich
- Center of Allergy and Environment (ZAUM) Technical University and Helmholtz Zentrum München München Germany
- Member of the German Center of Lung Research (DZL) and the Helmholtz I&I Initiative Munich Germany
| | - Ian M. Adcock
- National Heart and Lung Institute Imperial College London, and Royal Brompton and Harefield NHS Trust London UK
| | - Knut Brockow
- Department of Dermatology and Allergy Biederstein School of Medicine Technische Universität München Munich Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein School of Medicine Technische Universität München Munich Germany
| | - Markus Ollert
- Department of Infection and Immunity Luxembourg Institute of Health (LIH) Esch‐sur‐Alzette Luxembourg
- Department of Dermatology and Allergy Center Odense Research Centre for Anaphylaxis (ORCA) University of Southern Denmark Odense Denmark
| | - Adam M. Chaker
- Center of Allergy and Environment (ZAUM) Technical University and Helmholtz Zentrum München München Germany
- Member of the German Center of Lung Research (DZL) and the Helmholtz I&I Initiative Munich Germany
- Department of Otolaryngology Allergy Section Klinikum Rechts der Isar Technical University of Munich Munich Germany
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Holger Garn
- Biochemical Pharmacological Center (BPC) ‐ Molecular Diagnostics, Translational Inflammation Research Division & Core Facility for Single Cell Multiomics Philipps University of Marburg ‐ Medical Faculty Member of the German Center for Lung Research (DZL) Universities of Giessen and Marburg Lung Center (UGMLC) Marburg Germany
| | - Ryan S. Thwaites
- National Heart and Lung Institute Imperial College London, and Royal Brompton and Harefield NHS Trust London UK
| | - Alkis Togias
- Division of Allergy, Immunology and Transplantation National Institute of Allergy and Infectious Diseases National Institutes of Health Bethesda MD USA
| | - Marek L. Kowalski
- Department of Immunology and Allergy Medical University of Lodz Lodz Poland
| | - Trevor T. Hansel
- National Heart and Lung Institute Imperial College London, and Royal Brompton and Harefield NHS Trust London UK
| | - Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM) Technical University and Helmholtz Zentrum München München Germany
- Member of the German Center of Lung Research (DZL) and the Helmholtz I&I Initiative Munich Germany
| | - Carsten B. Schmidt‐Weber
- Center of Allergy and Environment (ZAUM) Technical University and Helmholtz Zentrum München München Germany
- Member of the German Center of Lung Research (DZL) and the Helmholtz I&I Initiative Munich Germany
| |
Collapse
|
19
|
The Use of Fecal Calprotectin Testing in Paediatric Disorders: A Position Paper of the European Society for Paediatric Gastroenterology and Nutrition Gastroenterology Committee. J Pediatr Gastroenterol Nutr 2021; 72:617-640. [PMID: 33716293 DOI: 10.1097/mpg.0000000000003046] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of the study was to review the evidence regarding the clinical use and value of fecal calprotectin (FC) measurements in different gastrointestinal disorders in children. METHODS A literature search was conducted in the PubMed, MEDLINE, EMBASE, and Cochrane databases until October 31, 2019. Subtopics were identified and each assigned to individual authors. RESULTS A total of 28 recommendations were voted on using the nominal voting technique. Recommendations are given related to sampling, measurement methods, and results interpretation. The 14 authors anonymously voted on each recommendation using a 9-point scale (1 strongly disagree to 9 fully agree). Consensus was considered achieved if at least 75% of the authors voted 6, 7, 8, or 9. CONCLUSIONS Consensus was reached for all recommendations. Limitations for the use of FC in clinical practice include variability in extraction methodology, performance of test kits as well as the need to establish local reference ranges because of the influence of individual factors, such as age, diet, microbiota, and drugs. The main utility of FC measurement at present is in the diagnosis and monitoring of inflammatory bowel disease (IBD) as well as to differentiate it from functional gastrointestinal disorders (FAPDs). FC, however, has neither utility in the diagnosis of infantile colic nor to differentiate between functional and organic constipation. A rise in FC concentration, may alert to the risk of developing necrotizing enterocolitis and help identifying gastrointestinal involvement in children with Henoch-Schönlein purpura. FC measurement is of little value in Cow's Milk Protein Allergy, coeliac disease (CD), and cystic fibrosis. FC does neither help to distinguish bacterial from viral acute gastroenteritis (AGE), nor to diagnose Helicobacter Pylori infection, small intestinal bacterial overgrowth (SIBO), acute appendicitis (AA), or intestinal polyps.
Collapse
|
20
|
Rachid R, Stephen-Victor E, Chatila TA. The microbial origins of food allergy. J Allergy Clin Immunol 2021; 147:808-813. [PMID: 33347905 PMCID: PMC8096615 DOI: 10.1016/j.jaci.2020.12.624] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/18/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Food allergy (FA) is a significant public health issue, propelled by its rapidly increasing prevalence. Its sharp rise into prominence has focused attention on causative environmental factors and their interplay with the immune system in disease pathogenesis. In that regard, there is now substantial evidence that alterations in the gut microbiome early in life imprint the host gut mucosal immunity and may play a critical role in precipitating FA. These changes may impact key steps in the development of the infant gut microbiome, including its shaping by maternal factors and upon the introduction of solid food (the weaning reaction). These early-life changes may have long-range effects on host immunity that manifest later in time as disease pathology. Experimental studies have shown that resetting the host intestinal immune responses by treatment with either a healthy fecal microbiota transplantation or defined commensal bacterial taxa can prevent or treat FA. The mechanisms by which these interventions suppress FA include restoration of gut immune regulatory checkpoints, notably the retinoic orphan receptor gamma T+ regulatory T cells, the epithelial barrier, and healthy immunoglobulin A responses to the gut commensals. These findings inform human studies currently in progress that evaluate the role of microbial therapies in FA.
Collapse
Affiliation(s)
- Rima Rachid
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Emmanuel Stephen-Victor
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
21
|
Vu K, Lou W, Tun HM, Konya TB, Morales-Lizcano N, Chari RS, Field CJ, Guttman DS, Mandal R, Wishart DS, Azad MB, Becker AB, Mandhane PJ, Moraes TJ, Lefebvre DL, Sears MR, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. From Birth to Overweight and Atopic Disease: Multiple and Common Pathways of the Infant Gut Microbiome. Gastroenterology 2021; 160:128-144.e10. [PMID: 32946900 DOI: 10.1053/j.gastro.2020.08.053] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Few studies, even those with cohort designs, test the mediating effects of infant gut microbes and metabolites on the onset of disease. We undertook such a study. METHODS Using structural equation modeling path analysis, we tested directional relationships between first pregnancy, birth mode, prolonged labor and breastfeeding; infant gut microbiota, metabolites, and IgA; and childhood body mass index and atopy in 1667 infants. RESULTS After both cesarean birth and prolonged labor with a first pregnancy, a higher Enterobacteriaceae/Bacteroidaceae ratio at 3 months was the dominant path to overweight; higher Enterobacteriaceae/Bacteroidaceae ratios and Clostridioides difficile colonization at 12 months were the main pathway to atopic sensitization. Depletion of Bifidobacterium after prolonged labor was a secondary pathway to overweight. Influenced by C difficile colonization at 3 months, metabolites propionate and formate were secondary pathways to child outcomes, with a key finding that formate was at the intersection of several paths. CONCLUSIONS Pathways from cesarean section and first pregnancy to child overweight and atopy share many common mediators of the infant gut microbiome, notably C difficile colonization.
