1
|
Liu H, Lu H, Wang Y, Yu C, He Z, Dong H. Unlocking the power of short-chain fatty acids in ameliorating intestinal mucosal immunity: a new porcine nutritional approach. Front Cell Infect Microbiol 2024; 14:1449030. [PMID: 39286812 PMCID: PMC11402818 DOI: 10.3389/fcimb.2024.1449030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Short-chain fatty acids (SCFAs), a subset of organic fatty acids with carbon chains ranging from one to six atoms in length, encompass acetate, propionate, and butyrate. These compounds are the endproducts of dietary fiber fermentation, primarily catalyzed by the glycolysis and pentose phosphate pathways within the gut microbiota. SCFAs act as pivotal energy substrates and signaling molecules in the realm of animal nutrition, exerting a profound influence on the intestinal, immune system, and intestinal barrier functions. Specifically, they contibute to 60-70% of the total energy requirements in ruminants and 10-25% in monogastric animals. SCFAs have demonstrated the capability to effectively modulate intestinal pH, optimize the absorption of mineral elements, and impede pathogen invasion. Moreover, they enhance the expression of proteins associated with intestinal tight junctions and stimulate mucus production, thereby refining intestinal tissue morphology and preserving the integrity of the intestinal structure. Notably, SCFAs also exert anti-inflammatory properties, mitigating inflammation within the intestinal epithelium and strengthening the intestinal barrier's defensive capabilities. The present review endeavors to synthesize recent findings regarding the role of SCFAs as crucial signaling intermediaries between the metabolic activities of gut microbiota and the status of porcine cells. It also provides a comprehensive overview of the current literature on SCFAs' impact on immune responses within the porcine intestinal mucosa.
Collapse
Affiliation(s)
- Haoyang Liu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Hongde Lu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Yuxuan Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Chenyun Yu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Zhiyuan He
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Hong Dong
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
2
|
Robinson JL, Gatford KL, Bailey DN, Roff AJ, Clifton VL, Morrison JL, Stark MJ. Preclinical models of maternal asthma and progeny outcomes: a scoping review. Eur Respir Rev 2024; 33:230174. [PMID: 38417970 PMCID: PMC10900068 DOI: 10.1183/16000617.0174-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/09/2023] [Indexed: 03/01/2024] Open
Abstract
There is an increased risk of adverse perinatal outcomes in the ∼17% of women with asthma during pregnancy. The mechanisms linking maternal asthma and adverse outcomes are largely unknown, but reflect joint effects of genetics and prenatal exposure to maternal asthma. Animal models are essential to understand the underlying mechanisms independent of genetics and comorbidities, and enable safe testing of interventions. This scoping review aimed to explore the methodology, phenotype, characteristics, outcomes and quality of published studies using preclinical maternal asthma models. MEDLINE (PubMed), Embase (Elsevier) and Web of Science were systematically searched using previously validated search strings for maternal asthma and for animal models. Two reviewers independently screened titles and abstracts, full texts, and then extracted and assessed the quality of each study using the Animal Research: Reporting of In Vivo Experiments (ARRIVE) 2.0 guidelines. Out of 3618 studies identified, 39 were eligible for extraction. Most studies were in rodents (86%) and all were models of allergic asthma. Maternal and progeny outcomes included airway hyperresponsiveness, airway resistance, inflammation, lung immune cells, lung structure and serum immunoglobulins and cytokines. Experimental design (100%), procedural details (97%) and rationale (100%) were most often reported. Conversely, data exclusion (21%), blinding (18%) and adverse events (8%) were reported in a minority of studies. Species differences in physiology and timing of development, the use of allergens not relevant to humans and a lack of comparable outcome measures may impede clinical translation. Future studies exploring models of maternal asthma should adhere to the minimum core outcomes set presented in this review.
Collapse
Affiliation(s)
- Joshua L Robinson
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kathryn L Gatford
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Danielle N Bailey
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Andrea J Roff
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Vicki L Clifton
- Mater Research Institute, University of Queensland, Brisbane, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Michael J Stark
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Department of Neonatal Medicine, Women's & Children's Hospital, Adelaide, Australia
| |
Collapse
|
3
|
Olujimi O, Ajakore S, Abuganloye D, Arowolo T, Steiner O, Goessler W, Towolawi T. Levels of toxic and trace metals in the breast milk of lactating mothers in Abeokuta, Ogun State, Nigeria. Toxicol Rep 2023; 11:168-173. [PMID: 37649468 PMCID: PMC10462791 DOI: 10.1016/j.toxrep.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
Background and objectives Breast milk is an essential source of nutrients and energy for infants. The study analyzed for the levels of essential, toxic and rare earth elements in the breast milk of lactating mothers within Abeokuta metropolis. Materials and methods Thirty-seven (37) breast milk samples were collected with consents of lactating mothers at Ogun State General Hospital in Abeokuta. The samples were digested using standard method and analyzed for essential, toxic and rare earth elements using Inductively Coupled Plasma Mass Spectrometer (ICP-MS). The data were subjected to descriptive analysis. Results The results showed higher concentrations of toxic elements than essential elements in the breast milk of lactating mothers, where five toxic metals: Ag, Ti, V, Pb and Ba were observed to be present in 11, 14, 15, 17 and 23 breast milk samples respectively. Two essential (P and S) and two toxic (Cd and Hg (except sample 19)) elements were observed to be present in all the breast milk samples. Rare Earth Elements (except Sr, U and Rb) were below the detection limit of the instrument. Though three breast milk samples (12, 14 and 17) were observed safe, they contained two toxic (Cd and Hg) and a rare earth trace (Rb) element. Conclusion It could be concluded that despite the inherent benefits of human breast milk with essential elements to the infants, it can still be a source of toxic and trace earth metals contamination.
