1
|
Charles N, Blank U. IgE-Mediated Activation of Mast Cells and Basophils in Health and Disease. Immunol Rev 2025; 331:e70024. [PMID: 40165512 DOI: 10.1111/imr.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Type 2-mediated immune responses protect the body against environmental threats at barrier surfaces, such as large parasites and environmental toxins, and facilitate the repair of inflammatory tissue damage. However, maladaptive responses to typically nonpathogenic substances, commonly known as allergens, can lead to the development of allergic diseases. Type 2 immunity involves a series of prototype TH2 cytokines (IL-4, IL-5, IL-13) and alarmins (IL-33, TSLP) that promote the generation of adaptive CD4+ helper Type 2 cells and humoral products such as allergen-specific IgE. Mast cells and basophils are integral players in this network, serving as primary effectors of IgE-mediated responses. These cells bind IgE via high-affinity IgE receptors (FcεRI) expressed on their surface and, upon activation by allergens, release a variety of mediators that regulate tissue responses, attract and modulate other inflammatory cells, and contribute to tissue repair. Here, we review the biology and effector mechanisms of these cells, focusing primarily on their role in mediating IgE responses in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Nicolas Charles
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine Site Bichat, Paris, France
- Laboratoire d'Excellence Inflamex, Université Paris Cité, Paris, France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine Site Bichat, Paris, France
- Laboratoire d'Excellence Inflamex, Université Paris Cité, Paris, France
| |
Collapse
|
2
|
Ehlers M, Jönsson F. Pathogenic and Nonpathogenic Antibody Responses in Allergic Diseases. Eur J Immunol 2025; 55:e202249978. [PMID: 40071673 PMCID: PMC11898564 DOI: 10.1002/eji.202249978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 03/15/2025]
Abstract
Allergen-specific antibodies, particularly of the IgE class, are a hallmark of many allergic diseases. Yet paradoxically, (1) a proportion of healthy individuals possess allergen-specific IgE without clinical signs of allergy; (2) some, but not all, allergic individuals develop a more severe disease over time or fail to respond to allergen-specific immunotherapy; and (3) allergen-specific IgG antibodies can inhibit IgE-mediated responses but they can also induce allergic reactions. In this review, we discuss the occurrence of and transition between nonpathogenic and pathogenic allergen-specific antibody responses in the light of a two-stage model. We recapitulate different factors and scenarios that may induce different inflammatory conditions and qualitatively distinct allergen-specific T- and B-cell responses, influencing IgE origins and affinities, IgE/IgG(4) ratios, IgG effector functions, antibody glycosylation patterns, Fc and glycan-binding receptor expression and involvement, and ultimately their propensity to elicit allergic responses. Differences in these antibody characteristics may determine the onset of symptomatic allergy and the severity or remission of the disease.
Collapse
Affiliation(s)
- Marc Ehlers
- Laboratories of Immunology and Antibody Glycan AnalysisInstitute of Nutritional MedicineUniversity of Lübeck and University Medical Center of Schleswig‐HolsteinLübeckGermany
- Airway Research Center NorthGerman Center for Lung Research (DZL)University of LübeckLübeckGermany
| | - Friederike Jönsson
- Institut PasteurUniversité de Paris Cité, Unit of Antibodies in Therapy and PathologyParisFrance
- CNRSParisFrance
| |
Collapse
|
3
|
Aguilera-Lizarraga J, Lopez-Lopez C, Jaramillo-Polanco J, Florens MV, Yu Y, Tsang QK, Chakraborty A, De Gand S, Pia F, Quan R, Cuende-Estévez M, Van Remoortel S, Strid J, Lomax AE, Berin MC, Craig AW, Kaufmann E, Ormiston ML, Vanner SJ, Hussein H, Boeckxstaens GE, Reed DE. Psychological Stress-Induced Local Immune Response to Food Antigens Increases Pain Signaling Across the Gut in Mice. Gastroenterology 2025:S0016-5085(25)00371-3. [PMID: 39978560 DOI: 10.1053/j.gastro.2025.01.246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND AIMS We recently showed that a bacterial infection can break oral tolerance to food and lead to immunoglobulin E (IgE)-dependent mast cell activation and food-induced abdominal pain, which could constitute an important pathogenic mechanism in postinfectious irritable bowel syndrome (IBS). Here, we investigated whether similar immune mechanisms in response to psychological stress lead to food-evoked pain signaling, and thus potentially explain the pathophysiology in a larger group of patients with IBS. METHODS Mice were exposed to ovalbumin (OVA) during water avoidance stress (WAS) and re-exposed to OVA 5 weeks later. Nociception was evaluated by visceromotor responses and afferent nerve recordings to intestinal distension, and patch-clamp recordings