Collapse
Affiliation(s)
- Khanh Vu
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Hein M Tun
- School of Public Health, University of Hong Kong, Hong Kong
| | - Theodore B Konya
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | | | - Radha S Chari
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| | - Rupasri Mandal
- The Metabolomics Innovation Centre, Edmonton, Alberta, Canada
| | - David S Wishart
- The Metabolomics Innovation Centre, Edmonton, Alberta, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Theo J Moraes
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Diana L Lefebvre
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Stuart E Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada; School of Public Health, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
22
|
Wei D, Au Yeung SL, Lu M, Xiao W, Lu J, Shen S, Lam KBH, Qiu X. Association between prenatal depressive symptoms and eczema in infants: The Born in Guangzhou Cohort Study. Pediatr Allergy Immunol 2020; 31:662-670. [PMID: 32301157 DOI: 10.1111/pai.13254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Eczema is a growing threat on infants' health, and the role of maternal depression in the risk of eczema's early onset is unclear. This study aimed to examine the associations of different exposure timing of prenatal depressive symptoms with offspring's eczema in infancy. METHODS The study was part of the ongoing prospective Born in Guangzhou Cohort Study. Maternal depressive symptoms were assessed at both early (<20th week of gestation) and late pregnancy (≥33rd week of gestation to delivery) using the Self-Rating Depression Scale. Information on the diagnosis of eczema was collected when the children were 1 year old. Multivariable logistic regression was used to examine the association between prenatal depressive symptoms and infants' eczema and test for moderation by parental history of allergic diseases. RESULTS In this population, 7.7% (447/5825) of mothers experienced persistent depressive symptoms during pregnancy, 10.1% (590/5825) had depressive symptoms only at early pregnancy, and 8.4% (489/5825) of women experienced depressive symptoms only at late pregnancy. After adjusting for potential confounders, higher risks of eczema were observed in infants of mothers with persistent prenatal depressive symptoms when compared to those children without maternal depressive symptoms throughout pregnancy (OR: 1.55, 95% CI 1.19-2.03). These associations were marginally more pronounced among children in families without parents affected by allergic diseases than in other families (P for interaction = .064 for courses of prenatal depressive symptoms). CONCLUSION Persistent maternal depressive symptoms during pregnancy increased the risk of infants' eczema, especially in children without family history of allergic diseases. These associations, if proved to be causal, could be an intervention target not only to improve women's health but also to prevent offspring's eczema.
Collapse
Affiliation(s)
- Dongmei Wei
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Woman and Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shiu Lun Au Yeung
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Minshan Lu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Woman and Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wanqing Xiao
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Woman and Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jinhua Lu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Woman and Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Songying Shen
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | | | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Woman and Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
23
|
Mine Y, Majumder K, Jin Y, Zeng Y. Chinese sweet tea (Rubus suavissimus) polyphenols attenuate the allergic responses in a Balb/c mouse model of egg allergy. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103827] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
24
|
Mojgani N, Shahali Y, Dadar M. Immune modulatory capacity of probiotic lactic acid bacteria and applications in vaccine development. Benef Microbes 2020; 11:213-226. [PMID: 32216470 DOI: 10.3920/bm2019.0121] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vaccination is one of the most important prevention tools providing protection against infectious diseases especially in children below the age of five. According to estimates, more than 5 million lives are saved annually by the implementation of six standard vaccines, including diphtheria, hepatitis B, Haemophilus influenza type b, polio, tetanus and yellow fever. Despite these efforts, we are faced with challenges in developing countries where increasing population and increasing disease burden and difficulties in vaccine coverage and delivery cause significant morbidity and mortality. Additionally, the high cost of these vaccines is also one of the causes for inappropriate and inadequate vaccinations in these regions. Thus, developing cost-effective vaccine strategies that could provide a stronger immune response with reduced vaccination schedules and maximum coverage is of critical importance. In last decade, different approaches have been investigated; among which live bacterial vaccines have been the focus of attention. In this regard, probiotic lactic acid bacteria have been extensively studied as safe and effective vaccine candidates. These microorganisms represent the largest group of probiotic bacteria in the intestine and are generally recognised as safe (GRAS) bacteria. They have also attracted attention due to their immunomodulatory actions and their effective role as novel vaccine adjuvants. A significant property of these bacteria is their ability to mimic natural infections, while intrinsically possessing mucosal adjuvant properties. Additionally, as live bacterial vaccines are administered orally or nasally, they have higher acceptance and better safety, but also avoid the risk of contamination due to needles and syringes. In this review, we emphasise the role of probiotic Lactobacillus strains as putative oral vaccine carriers and novel vaccine adjuvants.
Collapse
Affiliation(s)
- N Mojgani
- Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), P.O. Box 31975/148, Karaj, Iran
| | - Y Shahali
- Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), P.O. Box 31975/148, Karaj, Iran
| | - M Dadar
- Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), P.O. Box 31975/148, Karaj, Iran
| |
Collapse
|
25
|
Graversen KB, Bahl MI, Larsen JM, Ballegaard ASR, Licht TR, Bøgh KL. Short-Term Amoxicillin-Induced Perturbation of the Gut Microbiota Promotes Acute Intestinal Immune Regulation in Brown Norway Rats. Front Microbiol 2020; 11:496. [PMID: 32292395 PMCID: PMC7135894 DOI: 10.3389/fmicb.2020.00496] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
The intestinal gut microbiota is essential for maintaining host health. Concerns have been raised about the possible connection between antibiotic use, causing microbiota disturbances, and the increase in allergic and autoimmune diseases observed during the last decades. To elucidate the putative connection between antibiotic use and immune regulation, we have assessed the effects of the antibiotic amoxicillin on immune regulation, protein uptake, and bacterial community structure in a Brown Norway rat model. Daily intra-gastric administration of amoxicillin resulted in an immediate and dramatic shift in fecal microbiota, characterized by a reduction of within sample (α) diversity, reduced variation between animals (β diversity), increased relative abundance of Bacteroidetes and Gammaproteobacteria, with concurrent reduction of Firmicutes, compared to a water control group. In the small intestine, amoxicillin also affected microbiota composition significantly, but in a different way than observed in feces. The small intestine of control animals was vastly dominated by Lactobacillus, but this genus was much less abundant in the amoxicillin group. Instead, multiple different genera expanded after amoxicillin administration, with high variation between individual animals, thus the small intestinal α and β diversity were higher in the amoxicillin group compared to controls. After 1 week of daily amoxicillin administration, total fecal IgA level, relative abundance of small intestinal regulatory T cells and goblet cell numbers were higher in the amoxicillin group compared to controls. Several bacterial genera, including Escherichia/Shigella, Klebsiella (Gammaproteobacteria), and Bifidobacterium, for which the relative abundance was higher in the small intestine in the amoxicillin group than in controls, were positively correlated with the fraction of small intestinal regulatory T cells. Despite of epidemiologic studies showing an association between early life antibiotic consumption and later prevalence of inflammatory bowel diseases and food allergies, our findings surprisingly indicated that amoxicillin-induced perturbation of the gut microbiota promotes acute immune regulation. We speculate that the observed increase in relative abundance of small intestinal regulatory T cells is partly mediated by immunomodulatory lipopolysaccharides derived from outgrowth of Gammaproteobacteria.