Collapse
Affiliation(s)
- Olanrewaju Olujimi
- Department of Environmental Management and Toxicology, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Ogun State, Nigeria
| | - Sulaimon Ajakore
- Department of Environmental Management and Toxicology, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Ogun State, Nigeria
| | - Damilola Abuganloye
- Department of Environmental Management and Toxicology, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Ogun State, Nigeria
| | - Toyin Arowolo
- Department of Environmental Management and Toxicology, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Ogun State, Nigeria
| | - Oliver Steiner
- Institute for Chemistry, Department of Analytical Chemistry, Karl-Franzens University, Universitaesplatz 1, 8010 Graz, Austria
| | - Walter Goessler
- Institute for Chemistry, Department of Analytical Chemistry, Karl-Franzens University, Universitaesplatz 1, 8010 Graz, Austria
| | - Taofeek Towolawi
- Department of Environmental Health Science, College of Basic Medical and Health Sciences, Fountain University Osogbo, Osun State, Nigeria
| |
Collapse
|
4
|
Elesela S, Arzola-Martínez L, Rasky A, Ptaschinski C, Hogan SP, Lukacs NW. Mucosal IgA immune complex induces immunomodulatory responses in allergic airway and intestinal T H2 disease. J Allergy Clin Immunol 2023; 152:1607-1618.e1. [PMID: 37604310 DOI: 10.1016/j.jaci.2023.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND IgA is the most abundant immunoglobulin at the mucosal surface and although its role in regulating mucosal immunity is not fully understood, its presence is associated with protection from developing allergic disease. OBJECTIVE We sought to determine the role of IgA immune complexes for therapeutic application to mucosal allergic responses. METHODS Trinitrophenol (TNP)-specific IgA immune complexes were applied, using TNP-coupled ovalbumin (OVA), to airway and gut mucosal surfaces in systemically sensitized allergic animals to regulate allergen challenge responses. Animals were assessed for both pathologic and immune-mediated responses in the lung and gut, respectively, using established mouse models. RESULTS The mucosal application of IgA immune complexes in the lung and gut with TNP-OVA regulated TH2-driven allergic response in the lung and gut, reducing TH2 cytokines and mucus (lung) as well as diarrhea and temperature loss (gut), but increasing IL-10 and the number of regulatory T cells. The IgA-OVA immune complex did not alter peanut-induced anaphylaxis, indicating antigen specificity. Using OVA-specific DO.11-green fluorescent protein IL-4 reporter mouse-derived TH2-skewed cells in a transfer model demonstrated that mucosal IgA immune complex treatment reduced TH2-cell expansion and increased the number of regulatory T cells. To address a potential mechanism of action, TGF-β and IL-10 were induced in bone marrow-derived dendritic cells when they were exposed to IgA immune complex, suggesting a regulatory phenotype induced in dendritic cells that also led to an altered primary T-cell-mediated response in in vitro OVA-specific assays. CONCLUSIONS These studies highlight one possible mechanism of how allergen-specific IgA may provide a regulatory signal to reduce the development of allergic responses in the lung and gut.
Collapse
Affiliation(s)
- Srikanth Elesela
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Llilian Arzola-Martínez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Andrew Rasky
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Simon P Hogan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich.
| |
Collapse
|
5
|
Tomaszewska A, Jeleniewska A, Porębska K, Królikowska K, Rustecka A, Lipińska-Opałka A, Będzichowska A, Zdanowski R, Aleksandrowicz K, Kloc M, Kalicki B. Immunomodulatory Effect of Infectious Disease of a Breastfed Child on the Cellular Composition of Breast Milk. Nutrients 2023; 15:3844. [PMID: 37686876 PMCID: PMC10490220 DOI: 10.3390/nu15173844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Recent studies suggest that the content of immune components in milk is influenced by the mother's health and also by the infant she feeds. We aimed to evaluate the effect of a child's respiratory tract infection on the cellular composition of breast milk (neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations). Twenty-six breastfeeding mothers whose children were hospitalized for respiratory tract infections were enrolled in the study. The control group consisted of 23 mothers of healthy children. Regarding the children, baseline laboratory blood tests were performed, and nasal swabs were taken for the presence of RS virus. In the next step, milk samples were collected from the mothers to assess the cellular composition of the milk, including neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations. Significantly higher percentages of T lymphocytes (helper and cytotoxic lymphocytes) were observed in the milk of the studied mothers. There was a significantly higher percentage of milk lymphocytes in the group of affected children with confirmed RSV etiology than in children with excluded RSV etiology. A significant positive correlation was observed between the duration of infection and the percentage of milk NK cells and between milk CD19 lymphocytes and the child's serum leukocytosis. This study may provide evidence of a link between cells in breast milk and disease in the breastfed infant. The severity of the infection, its duration, and the etiological agent of the infection may affect the cellular composition of milk.