of sensory neurons incubated with intestinal supernatants. The role of IgE and type 2 immunity was evaluated using pharmacologic and genetic approaches. RESULTS Re-exposure to OVA increased pain signaling in the colon and small intestine only in mice exposed to OVA during WAS, in the absence of systemic allergy. OVA-induced increases in pain responses depended on mast cells, IgE, and signal transducer and activator of transcription 6 signaling. Notably, incubation of sensory neurons with ileum and colon supernatants from WAS/OVA+OVA mice lowered their threshold of excitability. Finally, treatment with histamine receptor H1 antagonist pyrilamine blocked the increased sensory neuron excitability, and reduced ileal afferent nerve firing to distension in WAS/OVA+OVA mice. CONCLUSIONS Psychological stress induces a type 2 immune response to food antigens, with IgE-mediated mast cell activation and increased pain signaling in the small intestine and colon in response to food. These findings may explain the potential role of psychological stress in food-induced symptoms in IBS.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Cintya Lopez-Lopez
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Josue Jaramillo-Polanco
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Morgane V Florens
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Quentin K Tsang
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Ananya Chakraborty
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Sofie De Gand
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Fedrica Pia
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Runze Quan
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - María Cuende-Estévez
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Samuel Van Remoortel
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - M Cecilia Berin
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Eva Kaufmann
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Mark L Ormiston
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Hind Hussein
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Center of Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
4
|
Plum T, Feyerabend TB, Rodewald HR. Beyond classical immunity: Mast cells as signal converters between tissues and neurons. Immunity 2024; 57:2723-2736. [PMID: 39662090 DOI: 10.1016/j.immuni.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Mast cells are regarded as effectors in immune defense against parasites and venoms and play an essential role in the pathology of allergic diseases. More recently, mast cells have been shown to receive stimuli derived from type 2 immunity, tissue damage, stress, and inflammation. Mast cells then rapidly convert these diverse signals into appropriate, organ-specific protective reflexes that can limit inflammation or reduce tissue damage. In this review, we consider functions of mast cells in sensations-such as pain, itch, and nausea-arising from tissue insults and inflammation and the ensuing protective responses. In light of emerging data highlighting the involvement of mast cells in neuroimmune communication, we also propose that mast cells are "signal converters" linking immunological and tissue states with nervous system responses.
Collapse
Affiliation(s)
- Thomas Plum
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| | - Thorsten B Feyerabend
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
5
|
Daëron M, Heyman B, Moulin AM. Life-threatening antibodies: The discovery of anaphylaxis. Immunol Rev 2024; 328:24-38. [PMID: 39495000 DOI: 10.1111/imr.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
It was at the turn of the 20th century, that immune serum was found both to save children dying from toxins of deadly pathogens, and to kill a dog within minutes following an injection of harmless doses of sea anemone toxins. This means that, before being formally identified in immune serum, antibodies were already known to be both protective and pathogenic. For this provocative finding, Charles Richet was awarded the 1913 Nobel Prize in Physiology or Medicine. Because, as its name said, anaphylaxis was understood as "the contrary of protection," unique mechanisms were found to explain it. Because, as its name did not initially say but finally said, allergy was understood as a reaction "other" than immunity, its symptoms were explained by mechanisms similar to those that explained anaphylaxis. We examined here the intricate relationships between anaphylaxis, allergy and immunity. Progressively anaphylaxis became one among other pathological effects of an immune response, and allergy an inflammatory disease among others. Looking at antibodies back in the past enables us not only to learn where they come from, but also to follow trends that contributed to shape immunology, some of which may persist in today's immunological thinking and say something about the future.