Collapse
Affiliation(s)
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jeppe Madura Larsen
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | |
Collapse
|
26
|
Kang LJ, Vu KN, Koleva PT, Field CJ, Chow A, Azad MB, Becker AB, Mandhane PJ, Moraes TJ, Sears MR, Lefebvre DL, Turvey SE, Subbarao P, Lou WYW, Scott JA, Kozyrskyj AL. Maternal psychological distress before birth influences gut immunity in mid-infancy. Clin Exp Allergy 2020; 50:178-188. [PMID: 31845414 DOI: 10.1111/cea.13551] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Maternal pre-postnatal psychosocial distress increases the risk for childhood allergic disease. This may occur through a host immunity pathway that involves intestinal secretory immunoglobulin A (sIgA). Experimental animal models show changes in the gut microbiome and immunity of offspring when exposed to direct or prenatal maternal stress, but little is known in humans. OBJECTIVE We determined the association between maternal depression and stress symptom trajectories and infant fecal sIgA concentrations. METHODS 1043 term infants from the Canadian Healthy Infant Longitudinal Development (CHILD) birth cohort were studied. Trajectories of maternal perceived stress and depression were based on scored scales administered in pregnancy and postpartum. sIgA was quantified in infant stool (mean age 3.7 months) with Immundiagnostik ELISA. Linear regression and logistic regression were employed to test associations. RESULTS Very low fecal sIgA concentrations were more common in infants of mothers in the antepartum and persistent depression trajectories (6% and 2% of women, respectively). Independent of breastfeeding status at fecal sampling, infant antibiotic exposure or other covariates, the antepartum depressive symptom trajectory was associated with reduced mean infant sIgA concentrations (β=-0.07, P < .01) and a two fold risk for lowest quartile concentrations (OR, 1.86; 95% CI: 1.02, 3.40). This lowering of sIgA yielded a large effect size in older infants (4-8 months)-breastfed and not. No associations were seen with postpartum depressive symptoms (7% of women) or with any of the perceived stress trajectories. CONCLUSION AND CLINICAL RELEVANCE Despite improved mood postpartum and independent of breastfeeding status, mothers experiencing antepartum depressive symptoms delivered offspring who exhibited lower fecal sIgA concentrations especially in later infancy. The implications of lowered sIgA concentrations in infant stool are altered microbe-sIgA interactions, greater risk for C difficile colonization and atopic disease in later years.
Collapse
Affiliation(s)
- Liane J Kang
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Khanh N Vu
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Petya T Koleva
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Angela Chow
- Department of Applied Health Science, Indiana University, Bloomington, IN, USA
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | | | - Theo J Moraes
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Diana L Lefebvre
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Stuart E Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Wendy Y W Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,School of Public Health, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Stockert K. Allergie, Mikrobiom und weitere epigenetische Faktoren. ALLERGIEPRÄVENTION 2020. [PMCID: PMC7123400 DOI: 10.1007/978-3-662-58140-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
28
|
Xiang H, Sun-Waterhouse D, Waterhouse GI, Cui C, Ruan Z. Fermentation-enabled wellness foods: A fresh perspective. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2019.08.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
29
|
Mine Y, Jin Y, Zhang H, Majumder K, Zeng Y, Sakurai T, Taniguchi Y, Takagaki R, Watanabe H, Mitsuzumi H. Therapeutic effects of isomaltodextrin in a BALB/c mouse model of egg allergy. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
30
|
Oral administration of a mixture of probiotics protects against food allergy via induction of CD103+ dendritic cells and modulates the intestinal microbiota. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
31
|
Fox AT, Wopereis H, Van Ampting MTJ, Oude Nijhuis MM, Butt AM, Peroni DG, Vandenplas Y, Candy DCA, Shah N, West CE, Garssen J, Harthoorn LF, Knol J, Michaelis LJ. A specific synbiotic-containing amino acid-based formula in dietary management of cow's milk allergy: a randomized controlled trial. Clin Transl Allergy 2019; 9:5. [PMID: 30651972 PMCID: PMC6332540 DOI: 10.1186/s13601-019-0241-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023] Open
Abstract
Background Here we report follow-up data from a double-blind, randomized, controlled multicenter trial, which investigated fecal microbiota changes with a new amino acid-based formula (AAF) including synbiotics in infants with non-immunoglobulin E (IgE)-mediated cow’s milk allergy (CMA). Methods Subjects were randomized to receive test product (AAF including fructo-oligosaccharides and Bifidobacterium breve M-16V) or control product (AAF) for 8 weeks, after which infants could continue study product until 26 weeks. Fecal percentages of bifidobacteria and Eubacterium rectale/Clostridium coccoides group (ER/CC) were assessed at 0, 8, 12, and 26 weeks. Additional endpoints included stool markers of gut immune status, clinical symptoms, and safety assessments including adverse events and medication use. Results The trial included 35 test subjects, 36 controls, and 51 in the healthy reference group. Study product was continued by 86% and 92% of test and control subjects between week 8–12, and by 71% and 80%, respectively until week 26. At week 26 median percentages of bifidobacteria were significantly higher in test than control [47.0% vs. 11.8% (p < 0.001)], whereas percentages of ER/CC were significantly lower [(13.7% vs. 23.6% (p = 0.003)]. Safety parameters were similar between groups. Interestingly use of dermatological medication and reported ear infections were lower in test versus control, p = 0.019 and 0.011, respectively. Baseline clinical symptoms and stool markers were mild (but persistent) and low, respectively. Symptoms reduced towards lowest score in both groups. Conclusion Beneficial effects of this AAF including specific synbiotics on microbiota composition were observed over 26 weeks, and shown suitable for dietary management of infants with non-IgE-mediated CMA. Trial Registration NTR3979 Electronic supplementary material The online version of this article (10.1186/s13601-019-0241-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adam T Fox
- 1Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Harm Wopereis
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.,3Wageningen University, Wageningen, The Netherlands
| | - Marleen T J Van Ampting
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Manon M Oude Nijhuis
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Assad M Butt
- 4Royal Alexandra Children's Hospital, Brighton, UK
| | | | | | | | | | | | - Johan Garssen
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.,9Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucien F Harthoorn
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.,3Wageningen University, Wageningen, The Netherlands
| | | | | |
Collapse
|
32
|
Tokuhara D, Kurashima Y, Kamioka M, Nakayama T, Ernst P, Kiyono H. A comprehensive understanding of the gut mucosal immune system in allergic inflammation. Allergol Int 2019; 68:17-25. [PMID: 30366757 DOI: 10.1016/j.alit.2018.09.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/23/2022] Open
Abstract
Despite its direct exposure to huge amounts of microorganisms and foreign and dietary antigens, the gut mucosa maintains intestinal homeostasis by utilizing the mucosal immune system. The gut mucosal immune system protects the host from the invasion of infectious pathogens and eliminates harmful non-self antigens, but it allows the cohabitation of commensal bacteria in the gut and the entry of dietary non-self antigens into the body via the mucosal surface. These physiological and immunological activities are regulated by the ingenious gut mucosal immune network, comprising such features as gut-associated lymphoid tissue, mucosal immune cells, cytokines, chemokines, antimicrobial peptides, secretory IgA, and commensal bacteria. The gut mucosal immune network keeps a fine tuned balance between active immunity (against pathogens and harmful non-self antigens) and immune tolerance (to commensal microbiota and dietary antigens), thus maintaining intestinal healthy homeostasis. Disruption of gut homeostasis results in persistent or severe gastrointestinal infection, inflammatory bowel disease, or allergic inflammation. In this review, we comprehensively introduce current knowledge of the gut mucosal immune system, focusing on its interaction with allergic inflammation.