Collapse
Affiliation(s)
- Agata Tomaszewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Alicja Jeleniewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Klaudia Porębska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Katarzyna Królikowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Rustecka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Lipińska-Opałka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agata Będzichowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Karolina Aleksandrowicz
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Małgorzata Kloc
- Transplant Immunology, The Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Bolesław Kalicki
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| |
Collapse
|
6
|
Mohamad Zainal NH, Mohd Nor NH, Saat A, Clifton VL. Childhood allergy susceptibility: The role of the immune system development in the in-utero period. Hum Immunol 2022; 83:437-446. [DOI: 10.1016/j.humimm.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
|
7
|
Brinkhaus M, van der Kooi EJ, Bentlage AEH, Ooijevaar-de Heer P, Derksen NIL, Rispens T, Vidarsson G. Human IgE does not bind to human FcRn. Sci Rep 2022; 12:62. [PMID: 34996950 PMCID: PMC8741920 DOI: 10.1038/s41598-021-03852-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/01/2021] [Indexed: 11/15/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is known to mediate placental transfer of IgG from mother to unborn. IgE is widely known for triggering immune responses to environmental antigens. Recent evidence suggests FcRn-mediated transplacental passage of IgE during pregnancy. However, direct interaction of FcRn and IgE was not investigated. Here, we compared binding of human IgE and IgG variants to recombinant soluble human FcRn with β2-microglobulin (sFcRn) in surface plasmon resonance (SPR) at pH 7.4 and pH 6.0. No interaction was found between human IgE and human sFcRn. These results imply that FcRn can only transport IgE indirectly, and thereby possibly transfer allergenic sensitivity from mother to fetus.
Collapse
Affiliation(s)
- Maximilian Brinkhaus
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Elvera J van der Kooi
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Arthur E H Bentlage
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Pleuni Ooijevaar-de Heer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX, Amsterdam, The Netherlands
| | - Ninotska I L Derksen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX, Amsterdam, The Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
López-Cervantes JP, Lønnebotn M, Jogi NO, Calciano L, Kuiper IN, Darby MG, Dharmage SC, Gómez-Real F, Hammer B, Bertelsen RJ, Johannessen A, Würtz AML, Mørkve Knudsen T, Koplin J, Pape K, Skulstad SM, Timm S, Tjalvin G, Krauss-Etschmann S, Accordini S, Schlünssen V, Kirkeleit J, Svanes C. The Exposome Approach in Allergies and Lung Diseases: Is It Time to Define a Preconception Exposome? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:12684. [PMID: 34886409 PMCID: PMC8657011 DOI: 10.3390/ijerph182312684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022]
Abstract
Emerging research suggests environmental exposures before conception may adversely affect allergies and lung diseases in future generations. Most studies are limited as they have focused on single exposures, not considering that these diseases have a multifactorial origin in which environmental and lifestyle factors are likely to interact. Traditional exposure assessment methods fail to capture the interactions among environmental exposures and their impact on fundamental biological processes, as well as individual and temporal factors. A valid estimation of exposure preconception is difficult since the human reproductive cycle spans decades and the access to germ cells is limited. The exposome is defined as the cumulative measure of external exposures on an organism (external exposome), and the associated biological responses (endogenous exposome) throughout the lifespan, from conception and onwards. An exposome approach implies a targeted or agnostic analysis of the concurrent and temporal multiple exposures, and may, together with recent technological advances, improve the assessment of the environmental contributors to health and disease. This review describes the current knowledge on preconception environmental exposures as related to respiratory health outcomes in offspring. We discuss the usefulness and feasibility of using an exposome approach in this research, advocating for the preconception exposure window to become included in the exposome concept.
Collapse
Affiliation(s)
- Juan Pablo López-Cervantes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Marianne Lønnebotn
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Nils Oskar Jogi
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Lucia Calciano
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | | | - Matthew G. Darby
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa;
| | - Shyamali C. Dharmage
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
| | - Francisco Gómez-Real
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5053 Bergen, Norway
| | - Barbara Hammer
- Department of Pulmonology, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Ane Johannessen
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
| | - Anne Mette Lund Würtz
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Toril Mørkve Knudsen
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Jennifer Koplin
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Kathrine Pape
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Svein Magne Skulstad
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Signe Timm
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark;
- Research Unit, Kolding Hospital, University Hospital of Southern Denmark, 6000 Kolding, Denmark
| | - Gro Tjalvin
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | | | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | - Vivi Schlünssen
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
- National Research Centre for the Working Environment, 2100 Copenhagen, Denmark
| | - Jorunn Kirkeleit
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Cecilie Svanes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| |
Collapse
|
9
|
Rio-Aige K, Azagra-Boronat I, Castell M, Selma-Royo M, Collado MC, Rodríguez-Lagunas MJ, Pérez-Cano FJ. The Breast Milk Immunoglobulinome. Nutrients 2021; 13:nu13061810. [PMID: 34073540 PMCID: PMC8230140 DOI: 10.3390/nu13061810] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022] Open
Abstract
Breast milk components contribute to the infant’s immune development and protection, and among other immune factors, immunoglobulins (Igs) are the most studied. The presence of IgA in milk has been known for a long time; however, less information is available about the presence of other Igs such as IgM, IgG, and their subtypes (IgG1, IgG2, IgG3, and IgG4) or even IgE or IgD. The total Ig concentration and profile will change during the course of lactation; however, there is a great variability among studies due to several variables that limit establishing a clear pattern. In this context, the aim of this review was firstly to shed light on the Ig concentration in breast milk based on scientific evidence and secondly to study the main factors contributing to such variability. A search strategy provided only 75 studies with the prespecified eligibility criteria. The concentrations and proportions found have been established based on the intrinsic factors of the study—such as the sampling time and quantification technique—as well as participant-dependent factors, such as lifestyle and environment. All these factors contribute to the variability of the immunoglobulinome described in the literature and should be carefully addressed for further well-designed studies and data interpretation.