Collapse
Affiliation(s)
- Marc Daëron
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université-CNRS-Inserm, Marseille, France
- Institut Pasteur-Université Paris Cité, Paris, France
- Institut d'histoire et de philosophie des sciences et des techniques, Université Paris 1 Panthéon Sorbonne-CNRS, Paris, France
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
6
|
Koenig JFE, Knudsen NPH, Phelps A, Bruton K, Hoof I, Lund G, Libera DD, Lund A, Christensen LH, Glass DR, Walker TD, Fang A, Waserman S, Jordana M, Andersen PS. Type 2-polarized memory B cells hold allergen-specific IgE memory. Sci Transl Med 2024; 16:eadi0944. [PMID: 38324637 DOI: 10.1126/scitranslmed.adi0944] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 12/18/2023] [Indexed: 02/09/2024]
Abstract
Allergen-specific immunoglobulin E (IgE) antibodies mediate pathology in diseases such as allergic rhinitis and food allergy. Memory B cells (MBCs) contribute to circulating IgE by regenerating IgE-producing plasma cells upon allergen encounter. Here, we report a population of type 2-polarized MBCs defined as CD23hi, IL-4Rαhi, and CD32low at both the transcriptional and surface protein levels. These MBC2s are enriched in IgG1- and IgG4-expressing cells while constitutively expressing germline transcripts for IgE. Allergen-specific B cells from patients with allergic rhinitis and food allergy were enriched in MBC2s. Furthermore, MBC2s generated allergen-specific IgE during sublingual immunotherapy, thereby identifying these cells as a major reservoir for IgE. The identification of MBC2s provides insights into the maintenance of IgE memory, which is detrimental in allergic diseases but could be beneficial in protection against venoms and helminths.
Collapse
Affiliation(s)
- Joshua F E Koenig
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Allyssa Phelps
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kelly Bruton
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ilka Hoof
- ALK-Abelló A/S, 2970 Hørsholm, Denmark
| | | | - Danielle Della Libera
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | | | - David R Glass
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Tina D Walker
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Allison Fang
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Susan Waserman
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Manel Jordana
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | |
Collapse
|
7
|
Haque TT, Weissler KA, Schmiechen Z, Laky K, Schwartz DM, Li J, Locci M, Turfkruyer M, Yao C, Schaughency P, Leak L, Lack J, Kanno Y, O'Shea J, Frischmeyer-Guerrerio PA. TGFβ prevents IgE-mediated allergic disease by restraining T follicular helper 2 differentiation. Sci Immunol 2024; 9:eadg8691. [PMID: 38241399 DOI: 10.1126/sciimmunol.adg8691] [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: 01/26/2023] [Accepted: 11/15/2023] [Indexed: 01/21/2024]
Abstract
Allergic diseases are common, affecting more than 20% of the population. Genetic variants in the TGFβ pathway are strongly associated with atopy. To interrogate the mechanisms underlying this association, we examined patients and mice with Loeys-Dietz syndrome (LDS) who harbor missense mutations in the kinase domain of TGFΒR1/2. We demonstrate that LDS mutations lead to reduced TGFβ signaling and elevated total and allergen-specific IgE, despite the presence of wild-type T regulatory cells in a chimera model. Germinal center activity was enhanced in LDS and characterized by a selective increase in type 2 follicular helper T cells (TFH2). Expression of Pik3cg was increased in LDS TFH cells and associated with reduced levels of the transcriptional repressor SnoN. PI3Kγ/mTOR signaling in LDS naïve CD4+ T cells was elevated after T cell receptor cross-linking, and pharmacologic inhibition of PI3Kγ or mTOR prevented exaggerated TFH2 and antigen-specific IgE responses after oral antigen exposure in an adoptive transfer model. Naïve CD4+ T cells from nonsyndromic allergic individuals also displayed decreased TGFβ signaling, suggesting that our mechanistic discoveries may be broadly relevant to allergic patients in general. Thus, TGFβ plays a conserved, T cell-intrinsic, and nonredundant role in restraining TFH2 development via the PI3Kγ/mTOR pathway and thereby protects against allergic disease.