Collapse
|
33
|
Jiménez-Saiz R, Patil SU. The Multifaceted B Cell Response in Allergen Immunotherapy. Curr Allergy Asthma Rep 2018; 18:66. [PMID: 30291463 DOI: 10.1007/s11882-018-0819-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
While allergen immunotherapy (AIT) for IgE-mediated diseases holds curative potential, the considerable heterogeneity in clinical outcomes may relate to the complex mechanisms of tolerance. The regulation of humoral immunity by AIT contributes to the suppression of allergic responses. Recent findings have revealed novel roles for IgA and IgG antibodies in the induction of tolerance. These mechanisms synergize with their ability to block allergen-IgE binding and mediate inhibitory signaling of effector cells of the allergic response. In addition, the regulatory activity of B cells in AIT extends beyond IL-10 secretion and induction of IgG4. Here, we review the evolution of the B cell response during AIT with special emphasis on the novel protective mechanisms entailing humoral immunity.
Collapse
Affiliation(s)
- Rodrigo Jiménez-Saiz
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University, Madrid, Spain
| | - Sarita U Patil
- Department of Medicine Division of Rheumatology, Allergy, and Immunology, Department of Pediatrics, Division of Allergy and Immunology, Food Allergy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Ulfman LH, Leusen JHW, Savelkoul HFJ, Warner JO, van Neerven RJJ. Effects of Bovine Immunoglobulins on Immune Function, Allergy, and Infection. Front Nutr 2018; 5:52. [PMID: 29988421 PMCID: PMC6024018 DOI: 10.3389/fnut.2018.00052] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
This review aims to provide an in depth overview of the current knowledge of the effects of bovine immunoglobulins on the human immune system. The stability and functional effects of orally ingested bovine immunoglobulins in milk products are described and potential mechanisms of action are discussed. Orally ingested bovine IgG (bovine IgG) can be recovered from feces, ranging from very low levels up to 50% of the ingested IgG that has passed through the gastrointestinal tract. In infants the recovered levels are higher than in adults most likely due to differences in stomach and intestinal conditions such as pH. This indicates that bovine IgG can be functionally active throughout the gastrointestinal tract. Indeed, a large number of studies in infants and adults have shown that bovine IgG (or colostrum as a rich source thereof) can prevent gastrointestinal tract infections, upper respiratory tract infections, and LPS-induced inflammation. These studies vary considerably in target group, design, source of bovine IgG, dosage, and endpoints measured making it hard to draw general conclusions on effectiveness of bovine immunoglobulin rich preparations. Typical sources of bovine IgG used in human studies are serum-derived IgG, colostrum, colostrum-derived IgG, or milk-derived immunoglobulins. In addition, many studies have used IgG from vaccinated cows, but studies using IgG from nonimmunized animals have also been reported to be effective. Mechanistically, bovine IgG binds to many human pathogens and allergens, can neutralize experimental infection of human cells, and limits gastrointestinal inflammation. Furthermore, bovine IgG binds to human Fc receptors which, enhances phagocytosis, killing of bacteria and antigen presentation and bovine IgG supports gastrointestinal barrier function in in vitro models. These mechanisms are becoming more and more established and explain why bovine IgG can have immunological effects in vivo. The inclusion of oral bovine immunoglobulins in specialized dairy products and infant nutrition may therefore be a promising approach to support immune function in vulnerable groups such as infants, children, elderly and immunocompromised patients.
Collapse
Affiliation(s)
| | - Jeanette H W Leusen
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Huub F J Savelkoul
- Wageningen University & Research, Cell Biology and Immunology, Wageningen, Netherlands.,Allergy Consortium Wageningen, Wageningen, Netherlands
| | - John O Warner
- National Institute of Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, Imperial College, London, United Kingdom
| | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Wageningen University & Research, Cell Biology and Immunology, Wageningen, Netherlands
| |
Collapse
|
35
|
Zhu Q, Li F, Wang J, Ma J, Sheng X. Upregulation of calprotectin in mild IgE-mediated ovalbumin hypersensitivity. Oncotarget 2018; 8:37342-37354. [PMID: 28454097 PMCID: PMC5514913 DOI: 10.18632/oncotarget.16954] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Calprotectin, also known as S100A8/A9, has been linked to gut inflammation caused by IgE-mediated food hypersensitivities, but the pathophysiologic abnormalities it causes remain to be determined. We created a mild food hypersensitivity model through oral gavage of ovalbumin in Norway brown rats without using immune adjuvant. Changes in the levels of calprotectin and inflammation-associated cytokines were then observed over time. We found that fecal calprotectin as well as jejunal and liver TLR4, TNF-α, NF-κB, IL-1β, and IL-6 were upregulated in hypersensitive rats. Additionally, the influence of calprotectin on CD4+ T and dendritic cells was observed by co-culturing CD4+ T cells with dendritic cells, which revealed a shift toward increased Th2 T cells in calprotectin-treated cultures. These results suggest that calprotectin, along with other inflammatory factors, promotes the inflammation seen in mild food allergy.