Collapse
Affiliation(s)
- Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46890 Paterna, Valencia, Spain; (M.S.-R.); (M.C.C.)
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (K.R.-A.); (I.A.-B.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| |
Collapse
|
10
|
Fujimura T, Lum SZC, Nagata Y, Kawamoto S, Oyoshi MK. Influences of Maternal Factors Over Offspring Allergies and the Application for Food Allergy. Front Immunol 2019; 10:1933. [PMID: 31507589 PMCID: PMC6716146 DOI: 10.3389/fimmu.2019.01933] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
The prevalence of food allergy has been steadily rising worldwide with the highest incidence noted among younger children, and increasingly recognized as a growing public concern. The first known ingestion of foods often causes allergic reaction, suggesting that sensitization of offspring with food allergens may occur during pregnancy and/or through breastfeeding. This creates a milieu that shapes the neonatal immune responses to these allergens. However, the effects of maternal allergen exposure and maternal sensitization with allergens on development of allergies in offspring remain controversial. This review discusses recent advances from human data in our understanding of how maternal factors, namely, food allergens, allergen-specific immunoglobulins, cytokines, genetics, and environmental factors transferred during pregnancy or breastfeeding influence offspring allergies and how such effects may be applicable to food allergy. Based on information obtained from mouse models of asthma and food allergy, the review also dissects the mechanisms by which maternal factors, including the impact of immune complexes, transforming growth factor-β, vitamin A, and regulatory T-cell responses, contribute to the induction of neonatal tolerance vs. development of allergic responses to maternally transferred allergens.
Collapse
Affiliation(s)
- Takashi Fujimura
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | | | - Yuka Nagata
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Seiji Kawamoto
- Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | - Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Maternal allergen-specific IgG may protect the child against allergic sensitization – literature review. GINECOLOGIA.RO 2019. [DOI: 10.26416/gine.24.2.2019.2376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
12
|
Happle C, Jirmo AC, Meyer-Bahlburg A, Habener A, Hoymann HG, Hennig C, Skuljec J, Hansen G. B cells control maternofetal priming of allergy and tolerance in a murine model of allergic airway inflammation. J Allergy Clin Immunol 2017; 141:685-696.e6. [PMID: 28601684 DOI: 10.1016/j.jaci.2017.03.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/25/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Allergic asthma is a chronic lung disease resulting from inappropriate immune responses to environmental antigens. Early tolerance induction is an attractive approach for primary prevention of asthma. OBJECTIVE We analyzed the mechanisms of perinatal tolerance induction to allergens, with particular focus on the role of B cells in preconception and early intrauterine immune priming. METHODS Wild-type (WT) and B cell-deficient mice received ovalbumin (OVA) intranasally before mating. Their offspring were analyzed in a murine model of allergic airway inflammation. RESULTS Although antigen application before conception protected WT progeny from allergy, it aggravated allergic airway inflammation in B cell-deficient offspring. B-cell transfer restored protection, demonstrating the crucial role of B cells in perinatal tolerance induction. Effective diaplacentar allergen transfer was detectable in pregnant WT mice but not in pregnant B-cell knockout dams, and antigen concentrations in WT amniotic fluid (AF) were higher than in IgG-free AF of B cell-deficient dams. Application of OVA/IgG immune complexes during pregnancy boosted OVA uptake by fetal dendritic cells (DCs). Fetal DCs in human subjects and mice expressed strikingly higher levels of Fcγ receptors compared with DCs from adults and were highly efficient in taking up OVA/IgG immune complexes. Moreover, murine fetal DCs effectively primed antigen-specific forkhead box P3+ regulatory T cells after in vitro coincubation with OVA/IgG-containing AF. CONCLUSION Our data support a decisive role for B cells and immunoglobulins during in utero tolerance priming. These findings improve the understanding of perinatal immunity and might support the development of effective primary prevention strategies for allergy and asthma in the future.
Collapse
Affiliation(s)
- Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Adan Chari Jirmo
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Almut Meyer-Bahlburg
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany; Department of Pediatrics, University Medicine Greifswald, Greifswald, Germany
| | - Anika Habener
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Heinz Gerd Hoymann
- Working Group for Airway Pharmacology, Fraunhofer Institute for Toxicology and Experimental Medicine Hannover, Hannover, Germany
| | - Christian Hennig
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Jelena Skuljec
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
13
|
Schoch JJ, Boull CL, Camilleri MJ, Tollefson MM, Hook KP, Polcari IC. Transplacental Transmission of Pemphigus Herpetiformis in the Setting of Maternal Lymphoma. Pediatr Dermatol 2015. [PMID: 26212699 DOI: 10.1111/pde.12649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pemphigus herpetiformis (PH) is characterized by grouped vesicular or papular pruritic lesions with histologic and immunopathologic features of pemphigus. PH can manifest at any age, and paraneoplastic cases have been reported. We describe a healthy boy born with acral crateriform erosions of the hands and feet whose 35-year-old mother had similar lesions. Biopsies from both patients were most consistent with PH. The mother was diagnosed with high-grade B-cell non-Hodgkin lymphoma and began chemotherapy with dexamethasone, and her lesions quickly improved. The infant had no additional lesions after 3 weeks of age and his acral erosions healed. To our knowledge, this is the first report of a diagnosis of PH in an infant and the first case of transplacental transmission of PH. This is also the first report of paraneoplastic PH in lymphoma. This case adds to the growing differential diagnosis of skin disease in postpartum women and their neonates.