Collapse
Affiliation(s)
- Tamara T Haque
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katherine A Weissler
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zoe Schmiechen
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Karen Laky
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jenny Li
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michela Locci
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathilde Turfkruyer
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen Yao
- Laboratory of Lymphocyte Nuclear Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paul Schaughency
- Collaborative Bioinformatics Resource, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lashawna Leak
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- Collaborative Bioinformatics Resource, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yuka Kanno
- Laboratory of Lymphocyte Nuclear Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John O'Shea
- Laboratory of Lymphocyte Nuclear Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela A Frischmeyer-Guerrerio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Plum T, Binzberger R, Thiele R, Shang F, Postrach D, Fung C, Fortea M, Stakenborg N, Wang Z, Tappe-Theodor A, Poth T, MacLaren DAA, Boeckxstaens G, Kuner R, Pitzer C, Monyer H, Xin C, Bonventre JV, Tanaka S, Voehringer D, Vanden Berghe P, Strid J, Feyerabend TB, Rodewald HR. Mast cells link immune sensing to antigen-avoidance behaviour. Nature 2023; 620:634-642. [PMID: 37438525 PMCID: PMC10432277 DOI: 10.1038/s41586-023-06188-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 05/10/2023] [Indexed: 07/14/2023]
Abstract
The physiological functions of mast cells remain largely an enigma. In the context of barrier damage, mast cells are integrated in type 2 immunity and, together with immunoglobulin E (IgE), promote allergic diseases. Allergic symptoms may, however, facilitate expulsion of allergens, toxins and parasites and trigger future antigen avoidance1-3. Here, we show that antigen-specific avoidance behaviour in inbred mice4,5 is critically dependent on mast cells; hence, we identify the immunological sensor cell linking antigen recognition to avoidance behaviour. Avoidance prevented antigen-driven adaptive, innate and mucosal immune activation and inflammation in the stomach and small intestine. Avoidance was IgE dependent, promoted by Th2 cytokines in the immunization phase and by IgE in the execution phase. Mucosal mast cells lining the stomach and small intestine rapidly sensed antigen ingestion. We interrogated potential signalling routes between mast cells and the brain using mutant mice, pharmacological inhibition, neural activity recordings and vagotomy. Inhibition of leukotriene synthesis impaired avoidance, but overall no single pathway interruption completely abrogated avoidance, indicating complex regulation. Collectively, the stage for antigen avoidance is set when adaptive immunity equips mast cells with IgE as a telltale of past immune responses. On subsequent antigen ingestion, mast cells signal termination of antigen intake. Prevention of immunopathology-causing, continuous and futile responses against per se innocuous antigens or of repeated ingestion of toxins through mast-cell-mediated antigen-avoidance behaviour may be an important arm of immunity.
Collapse
Affiliation(s)
- Thomas Plum
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany.
| | - Rebecca Binzberger
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Robin Thiele
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Fuwei Shang
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Daniel Postrach
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Candice Fung
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Marina Fortea
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Zheng Wang
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | | | - Tanja Poth
- Center for Model System and Comparative Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Duncan A A MacLaren
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center, Heidelberg, Germany
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Rohini Kuner
- Pharmacology Institute, Heidelberg University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center, Heidelberg, Germany
| | - Cuiyan Xin
- Division of Renal Medicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph V Bonventre
- Division of Renal Medicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Tanaka
- Laboratory of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Thorsten B Feyerabend
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
9
|
Florsheim EB, Bachtel ND, Cullen JL, Lima BGC, Godazgar M, Carvalho F, Chatain CP, Zimmer MR, Zhang C, Gautier G, Launay P, Wang A, Dietrich MO, Medzhitov R. Immune sensing of food allergens promotes avoidance behaviour. Nature 2023; 620:643-650. [PMID: 37437602 PMCID: PMC10432274 DOI: 10.1038/s41586-023-06362-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
In addition to its canonical function of protection from pathogens, the immune system can also alter behaviour1,2. The scope and mechanisms of behavioural modifications by the immune system are not yet well understood. Here, using mouse models of food allergy, we show that allergic sensitization drives antigen-specific avoidance behaviour. Allergen ingestion activates brain areas involved in the response to aversive stimuli, including the nucleus of tractus solitarius, parabrachial nucleus and central amygdala. Allergen avoidance requires immunoglobulin E (IgE) antibodies and mast cells but precedes the development of gut allergic inflammation. The ability of allergen-specific IgE and mast cells to promote avoidance requires cysteinyl leukotrienes and growth and differentiation factor 15. Finally, a comparison of C57BL/6 and BALB/c mouse strains revealed a strong effect of the genetic background on the avoidance behaviour. These findings thus point to antigen-specific behavioural modifications that probably evolved to promote niche selection to avoid unfavourable environments.