Collapse
Affiliation(s)
- Qingling Zhu
- Department of Child and Adolescent Healthcare, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Feng Li
- Department of Child and Adolescent Healthcare, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Junli Wang
- Department of Child and Adolescent Healthcare, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Jingqiu Ma
- Department of Child and Adolescent Healthcare, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Xiaoyang Sheng
- Department of Child and Adolescent Healthcare, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| |
Collapse
|
36
|
Kang LJ, Koleva PT, Field CJ, Giesbrecht GF, Wine E, Becker AB, Mandhane PJ, Turvey SE, Subbarao P, Sears MR, Scott JA, Kozyrskyj AL. Maternal depressive symptoms linked to reduced fecal Immunoglobulin A concentrations in infants. Brain Behav Immun 2018; 68:123-131. [PMID: 29032226 DOI: 10.1016/j.bbi.2017.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/30/2017] [Accepted: 10/10/2017] [Indexed: 01/23/2023] Open
Abstract
Secretory Immunoglobulin A (sIgA) plays a critical role to infant gut mucosal immunity. Delayed IgA production is associated with greater risk of allergic disease. Murine models of stressful events during pregnancy and infancy show alterations in gut immunity and microbial composition in offspring, but little is known about the stress-microbiome-immunity pathways in humans. We investigated differences in infant fecal sIgA concentrations according to the presence of maternal depressive symptoms during and after pregnancy. A subsample of 403 term infants from the Canadian Healthy Infant Longitudinal Development (CHILD) cohort were studied. Their mothers completed the Center of Epidemiologic Studies Depression Scale when enrolled prenatally and again postpartum. Quantified by Immundiagnostik sIgA ELISA kit, sIgA from infant stool was compared across maternal depressive symptom categories using Mann-Whitney U-tests and logistic regression models that controlled for various covariates. Twelve percent of women reported clinically significant depressive symptoms only prenatally, 8.7% had only postpartum symptoms and 9.2% had symptoms both pre and postnatally. Infants born to mothers with pre and postnatal symptoms had significantly lower median sIgA concentrations than those in the reference group (4.4 mg/g feces vs. 6.3 mg/g feces; p = 0.033). The odds for sIgA concentrations in the lowest quartile was threefold higher (95% CI: 1.25-7.55) when mothers had pre and postnatal symptoms, after controlling for breastfeeding status, infant age, antibiotics exposure and other covariates. Postnatal symptoms were not associated with fecal sIgA, independently of breastfeeding status. Infants born to mothers with depressive symptoms appear to have lower fecal sIgA concentrations, predisposing them to higher risk for allergic disease.
Collapse
Affiliation(s)
- Liane J Kang
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Petya T Koleva
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-126A Li Ka Shing Center for Health Research Innovation, Edmonton, Alberta T6G 2E1, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, CDC, Owerko Centre, Room 355, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Department of Community Health Sciences, University of Calgary, CDC, Owerko Centre, Room 355, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Eytan Wine
- Department of Pediatrics, University of Alberta, 4-577 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, 513 - 715 McDermot Avenue Winnipeg, Manitoba R3E 3P4, Canada
| | - Piushkumar J Mandhane
- Department of Pediatrics, University of Alberta, 1048B Research Transition Facility, Edmonton, Alberta T6G 2V2, Canada
| | - Stuart E Turvey
- Department of Pediatrics, University of British Columbia, BC Children's Hospital, Room A2-147, 950 W 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Peter Gilgan Center for Research and Learning, 686 Bay Street, 10-9716, Toronto, Ontario M5G 0A4, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, 50 Charlton Avenue E., Hamilton, Ontario L8N 4A6, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, 223 College Street, Toronto, Ontario M5T 1R4, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada; School of Public Health, University of Alberta, 3-527 Edmonton Clinic Health Academy, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada.
| | | |
Collapse
|
37
|
Jenmalm MC. The mother-offspring dyad: microbial transmission, immune interactions and allergy development. J Intern Med 2017; 282:484-495. [PMID: 28727206 DOI: 10.1111/joim.12652] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The increasing prevalence of allergy in affluent countries may be caused by reduced intensity and diversity of microbial stimulation, resulting in abnormal postnatal immune maturation. Most studies investigating the underlying immunomodulatory mechanisms have focused on postnatal microbial exposure, for example demonstrating that the gut microbiota differs in composition and diversity during the first months of life in children who later do or do not develop allergic disease. However, it is also becoming increasingly evident that the maternal microbial environment during pregnancy is important in childhood immune programming, and the first microbial encounters may occur already in utero. During pregnancy, there is a close immunological interaction between the mother and her offspring, which provides important opportunities for the maternal microbial environment to influence the immune development of the child. In support of this theory, combined pre- and postnatal supplementations seem to be crucial for the preventive effect of probiotics on infant eczema. Here, the influence of microbial and immune interactions within the mother-offspring dyad on childhood allergy development will be discussed. In addition, how perinatal transmission of microbes and immunomodulatory factors from mother to offspring may shape appropriate immune maturation during infancy and beyond, potentially via epigenetic mechanisms, will be examined. Deeper understanding of these interactions between the maternal and offspring microbiome and immunity is needed to identify efficacious preventive measures to combat the allergy epidemic.
Collapse
Affiliation(s)
- M C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.,International Inflammation (in-FLAME) Network of the World Universities Network, Sydney, NSW, Australia
| |
Collapse
|
38
|
Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, Macia L, Mackay CR. Dietary Fiber and Bacterial SCFA Enhance Oral Tolerance and Protect against Food Allergy through Diverse Cellular Pathways. Cell Rep 2017; 15:2809-24. [PMID: 27332875 DOI: 10.1016/j.celrep.2016.05.047] [Citation(s) in RCA: 432] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/02/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023] Open
Abstract
The incidence of food allergies in western countries has increased dramatically in recent decades. Tolerance to food antigens relies on mucosal CD103(+) dendritic cells (DCs), which promote differentiation of regulatory T (Treg) cells. We show that high-fiber feeding in mice improved oral tolerance and protected from food allergy. High-fiber feeding reshaped gut microbial ecology and increased the release of short-chain fatty acids (SCFAs), particularly acetate and butyrate. High-fiber feeding enhanced oral tolerance and protected against food allergy by enhancing retinal dehydrogenase activity in CD103(+) DC. This protection depended on vitamin A in the diet. This feeding regimen also boosted IgA production and enhanced T follicular helper and mucosal germinal center responses. Mice lacking GPR43 or GPR109A, receptors for SCFAs, showed exacerbated food allergy and fewer CD103(+) DCs. Dietary elements, including fiber and vitamin A, therefore regulate numerous protective pathways in the gastrointestinal tract, necessary for immune non-responsiveness to food antigens.
Collapse
Affiliation(s)
- Jian Tan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Craig McKenzie
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Gera Goverse
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Carola G Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, ACT 0200, Australia
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Laurence Macia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Department of Physiology, Faculty of Medicine, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Department of Physiology, Faculty of Medicine, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
39
|
Lozano-Ojalvo D, López-Fandiño R. Immunomodulating peptides for food allergy prevention and treatment. Crit Rev Food Sci Nutr 2017; 58:1629-1649. [PMID: 28102702 DOI: 10.1080/10408398.2016.1275519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Among the most promising strategies currently assayed against IgE-mediated allergic diseases stands the possibility of using immunomodulating peptides to induce oral tolerance toward offending food allergens or even to prevent allergic sensitization. This review focuses on the beneficial effects of food derived immunomodulating peptides on food allergy, which can be directly exerted in the intestinal tract or once being absorbed through the intestinal epithelial barrier to interact with immune cells. Food peptides influence intestinal homeostasis by maintaining and reinforcing barrier function or affecting intestinal cell-signalling to nearby immune cells and mucus secretion. In addition, they can stimulate cells of the innate and adaptive immune system while supressing inflammatory responses. Peptides represent an attractive alternative to whole allergens to enhance the safety and efficacy of immunotherapy treatments. The conclusions drawn from curative and preventive experiments in murine models are promising, although there is a need for more pre-clinical studies to further explore the immunomodulating strategy and its mechanisms and for a deeper knowledge of the peptide sequence and structural requirements that determine the immunoregulatory function.