Collapse
Affiliation(s)
| | - Christina L Boull
- Division of Pediatric Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Michael J Camilleri
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota.,Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Kristen P Hook
- Division of Pediatric Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Ingrid C Polcari
- Division of Pediatric Dermatology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
14
|
Abstract
Recently, therapeutic monoclonal antibodies have been introduced for the treatment of advanced melanoma and other diseases. It remains unclear whether these drugs can be safely administered to women who are breast feeding because of the potential hazardous side effects for nursing infants. One such therapy for metastatic melanoma is ipilimumab, a human monoclonal antibody that blocks cytotoxic T-lymphocyte-antigen-4, and is the preferred treatment for patients with metastatic melanoma when other molecular therapies are not viable. This study measured ipilimumab levels in the breast milk of a patient undergoing treatment that were enough to raise concerns for a nursing infant exposed to ipilimumab.
Collapse
|
15
|
López-Expósito I, Srivastava KD, Birmingham N, Castillo A, Miller RL, Li XM. Maternal Antiasthma Simplified Herbal Medicine Intervention therapy prevents airway inflammation and modulates pulmonary innate immune responses in young offspring mice. Ann Allergy Asthma Immunol 2014; 114:43-51.e1. [PMID: 25465920 DOI: 10.1016/j.anai.2014.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/08/2014] [Accepted: 10/14/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Maternal asthma is a risk factor for asthma in offspring; however, transmission of the risk for allergic asthma without direct offspring sensitization has not been explored. OBJECTIVE To determine whether offspring from mothers with ovalbumin (OVA)-sensitized asthma would develop airway disease at first-ever exposure to OVA and whether preconception maternal treatment with the Antiasthma Simplified Herbal Medicine Intervention (ASHMI) or dexamethasone (DEX) could modify this risk in offspring. METHODS Female BALB/c mice (F0) with OVA-induced asthma were generated using established protocols. Mice with asthma were treated with ASHMI, DEX, or water for 6 to 7 weeks. Naive mice served as controls. Subsequently, mice were mated. Twelve-day-old F1 offspring received 3 consecutive intranasal low- or high-dose OVA exposures without sensitization. Forty-eight hours later, airway inflammation, mucus hypersecretion, serum antibodies, and cytokines were evaluated. RESULTS Offspring from OVA-sensitized mothers, but not naive mothers, showed eosinophilic and neutrophilic airway inflammation, and mucus hyperplasia after OVA exposure and he presence of OVA-specific IgG1 and IgG2a. Offspring of ASHMI- and DEX-treated mothers showed decreased airway inflammation and mucus hypersecretion after low-dose OVA (P < .05-.001 for the 2 comparisons vs offspring of OVA/Sham mothers). Offspring of ASHMI-treated, but not DEX-treated, mothers were protected after the high-dose OVA challenge (P < .05-.01 vs offspring OVA/Sham). Maternal ASHMI therapy was associated with increased IgG2a (P < .01 vs offspring of OVA/Sham mothers) and decreased bronchoalveolar lavage fluid CXCL-1 and eotaxin-1 levels (P < .01 and P < .05, respectively, vs offspring of OVA/Sham mothers). CONCLUSION Offspring of mothers with OVA-induced asthma developed airway inflammation and mucus to first-ever OVA exposure without prior sensitization. Maternal therapy with ASHMI was superior to DEX in decreasing offspring susceptibility to airway disease and could be a strategy to lower asthma prevalence.
Collapse
Affiliation(s)
- Iván López-Expósito
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York; Department of Bioactivity and Food Analysis, Institute in Food Science Research (CIAL), CSIC-UAM, Madrid, Spain
| | - Kamal D Srivastava
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York.
| | - Neil Birmingham
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York
| | - Alexandra Castillo
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York
| | - Rachel L Miller
- Division of Pulmonary, Allergy, Critical Care Medicine, Department of Medicine; Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Columbia University, New York, New York
| | - Xiu-Min Li
- Department of Pediatrics, Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
16
|
|
17
|
Yamaki K, Miyatake K, Nakashima T, Morioka A, Yamamoto M, Ishibashi Y, Ito A, Kuranishi A, Yoshino S. Intravenous IgA complexed with antigen reduces primary antibody response to the antigen and anaphylaxis upon antigen re-exposure by inhibiting Th1 and Th2 activation in mice. Immunopharmacol Immunotoxicol 2014; 36:316-28. [PMID: 25077632 DOI: 10.3109/08923973.2014.946143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CONTEXT Serum IgG, IgE and IgM have been shown to enhance the primary antibody responses upon exposure to the soluble antigens recognized by those antibodies. However, how IgA affects these responses remains unknown. OBJECTIVE We investigated the effects of intravenously administered monoclonal IgA on the immune responses in mice. MATERIALS AND METHODS DBA/1J mice were immunized with ovalbumin in the presence or absence of anti-ovalbumin monoclonal IgA. The Th1 and Th2 immune responses to ovalbumin and the anaphylaxis induced by re-exposure to ovalbumin were measured. RESULTS IgA complexed with antigen attenuated the primary antibody responses to the antigen in mice, in contrast to IgG2b and IgE. The primary antibody responses, i.e. the de novo synthesis of anti-ovalbumin IgG2a, IgG1 and IgE in the serum, and the subsequent anaphylaxis induced with re-exposure to ovalbumin were reduced by the co-injection of anti-ovalbumin monoclonal IgA at ovalbumin immunization. The Th1, Th2 and Tr1 cytokines interferon-γ, interleukin-4 and interleukin-10, respectively, released from ovalbumin-restimulated cultured splenocytes collected from allergic mice were also reduced by the treatment. The induction of interferon-γ and interleukin-4 secretion by splenocytes from ovalbumin-immunized mice stimulated in vitro with ovalbumin was also significantly reduced by the antigen complexed with anti-ovalbumin IgA. CONCLUSION These data suggest that the direct inhibition of Th1 and Th2 activation by anti-ovalbumin monoclonal IgA participates in the inhibition of the primary antibody responses. IgA plays important immunosuppressive roles under physiological and pathological conditions and is a promising candidate drug for the treatment of immune disorders.