Collapse
Affiliation(s)
- Esther B Florsheim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- Biodesign Institute, Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA.
| | - Nathaniel D Bachtel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jaime L Cullen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Bruna G C Lima
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mahdieh Godazgar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Fernando Carvalho
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carolina P Chatain
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Marcelo R Zimmer
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Cuiling Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gregory Gautier
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Université Paris Cité, Paris, France
| | - Pierre Launay
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Université Paris Cité, Paris, France
| | - Andrew Wang
- Department of Medicine (Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Marcelo O Dietrich
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Kow ASF, Khoo LW, Tan JW, Abas F, Lee MT, Israf DA, Shaari K, Tham CL. Clinacanthus nutans aqueous leaves extract exerts anti-allergic activity in preclinical anaphylactic models via alternative IgG pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:116003. [PMID: 36464074 DOI: 10.1016/j.jep.2022.116003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Allergy is mediated by the crosslinking of immunoglobulins (Ig) -E or -G to their respective receptors, which degranulates mast cells, macrophages, basophils, or neutrophils, releasing allergy-causing mediators. The removal of these mediators such as histamine, platelet-activating factor (PAF) and interleukins (ILs) released by effector cells will alleviate allergy. Clinacanthus nutans (C. nutans), an herbal plant in Southeast Asia, is used traditionally to treat skin rash, an allergic symptom. Previously, we have reported that C. nutans aqueous leaves extract (CNAE) was able to suppress the release of β-hexosaminidase and histamine but not interleukin-4 (IL-4) and tumor necrosis factor-alpha (TNF-α) in the IgE-induced mast cell degranulation model at 5 mg/mL and above. We also found that CNAE could protect rats against ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) through the downregulation and upregulation of certain metabolites using proton nuclear magnetic resonance (1H-NMR) metabolomics approach. AIM OF THE STUDY As allergy could be mediated by both IgE and IgG, we further evaluated the anti-allergy potential of CNAE in both in vitro model of IgG-induced macrophage activation and in vivo anaphylaxis models to further dissect the mechanism of action underlying the anti-allergic properties of CNAE. MATERIAL & METHODS The anti-allergy potential of CNAE was evaluated in in vivo anaphylaxis models of ovalbumin-challenged active systemic anaphylaxis (OVA-ASA) and IgE-challenged passive systemic anaphylaxis (PSA) using Sprague Dawley rats as well as IgG-challenged passive systemic anaphylaxis (IgG-PSA) using C57BL/6 mice. Meanwhile, in vitro model of IgG-induced macrophage activation model was performed using IC-21 macrophages. The release of soluble mediators from both IgE and IgG-mediated pathways were measured using enzyme-linked immunosorbent assay (ELISA). The signaling molecules targeted by CNAE were identified by performing Western blot. RESULTS IgG, platelet-activating factor (PAF) and IL-6 was suppressed by CNAE in OVA-ASA, but not IgE. In addition, CNAE significantly suppressed PAF and IL-6 in IgG-PSA but did not suppress histamine, IL-4 and leukotrienes C4 (LTC4) in IgE-PSA. CNAE also inhibited IL-6 and TNF-α by inhibiting the phosphorylation of ERK1/2 in the IgG-induced macrophage activation model. CONCLUSION Overall, our findings supported that CNAE exerts its anti-allergic properties by suppressing the IgG pathway and its mediators by inhibiting ERK1/2 phosphorylation, thus providing scientific evidence supporting its traditional use in managing allergy.
Collapse
Affiliation(s)
- Audrey Siew Foong Kow
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | - Leng Wei Khoo
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ji Wei Tan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia; School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Malaysia.