Collapse
Affiliation(s)
- Daniel Lozano-Ojalvo
- a Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM) , Madrid , Spain
| | - Rosina López-Fandiño
- a Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM) , Madrid , Spain
| |
Collapse
|
40
|
Genda T, Sasaki Y, Kondo T, Hino S, Nishimura N, Tsukahara T, Sonoyama K, Morita T. Fructo-oligosaccharide-Induced Transient Increases in Cecal Immunoglobulin A Concentrations in Rats Are Associated with Mucosal Inflammation in Response to Increased Gut Permeability. J Nutr 2017; 147:1900-1908. [PMID: 28835391 DOI: 10.3945/jn.117.253955] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 05/31/2017] [Accepted: 07/26/2017] [Indexed: 11/14/2022] Open
Abstract
Background: The mechanism underlying transient increases in immunoglobulin (Ig) A concentrations in the cecal contents of rats fed fructo-oligosaccharide (FOS) is unclear.Objective: This study was designed to test whether increased IgA concentrations represent one aspect of the inflammatory response to increased permeability induced by FOS in the cecum.Methods: Seven-week-old male Wistar rats were fed a fiber-free semipurified diet (FFP) with or without supplemental FOS (60 g/kg diet) for 9 or 58 d [experiment (expt.) 1], 7 d (expt. 2), or 7 or 56 d (expt. 3). In addition to measuring IgA concentrations in cecal content, we assessed gut permeability, inflammatory responses (expt. 1), the number of IgA plasma cells in the cecal lamina propria, polymeric Ig receptor (pIgR) expression in the cecal mucosa (expt. 2), and the condition of the cecal mucus layer (expt. 3).Results: The cecal IgA concentration in the FOS-fed rats was 15-fold higher than that of the rats fed FFP for 9 d (P < 0.05). Gut permeability estimated by urinary chromium-EDTA excretion, bacterial translocation to mesenteric lymph nodes, myeloperoxidase activity, and expression of inflammatory cytokine genes in the cecal mucosa was greater in the FOS-fed rats than in the rats fed FFP for 9 d. These effects were not observed in the rats fed FOS for 58 d (expt. 1). Accompanying the higher cecal IgA concentration, pIgR protein and the number of IgA plasma cells in the cecal mucosa were higher in the FOS-fed rats than in the rats fed FFP for 7 d (expt. 2). Destruction of the mucus layer on the epithelial surface, as evidenced by Alcian blue staining in the cecal sections, was evident in the rats fed FOS for 7 d, but the mucus layer appeared normal in the rats fed FOS for 56 d (expt. 3).Conclusions: These findings suggest that transient increases in cecal IgA concentrations induced by FOS in rats are associated with mucosal inflammation in response to increased gut permeability; these are presumably evoked by disruption of the cecal mucus barrier. The observed responses could contribute to the maturation of the gut immune system.
Collapse
Affiliation(s)
| | - Yuta Sasaki
- Graduate School of Science and Technology and
| | | | - Shingo Hino
- College of Agriculture, Academic Institute, Shizuoka University, Shizuoka, Japan
| | - Naomichi Nishimura
- College of Agriculture, Academic Institute, Shizuoka University, Shizuoka, Japan
| | | | - Kei Sonoyama
- Division of Fundamental AgriScience, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Tatsuya Morita
- College of Agriculture, Academic Institute, Shizuoka University, Shizuoka, Japan;
| |
Collapse
|
41
|
West CE, Dzidic M, Prescott SL, Jenmalm MC. Bugging allergy; role of pre-, pro- and synbiotics in allergy prevention. Allergol Int 2017; 66:529-538. [PMID: 28865967 DOI: 10.1016/j.alit.2017.08.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Large-scale biodiversity loss and complex changes in social behaviors are altering human microbial ecology. This is increasingly implicated in the global rise in inflammatory diseases, most notably the "allergy epidemic" in very early life. Colonization of human ecological niches, particularly the gastrointestinal tract, is critical for normal local and systemic immune development and regulation. Disturbances in composition, diversity and timing of microbial colonization have been associated with increased allergy risk, indicating the importance of strategies to restore a dysbiotic gut microbiota in the primary prevention of allergic diseases, including the administration of probiotics, prebiotics and synbiotics. Here, we summarize and discuss findings of randomized clinical trials that have examined the effects of these microbiome-related strategies on short and long-term allergy preventative effects - including new guidelines from the World Allergy Organization which now recommend probiotics and prebiotics for allergy prevention under certain conditions. The relatively low quality evidence, limited comparative studies and large heterogeneity between studies, have collectively hampered recommendations on specific probiotic strains, specific timing and specific conditions for the most effective preventive management. At the same time the risk of using available products is low. While further research is needed before specific practice guidelines on supplement probiotics and prebiotics, it is equally important that the underlying dietary and lifestyle factors of dysbiosis are addressed at both the individual and societal levels.
Collapse
Affiliation(s)
- Christina E West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden; inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA.
| | - Majda Dzidic
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Susan L Prescott
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; School of Paediatrics and Child Health, University of Western Australia and Princess Margaret Hospital for Children, Perth, Australia
| | - Maria C Jenmalm
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
42
|
Human Milk and Allergic Diseases: An Unsolved Puzzle. Nutrients 2017; 9:nu9080894. [PMID: 28817095 PMCID: PMC5579687 DOI: 10.3390/nu9080894] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to the development of allergic sensitisation and allergic disease. Studies vary in methodology and definition of outcomes, which lead to considerable heterogeneity. Human milk composition varies both within and between individuals, which may partially explain conflicting data. It is known that human milk composition is very complex and contains variable levels of immune active molecules, oligosaccharides, metabolites, vitamins and other nutrients and microbial content. Existing evidence suggests that modulation of human breast milk composition has potential for preventing allergic diseases in early life. In this review, we discuss associations between breastfeeding/human milk composition and allergy development.
Collapse
|
43
|
Smole U, Schabussova I, Pickl WF, Wiedermann U. Murine models for mucosal tolerance in allergy. Semin Immunol 2017; 30:12-27. [PMID: 28807539 DOI: 10.1016/j.smim.2017.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/21/2017] [Indexed: 02/07/2023]
Abstract
Immunity is established by a fine balance to discriminate between self and non-self. In addition, mucosal surfaces have the unique ability to establish and maintain a state of tolerance also against non-self constituents such as those represented by the large numbers of commensals populating mucosal surfaces and food-derived or air-borne antigens. Recent years have seen a dramatic expansion in our understanding of the basic mechanisms and the involved cellular and molecular players orchestrating mucosal tolerance. As a direct outgrowth, promising prophylactic and therapeutic models for mucosal tolerance induction against usually innocuous antigens (derived from food and aeroallergen sources) have been developed. A major theme in the past years was the introduction of improved formulations and novel adjuvants into such allergy vaccines. This review article describes basic mechanisms of mucosal tolerance induction and contrasts the peculiarities but also the interdependence of the gut and respiratory tract associated lymphoid tissues in that context. Particular emphasis is put on delineating the current prophylactic and therapeutic strategies to study and improve mucosal tolerance induction in allergy.