Collapse
Affiliation(s)
- Kouya Yamaki
- Department of Pharmacology, Kobe Pharmaceutical University , Kobe, Hyogo , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Mother's milk is the fundamental food for infants. It contains proteins, fat, carbohydrates and essential metals which are necessary to ensure correct functioning of the organism. Unfortunately, breast milk is a potential source of toxic metals, which are dangerous for a baby. In Poland, previous research concerning the content of metals in breast milk was very scarce or its results were unavailable. The present study aimed at assessing the content of Cd, Pb, Cu and Zn in human breast milk, as well as estimating the mean weekly intake of these metals by breast-fed infants from Poland. The average concentrations of Cd, Pb, Cu and Zn were 2.114 μg/l, 6.331 μg/l, 0.137 mg/l and 1.623 mg/l, respectively. The admissible levels of supply of these toxic metals has not been exceeded, but their contents were high, particularly in 6-month-old infants (nearly 85 % TWI for Cd and nearly 70 % BMDL₀₁ for Pb). The daily intake of Cu and Zn did not fully satisfy the infant's requirements determined by Polish standards and WHO recommendations. Since the lifestyle of lactating women has a direct influence on the content of these elements in breast milk, women should be educated in this respect with particular focus on eliminating tobacco smoking, both by breastfeeding mothers and by their direct environment.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Department of Bromatology and Food Physiology, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland,
| |
Collapse
|
19
|
Matson AP, Cloutier MM, Dhongade A, Puddington L, Rafti E. Maternal allergy is associated with surface-bound IgE on cord blood basophils. Pediatr Allergy Immunol 2013; 24:614-21. [PMID: 23980848 PMCID: PMC3798094 DOI: 10.1111/pai.12113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND The cell type(s) mediating the maternal influence on allergic disease in children remain unclear. We set out to define the relationship between maternal allergy and frequencies of cord blood (CB) basophils, and plasmacytoid dendritic cells (pDCs); to characterize surface-bound IgE and FcεRI expressions on these cells; and to investigate the association between maternal and CB serum IgE levels with surface-bound IgE and FcεRI expressions. METHODS One hundred and three mother/infant dyads were recruited prenatally, and maternal allergic history was recorded. Maternal blood was collected prior to delivery, and CB was collected after birth. Flow cytometry was used to identify CB basophils and pDCs and to determine surface-bound IgE and FcεRI expressions. RESULTS Frequencies of CB basophils and pDCs were low and not related to maternal history of allergy. Percentages of CB basophils with surface-bound IgE were significantly higher in infants of allergic mothers compared with infants of non-allergic mothers (median, 59.60% vs. 19.70%, p = 0.01). IgE on CB basophils correlated with CB IgE levels (r = 0.72, p < 0.0001), but not with maternal IgE levels (r = 0.26, p = 0.06). IgE on CB pDCs was low and not significantly associated with maternal or CB IgE levels. Similarly, FcεRI expression by CB basophils and pDCs was not significantly associated with maternal or CB IgE levels. CONCLUSIONS Frequencies of CB basophils and pDCs are not influenced by maternal allergy. CB basophils and pDCs have surface-bound IgE and express FcεRI; however, only IgE on CB basophils appears influenced by maternal allergy.
Collapse
Affiliation(s)
- Adam P Matson
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA.
| | | | | | | | | |
Collapse
|
20
|
Horton RE, Vidarsson G. Antibodies and their receptors: different potential roles in mucosal defense. Front Immunol 2013; 4:200. [PMID: 23882268 PMCID: PMC3712224 DOI: 10.3389/fimmu.2013.00200] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/03/2013] [Indexed: 01/07/2023] Open
Abstract
Over recent years it has become increasingly apparent that mucosal antibodies are not only restricted to the IgM and IgA isotypes, but that also other isotypes and particularly IgG can be found in significant quantities at some mucosal surfaces, such as in the genital tract. Their role is more complex than traditionally believed with, among other things, the discovery of novel function of mucosal immunoglobulin receptors. A thorough knowledge in the source and function and mucosal immunoglobulins is particularly important in development of vaccines providing mucosal immunity, and also in the current climate of microbicide development, to combat major world health issues such as HIV. We present here a comprehensive review of human antibody mediated mucosal immunity.