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43400, Malaysia; Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Ming-Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan; Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Khozirah Shaari
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.
| |
Collapse
|
11
|
Florsheim EB, Bachtel ND, Cullen J, Lima BGC, Godazgar M, Zhang C, Carvalho F, Gautier G, Launay P, Wang A, Dietrich MO, Medzhitov R. Immune sensing of food allergens promotes aversive behaviour. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524823. [PMID: 36712030 PMCID: PMC9882358 DOI: 10.1101/2023.01.19.524823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In addition to its canonical function in protecting from pathogens, the immune system can also promote behavioural alterations 1â€"3 . The scope and mechanisms of behavioural modifications by the immune system are not yet well understood. Using a mouse food allergy model, here we show that allergic sensitization drives antigen-specific behavioural aversion. Allergen ingestion activates brain areas involved in the response to aversive stimuli, including the nucleus of tractus solitarius, parabrachial nucleus, and central amygdala. Food aversion requires IgE antibodies and mast cells but precedes the development of gut allergic inflammation. The ability of allergen-specific IgE and mast cells to promote aversion requires leukotrienes and growth and differentiation factor 15 (GDF15). In addition to allergen-induced aversion, we find that lipopolysaccharide-induced inflammation also resulted in IgE-dependent aversive behaviour. These findings thus point to antigen-specific behavioural modifications that likely evolved to promote niche selection to avoid unfavourable environments.
Collapse
Affiliation(s)
- Esther B. Florsheim
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA,Centre for Immunotherapy, Vaccines, and Virotherapy (CIVV), Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ 85284, USA,Correspondence: and
| | - Nathaniel D. Bachtel
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Jaime Cullen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Bruna G. C. Lima
- Department of Pharmacology, University of São Paulo, São Paulo, SP 05508-000 SP, Brazil,Centre for Immunotherapy, Vaccines, and Virotherapy (CIVV), Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ 85284, USA
| | - Mahdieh Godazgar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Cuiling Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Fernando Carvalho
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Gregory Gautier
- INSERM UMRS 1149; CNRS ERL 8252; University Paris Diderot, Sorbonne Paris Cite, Laboratoire d’excellence INFLAMEX, Paris 75018, France
| | - Pierre Launay
- INSERM UMRS 1149; CNRS ERL 8252; University Paris Diderot, Sorbonne Paris Cite, Laboratoire d’excellence INFLAMEX, Paris 75018, France
| | - Andrew Wang
- Department of Internal Medicine and Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marcelo O. Dietrich
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA,Howard Hughes Medical Institute,Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA,Correspondence: and
| |
Collapse
|
12
|
Oettgen HC. Mast cells in food allergy: Inducing immediate reactions and shaping long-term immunity. J Allergy Clin Immunol 2023; 151:21-25. [PMID: 36328809 DOI: 10.1016/j.jaci.2022.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/08/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
Abstract
Mast cells are distributed throughout the gastrointestinal tract and function as the main effector cells of IgE-mediated allergic reactions to foods. Allergen-induced cross-linking of IgE antibodies bound to high-affinity IgE receptors, FcεRI, on the surface of mast cells triggers their activation, resulting in the release of mediators of immediate hypersensitivity. These mediators rapidly induce both local gastrointestinal and systemic physiological responses including anaphylaxis. Emerging evidence has revealed that, in addition to inciting immediate reactions, mast cells are key regulators of adaptive immunity to foods. In the gastrointestinal mucosa they provide the priming cytokines that initiate and, over time, consolidate adaptive TH2 responses to ingested allergens as well as TNF and chemokines that orchestrate the recruitment of tissue-infiltrating leukocytes that drive type 2 tissue inflammation. Patients with atopic dermatitis have increased intestinal mast cell numbers and are at a greater risk for food allergy. Recent studies have uncovered a skin-gut axis in which epicutaneous allergen exposure drives intestinal mast cell expansion. The activating effects of IgE antibodies in mast cells are countered by food-specific IgG antibodies that signal via the inhibitory IgG receptor, FcγR2b, suppressing both immediate allergic reactions to foods and the type 2 immune adjuvant activity of mast cells.