Collapse
Affiliation(s)
- Ursula Smole
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
44
|
Laforest-Lapointe I, Arrieta MC. Patterns of Early-Life Gut Microbial Colonization during Human Immune Development: An Ecological Perspective. Front Immunol 2017; 8:788. [PMID: 28740492 PMCID: PMC5502328 DOI: 10.3389/fimmu.2017.00788] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 06/22/2017] [Indexed: 12/24/2022] Open
Abstract
Alterations in gut microbial colonization during early life have been reported in infants that later developed asthma, allergies, type 1 diabetes, as well as in inflammatory bowel disease patients, previous to disease flares. Mechanistic studies in animal models have established that microbial alterations influence disease pathogenesis via changes in immune system maturation. Strong evidence points to the presence of a window of opportunity in early life, during which changes in gut microbial colonization can result in immune dysregulation that predisposes susceptible hosts to disease. Although the ecological patterns of microbial succession in the first year of life have been partly defined in specific human cohorts, the taxonomic and functional features, and diversity thresholds that characterize these microbial alterations are, for the most part, unknown. In this review, we summarize the most important links between the temporal mosaics of gut microbial colonization and the age-dependent immune functions that rely on them. We also highlight the importance of applying ecology theory to design studies that explore the interactions between this complex ecosystem and the host immune system. Focusing research efforts on understanding the importance of temporally structured patterns of diversity, keystone groups, and inter-kingdom microbial interactions for ecosystem functions has great potential to enable the development of biologically sound interventions aimed at maintaining and/or improving immune system development and preventing disease.
Collapse
Affiliation(s)
- Isabelle Laforest-Lapointe
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
45
|
Debuf MJ, Claeys T, Stalens JP, Cornette L. Hematochezia caused by eosinophilic proctocolitis in a newborn before oral feeding: a case report. J Med Case Rep 2017; 11:160. [PMID: 28619048 PMCID: PMC5472870 DOI: 10.1186/s13256-017-1318-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/11/2017] [Indexed: 01/22/2023] Open
Abstract
Background Hematochezia is a frequent symptom in early infancy. However, it occurs very rarely within the immediate neonatal period, and its occurrence before any oral intake is particularly rare. Because of the “congenital” presentation of hematochezia in our patient, we initially considered our case to be a non-classical, potentially severe type of food protein-induced allergic proctocolitis. This diagnosis needs to be confirmed by an abnormal oral challenge test once the hematochezia has disappeared. If such a challenge cannot demonstrate an allergic origin, then the etiology of the hematochezia could be a neonatal transient eosinophilic colitis. Only two similar cases have been described so far. Case presentation We report the case of a black baby boy of African origin born at 36 weeks 5 days of gestational age who presented with massive hematochezia immediately after birth. A rectosigmoidoscopy revealed a severe inflammation associated with diffuse eosinophilic infiltration on biopsy. His clinical outcome was favorable after introduction of an amino acid formula diet. We initially considered our case to be a non-classical, potentially severe type of food protein-induced allergic proctocolitis but reintroduction of standard formula milk at the age of 3 months was successful. So, our patient is the first newborn in Europe who fits the diagnosis of “neonatal transient eosinophilic colitis.” Conclusions We discuss the possible etiology of “congenital” eosinophilic inflammation of the distal colon and conclude that hematochezia in well-looking neonates, in the absence of negative challenge tests later on, is more likely to be a neonatal transient eosinophilic colitis than an allergic proctocolitis. This new entity could be more frequent than previously thought, changing our medical care strategies for this kind of neonatal symptom.
Collapse
Affiliation(s)
- Marie-Julie Debuf
- Department of Pediatrics, Université Catholique de Louvain, Brussels, Belgium
| | - Tania Claeys
- Department of Pediatrics, AZ Sint-Jan Brugge-Oostende, Bruges, Belgium
| | | | - Luc Cornette
- Department of Neonatology, AZ Sint-Jan Brugge-Oostende, Bruges, Belgium.
| |
Collapse
|
46
|
Kaikiri H, Miyamoto J, Kawakami T, Park SB, Kitamura N, Kishino S, Yonejima Y, Hisa K, Watanabe J, Ogita T, Ogawa J, Tanabe S, Suzuki T. Supplemental feeding of a gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, alleviates spontaneous atopic dermatitis and modulates intestinal microbiota in NC/nga mice. Int J Food Sci Nutr 2017; 68:941-951. [PMID: 28438083 DOI: 10.1080/09637486.2017.1318116] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The present study investigated the antiallergic and anti-inflammatory effects of 10-hydroxy-cis-12-octadecenoic acid (HYA), a novel gut microbial metabolite of linoleic acid, in NC/Nga mice, a model of atopic dermatitis (AD). Feeding HYA decreased the plasma immunoglobulin E level and skin infiltration of mast cells with a concomitant decrease in dermatitis score. HYA feeding decreased TNF-α and increased claudin-1, a tight junction protein, levels in the mouse skin. Cytokine expression levels in the skin and intestinal Peyer's patches cells suggested that HYA improved the Th1/Th2 balance in mice. Immunoglobulin A concentration in the feces of the HYA-fed mice was approximately four times higher than that in the control mice. Finally, denaturing gradient gel electrophoresis of the PCR-amplified 16 S rRNA gene of fecal microbes indicated the modification of microbiota by HYA. Taken together, the alterations in the intestinal microbiota might be, at least in part, associated with the antiallergic effect of HYA.