Collapse
Affiliation(s)
- Rachel E Horton
- Institute for Glycomics, Griffith University , Gold Coast, QLD , Australia
| | | |
Collapse
|
21
|
Paveglio S, Puddington L, Rafti E, Matson AP. FcRn-mediated intestinal absorption of IgG anti-IgE/IgE immune complexes in mice. Clin Exp Allergy 2013. [PMID: 23181795 DOI: 10.1111/j.1365-2222.2012.04043.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The mechanism(s) responsible for the acquisition of maternal antibody isotypes other than IgG are not fully understood. OBJECTIVE To define the ability of the neonatal Fc receptor for IgG uptake (FcRn) to mediate intestinal absorption of IgG(1) anti-IgE/IgE immune complexes. METHODS C57BL/6 allergic ovalbumin (OVA)-immune foster mothers were generated to nurse naïve FcRn(+/-) or FcRn(-/-) progeny. At the time of weaning, serum levels of OVA-specific antibodies and IgG(1) anti-IgE/IgE immune complexes were determined in allergic foster mothers and FcRn(+/+), FcRn(+/-), or FcRn(-/-) breastfed offspring. In separate experiments, FcRn(+/-) or FcRn(-/-) neonatal mice were gavage fed TNP-specific IgE as IgG(1) anti-IgE/IgE immune complexes, IgG(1) isotype control and IgE, or IgE alone. Mice were killed 2 h after feeding to determine serum levels and biological activity of absorbed TNP-specific IgE. RESULTS As expected, the absorption of maternal OVA-specific IgG(1) in FcRn(-/-) offspring was at levels 10(3) -10(4) less than observed in FcRn(+/+) or FcRn(+/-) offspring. Surprisingly, FcRn expression also influenced the absorption of maternal IgE. OVA-specific IgE was detected in FcRn(+/+) and FcRn(+/-) offspring, but not in FcRn(-/-) offspring. IgG(1) anti-IgE/IgE immune complexes were detected in allergic foster mothers and correlated strongly with levels in FcRn(+/+) and FcRn(+/-) offspring (ρ = 0.88, P < 0.0001). Furthermore, FcRn expression was required for neonatal mice to absorb TNP-specific IgE when fed as IgG(1) anti-IgE/IgE immune complexes. When immune complexes were generated with IgG(1) anti-IgE directed against the Cε4 domain, the absorbed IgE was able to function in antigen-dependent basophil degranulation. CONCLUSIONS AND CLINICAL RELEVANCE These data demonstrate a novel mechanism by which FcRn may facilitate absorption of maternal antibodies other than IgG. These findings are clinically relevant because FcRn mediates the transplacental passage of maternal IgG to the fetus. This raises the possibility that FcRn could mediate the transplacental passage of maternal IgE as IgG anti-IgE/IgE immune complexes.
Collapse
Affiliation(s)
- S Paveglio
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | |
Collapse
|
22
|
Hansen JS, Andreassen M, Nygaard UC, Løvik M. Offspring IgE responses are influenced by levels of maternal IgG transferred in early life. Am J Reprod Immunol 2013; 70:359-71. [PMID: 23692081 DOI: 10.1111/aji.12139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 04/23/2013] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Maternal immune responses may interfere with offspring allergy development as maternal immunization may suppress IgE development, while maternal allergy may promote allergy. Therefore, we investigated the effect of two different maternal treatments on airway allergy in female and male offspring. METHOD OF STUDY Pregnant mice were immunized (IMM) with ovalbumin (OVA) or immunized and airway-challenged (IMM+AI). At different ages, airway allergy to OVA was induced in offspring by intranasal sensitization. RESULTS Maternal IgG1 was found at higher levels in IMM+AI than in IMM offspring. After sensitization, the suppression of OVA-specific IgE and IgG1 was complete in juvenile offspring but waned with age concurrently with maternal IgG1 levels. Cytokine secretion, lung inflammation, and B cell priming were not suppressed although IgE responses were. CONCLUSIONS High compared with low levels of maternal IgG1 were associated with lower TH 2 antibody production after adult offspring were re-exposed to OVA. Thus, offspring allergy-related responses appeared to be shaped by maternal antibody levels.
Collapse
Affiliation(s)
- Jitka S Hansen
- Norwegian Institute of Public Health, Oslo, Norway; National Research Centre for the Working Environment, Copenhagen, Denmark
| | | | | | | |
Collapse
|
23
|
Affiliation(s)
- S. P. Hogan
- Division of Allergy and Immunology; Cincinnati Children's Hospital Medical Center; Cincinnati; OH; USA
| |
Collapse
|
24
|
Berin MC. Mucosal antibodies in the regulation of tolerance and allergy to foods. Semin Immunopathol 2012; 34:633-42. [PMID: 22777546 DOI: 10.1007/s00281-012-0325-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 06/20/2012] [Indexed: 01/01/2023]
Abstract
The intestinal mucosa is densely packed with antibody-secreting B cells, the majority of which produce IgA. Mucosal antibodies have traditionally been thought of as neutralizing antibodies that exclude antigens, but they also function in antigen sampling, allowing for selective transcytosis of antigens from the intestinal lumen. IgE-mediated antigen uptake can facilitate the development of allergic reactions to foods, but emerging evidence indicates that IgG-mediated antigen uptake may also play an important role in the development of immune tolerance to foods, particularly in the neonate. This review will focus on the role of intestinal immunoglobulins in the development of clinical tolerance and allergy to food antigens.