Collapse
Affiliation(s)
- Hiroko Kaikiri
- a Graduate School of Biosphere Science , Hiroshima University , Higashi-Hiroshima , Japan
| | - Junki Miyamoto
- a Graduate School of Biosphere Science , Hiroshima University , Higashi-Hiroshima , Japan
| | - Takahiro Kawakami
- a Graduate School of Biosphere Science , Hiroshima University , Higashi-Hiroshima , Japan
| | - Si-Bum Park
- b Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Nahoko Kitamura
- b Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | | | - Yasunori Yonejima
- c Research and Development Department , Nitto Pharmaceutical Industries, Ltd , Muko , Japan
| | - Keiko Hisa
- c Research and Development Department , Nitto Pharmaceutical Industries, Ltd , Muko , Japan
| | - Jun Watanabe
- d National Food Research Institute, National Agriculture and Food Research Organization , Tsukuba , Japan
| | - Tasuku Ogita
- d National Food Research Institute, National Agriculture and Food Research Organization , Tsukuba , Japan
| | - Jun Ogawa
- b Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Soichi Tanabe
- a Graduate School of Biosphere Science , Hiroshima University , Higashi-Hiroshima , Japan
| | - Takuya Suzuki
- a Graduate School of Biosphere Science , Hiroshima University , Higashi-Hiroshima , Japan
| |
Collapse
|
47
|
Abstract
Secretory immunoglobulin A (IgA) plays a critical role in gut mucosal immune defense. Initially provided by breastmilk, IgA production by the infant gut is gradually stimulated by developing gut microbiota. This study reports associations between infant fecal IgA concentrations 4 months after birth, breastfeeding status and other pre/postnatal exposures in 47 infants in the Canadian Healthy Infant Longitudinal Development cohort. Breastfed infants and first-born infants had higher median fecal IgA concentrations (23.11 v. 9.34 µg/g protein, P<0.01 and 22.19 v. 8.23 µg/g protein, P=0.04). IgA levels increased successively with exclusivity of breastfeeding (β-coefficient, 0.37, P<0.05). This statistical association was independent of maternal parity and household pets. In the absence of breastfeeding, female sex and pet exposure elevated fecal IgA to levels found in breastfed infants. In addition to breastfeeding, infant fecal IgA associations with pre/postnatal exposures may affect gut immunity and risk of allergic disease.
Collapse
|
48
|
Dzidic M, Abrahamsson TR, Artacho A, Björkstén B, Collado MC, Mira A, Jenmalm MC. Aberrant IgA responses to the gut microbiota during infancy precede asthma and allergy development. J Allergy Clin Immunol 2016; 139:1017-1025.e14. [PMID: 27531072 DOI: 10.1016/j.jaci.2016.06.047] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/11/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although a reduced gut microbiota diversity and low mucosal total IgA levels in infancy have been associated with allergy development, IgA responses to the gut microbiota have not yet been studied. OBJECTIVE We sought to determine the proportions of IgA coating together with the characterization of the dominant bacteria, bound to IgA or not, in infant stool samples in relation to allergy development. METHODS A combination of flow cytometric cell sorting and deep sequencing of the 16S rDNA gene was used to characterize the bacterial recognition patterns by IgA in stool samples collected at 1 and 12 months of age from children staying healthy or having allergic symptoms up to 7 years of age. RESULTS The children with allergic manifestations, particularly asthma, during childhood had a lower proportion of IgA bound to fecal bacteria at 12 months of age compared with healthy children. These alterations cannot be attributed to differences in IgA levels or bacterial load between the 2 groups. Moreover, the bacterial targets of early IgA responses (including coating of the Bacteroides genus), as well as IgA recognition patterns, differed between healthy children and children with allergic manifestations. Altered IgA recognition patterns in children with allergy were observed already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breast-fed children. CONCLUSION An aberrant IgA responsiveness to the gut microbiota during infancy precedes asthma and allergy development, possibly indicating an impaired mucosal barrier function in allergic children.
Collapse
Affiliation(s)
- Majda Dzidic
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden; Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Paediatrics, Linköping University, Linköping, Sweden
| | - Alejandro Artacho
- Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain
| | - Bengt Björkstén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Alex Mira
- Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain.
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.
| |
Collapse
|
49
|
Bridgman SL, Konya T, Azad MB, Guttman DS, Sears MR, Becker AB, Turvey SE, Mandhane PJ, Subbarao P, Scott JA, Field CJ, Kozyrskyj AL. High fecal IgA is associated with reduced Clostridium difficile colonization in infants. Microbes Infect 2016; 18:543-9. [PMID: 27235197 DOI: 10.1016/j.micinf.2016.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022]
Abstract
Colonization of infants with Clostridium difficile is on the rise. Although better tolerated by infants than adults, it is a risk factor for future allergic disease. The present study describes associations between infant fecal immunoglobulin A (IgA) and colonization with C. difficile in 47 infants enrolled in the Canadian Healthy Infant Longitudinal Development (CHILD) study. C. difficile colonization was observed in over half (53%) of the infants. Median IgA was lower in infants colonized with C. difficile (10.9 μg versus 25.5 μg per g protein; p = 0.18). A smaller proportion of infants with IgA in the highest tertile were colonized with C. difficile compared to the other tertiles (31.3% versus 64.5%, p = 0.03). In unadjusted analysis, odds of colonization with C. difficile was reduced by 75% (OR 0.25 95% CI 0.07, 0.91 p = 0.04) among infants with IgA in the highest tertile compared to those in the other tertiles. Following adjustment for parity, birth mode and breastfeeding, this association was even stronger (aOR 0.17, 95% CI 0.03, 0.94, p = 0.04). Our study provides evidence that high fecal IgA, independent of breastfeeding, is associated with reduced likelihood of C. difficile colonization in infancy.
Collapse
Affiliation(s)
| | - Tedd Konya
- Dalla Lana School of Public Health, University of Toronto, ON, Canada
| | - Meghan B Azad
- Department of Pediatrics, University of Alberta, AB, Canada; Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, MB, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, de Groote School of Medicine, McMaster University, ON, Canada
| | - Allan B Becker
- Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, MB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, Child & Family Research Institute and BC Children's Hospital, University of British Columbia, BC, Canada
| | | | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, ON, Canada
| | | | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, ON, Canada
| | - Catherine J Field
- Agriculture, Food and Nutritional Sciences, University of Alberta, AB, Canada
| | | |
Collapse
|
50
|
Effects of Bifidobacterium supplementation on intestinal microbiota composition and the immune response in healthy infants. World J Pediatr 2016; 12:177-82. [PMID: 25846071 DOI: 10.1007/s12519-015-0025-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 12/22/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND Intestinal microbiotas are thought to be the most important source of maturational stimuli to the development of the immune system. However, few studies have focused on the development of T helper (Th) 1 immune response and antibody response to vaccinations in healthy infants, especially in a large cohort. Through this randomized, double-blind control trial, we investigated the effects of Bifidobacterium longum BB536 (BB536) supplementation on intestinal microbiota composition and the immune response in term infants. METHODS In total, 300 healthy newborns were recruited, randomized and fed formula either supplemented with BB536 or with no supplementation. Stool samples were analyzed at months 2, 4 and 11. The representative cytokine for Th1 [interferon-γ (IFN-γ)] and Th2 [interleukin-4 (IL-4)] secretion cells were measured using enzyme-linked immunospot assay at 4 and 7 months of age. The antibody response to vaccines was measured at months 7 and 11. RESULTS A total of 264 infants completed the study. The amount of bifidobacteria and the bifidobacteria/ Enterobacteriaceae ratio (B/E) were significantly higher in the BB536 supplementation group at months 2 and 4. The number of IFN-γ secretion cells and the ratio of IFN-γ/IL-4 secretion cells were increased in the BB536 supplementation group at 7 months. Moreover, the higher value of B/E in the early stages seems to be related to the increased Th1 response. No difference was observed between groups in the antibody response after vaccination. CONCLUSION BB536 has positive effects on establishing a healthy intestinal microbiota early in life, and it also plays an important role in improving the Th1 immune response.
Collapse
|