Collapse
Affiliation(s)
- M Cecilia Berin
- Division of Allergy and Immunology, Department of Pediatrics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
25
|
Roth M. Is there a regulatory role of immunoglobulins on tissue forming cells relevant in chronic inflammatory lung diseases? J Allergy (Cairo) 2011; 2011:721517. [PMID: 22121383 PMCID: PMC3216316 DOI: 10.1155/2011/721517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 08/29/2011] [Indexed: 11/17/2022] Open
Abstract
Epithelial cells, fibroblasts and smooth muscle cells together form and give structure to the airway wall. These three tissue forming cell types are structure giving elements and participate in the immune response to inhaled particles including allergens and dust. All three cell types actively contribute to the pathogenesis of chronic inflammatory lung diseases such as asthma and chronic obstructive pulmonary disease (COPD). Tissue forming cells respond directly to allergens through activated immunoglobulins which then bind to their corresponding cell surface receptors. It was only recently reported that allergens and particles traffic through epithelial cells without modification and bind to the immunoglobulin receptors on the surface of sub-epithelial mesenchymal cells. In consequence, these cells secrete pro-inflammatory cytokines, thereby extending the local inflammation. Furthermore, activation of the immunoglobulin receptors can induce proliferation and tissue remodeling of the tissue forming cells. New studies using anti-IgE antibody therapy indicate that the inhibition of immunoglobulins reduces the response of tissue forming cells. The unmeasured questions are: (i) why do tissue forming cells express immunoglobulin receptors and (ii) do tissue forming cells process immunoglobulin receptor bound particles? The focus of this review is to provide an overview of the expression and function of various immunoglobulin receptors.
Collapse
Affiliation(s)
- Michael Roth
- Pulmonary Cell Research, Department of Research and Pneumology, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
26
|
López-Expósito I, Järvinen KM, Castillo A, Seppo AE, Song Y, Li XM. Maternal peanut consumption provides protection in offspring against peanut sensitization that is further enhanced when co-administered with bacterial mucosal adjuvant. Food Res Int 2011; 44:1649-1656. [PMID: 21927544 DOI: 10.1016/j.foodres.2011.04.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aims of the present study were to assess whether protection against peanut (PN) sensitization can be conferred by maternal PN consumption alone and if so, whether protection was increased by mucosal adjuvant co-administration. Mice were fed with low dose of either PN or PN with cholera toxin (CT) preconceptionally, and during pregnancy and lactation. Offspring serum PN-specific immunoglobulins and cellular responses by splenocytes and mesenteric lymph node (MLN) cells were determined after an active PN sensitization protocol. Milk was collected from lactating mothers of 11-21-day-old pups for evaluation of PN-specific immunoglobulin levels. We found that offspring of PN fed mothers exhibited lower PN-specific IgE levels and reduced PN-stimulated splenocyte and MLN cells cytokine secretion than offspring of non PN fed mothers. CT co-administration with PN enhanced these responses.. Milk from mothers fed PN and CT, but not PN alone preconceptionally and during pregnancy and lactation contained markedly and significantly increased levels of both peanut-specific IgG2a and IgA. Our study demonstrated that maternal feeding of PN alone had a protective effect against PN sensitization of the progeny, which was enhanced by co-administration of a mucosal adjuvant. Increased levels of PN-specific IgG2a and/or IgA in milk were seen when PN and CT were administered together, suggesting that transmission of maternal immunoglobulins may play a role in the observed protection.
Collapse
Affiliation(s)
- Iván López-Expósito
- Pediatric Allergy and Immunology, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
27
|
Transmission of murine cytomegalovirus in breast milk: a model of natural infection in neonates. J Virol 2011; 85:5115-24. [PMID: 21367905 DOI: 10.1128/jvi.01934-10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vertical transmission of viruses in breast milk can expose neonates to infectious pathogens at a time when the capacity of their immune system to control infections is limited. We developed a mouse model to study the outcomes of acquisition of murine cytomegalovirus (MCMV) when neonates are breastfed by mothers with acute or latent infection. Breast milk leukocytes collected from lactating mice were examined for the presence of MCMV IE-1 mRNA by reverse transcription-PCR (RT-PCR) with Southern analysis. As determined by this criterion, breast milk leukocytes from both acute and latent mothers were positive for MCMV. This mimics the outcome seen in humans with latent cytomegalovirus infection, where reactivation of virus occurs specifically in the lactating mammary gland. Interestingly, intraperitoneal injection of breast milk collected from mothers with latent infection was sufficient to transfer MCMV to neonatal mice, demonstrating that breast milk was a source of virus. Furthermore, we found that MCMV was transmitted from infected mothers to breastfed neonates, with MCMV IE-1 mRNA or infectious virus present in multiple organs, including the brain. In fact, 1 day of nursing was sufficient to transmit MCMV from latent mothers to breastfed neonatal mice. Together, these data validate this mouse model of vertical transmission of MCMV from mothers with acute or latent MCMV infection to breastfed neonates. Its relevance to human disease should prove useful in future studies designed to elucidate the immunological and pathological ramifications of neonatal infection acquired via this natural route.
Collapse
|
28
|
Kuo TT, Baker K, Yoshida M, Qiao SW, Aveson VG, Lencer WI, Blumberg RS. Neonatal Fc receptor: from immunity to therapeutics. J Clin Immunol 2010; 30:777-89. [PMID: 20886282 PMCID: PMC2970823 DOI: 10.1007/s10875-010-9468-4] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor (FcRn), also known as the Brambell receptor and encoded by Fcgrt, is a MHC class I like molecule that functions to protect IgG and albumin from catabolism, mediates transport of IgG across epithelial cells, and is involved in antigen presentation by professional antigen presenting cells. Its function is evident in early life in the transport of IgG from mother to fetus and neonate for passive immunity and later in the development of adaptive immunity and other functions throughout life. The unique ability of this receptor to prolong the half-life of IgG and albumin has guided engineering of novel therapeutics. Here, we aim to summarize the basic understanding of FcRn biology, its functions in various organs, and the therapeutic design of antibody- and albumin-based therapeutics in light of their interactions with FcRn.
Collapse
Affiliation(s)
- Timothy T Kuo
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|