Collapse
Affiliation(s)
- Hans C Oettgen
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
13
|
Todorova B, Godon O, Conde E, Gillis CM, Iannascoli B, Richard-Le Goff O, Fiole D, Roumenina LT, Leusen JHW, Murphy AJ, Macdonald LE, Reber LL, Jönsson F, Bruhns P. IgG Subclass-Dependent Pulmonary Antigen Retention during Acute IgG-Dependent Systemic Anaphylaxis in Mice. THE JOURNAL OF IMMUNOLOGY 2022; 209:1243-1251. [DOI: 10.4049/jimmunol.2200234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/25/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Mouse models of active systemic anaphylaxis rely predominantly on IgG Abs forming IgG–allergen immune complexes that induce IgG receptor–expressing neutrophils and monocytes/macrophages to release potent mediators, leading to systemic effects. Whether anaphylaxis initiates locally or systemically remains unknown. In this study, we aimed at identifying the anatomical location of IgG–allergen immune complexes during anaphylaxis. Active systemic anaphylaxis was induced following immunization with BSA and i.v. challenge with fluorescently labeled BSA. Ag retention across different organs was examined using whole-body fluorescence imaging, comparing immunized and naive animals. Various mouse models and in vivo deletion strategies were employed to determine the contribution of IgG receptors, complement component C1q, myeloid cell types, and anaphylaxis mediators. We found that following challenge, Ag diffused systemically, but specifically accumulated in the lungs of mice sensitized to that Ag, where it formed large Ab-dependent aggregates in the vasculature. Ag retention in the lungs did not rely on IgG receptors, C1q, neutrophils, or macrophages. IgG2a-mediated, but neither IgG1- nor IgG2b-mediated, passive systemic anaphylaxis led to Ag retention in the lung. Neutrophils and monocytes significantly accumulated in the lungs after challenge and captured high amounts of Ag, which led to downmodulation of surface IgG receptors and triggered their activation. Thus, within minutes of systemic injection in sensitized mice, Ag formed aggregates in the lung and liver vasculature, but accumulated specifically and dose-dependently in the lung. Neutrophils and monocytes recruited to the lung captured Ag and became activated. However, Ag aggregation in the lung vasculature was not necessary for anaphylaxis induction.
Collapse
Affiliation(s)
- Biliana Todorova
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Ophélie Godon
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Eva Conde
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Caitlin M. Gillis
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Bruno Iannascoli
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Odile Richard-Le Goff
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Daniel Fiole
- †Unité Biothérapies Anti-Infectieuses et Immunité, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
- ‡Unit of Human Histopathology and Animal Models, Institut Pasteur, Université de Paris, Paris, France
| | - Lubka T. Roumenina
- §Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | | | | | | | - Laurent L. Reber
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
- #Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Friederike Jönsson
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Pierre Bruhns
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| |
Collapse
|
14
|
Lama JK, Iijima K, Kobayashi T, Kita H. Blocking the inhibitory receptor programmed cell death 1 prevents allergic immune response and anaphylaxis in mice. J Allergy Clin Immunol 2022; 150:178-191.e9. [PMID: 35092762 PMCID: PMC9271539 DOI: 10.1016/j.jaci.2022.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Food allergy and acute anaphylaxis can be life-threatening. While T follicular helper (Tfh) cells play a pivotal role in the allergic immune responses, the immunologic mechanisms that regulate the production of antibodies (Abs) that mediate anaphylaxis are not fully understood. OBJECTIVE The aim of this study was to investigate the role of the inhibitory receptor programmed cell death protein 1 (PD-1), which is highly expressed on Tfh cells, in allergic immune responses using an animal model of peanut allergy and anaphylaxis. METHODS Naive wild-type mice were exposed to peanut flour intranasally and then challenged with peanut extract to induce systemic anaphylaxis. The roles of PD-1 were examined by blocking Abs and using gene-deficient animals. A hapten model and passive cutaneous anaphylaxis were used to characterize allergen-specific Abs. RESULTS Treatment with anti-PD-1 enhanced development of Tfh cells and germinal center B cells in mice exposed to peanut flour. Nonetheless, anti-PD-1 or its ligand fully protected mice from developing anaphylaxis. Anti-PD-1 treatment or genetic deficiency of PD-1 in CD4+ T cells inhibited production of peanut-specific IgE and increased the levels of IgG. The passive cutaneous anaphylaxis showed that peanut-specific Abs generated in anti-PD-1-treated animals prevented, rather than provoked, anaphylaxis when transferred to naive animals. Anti-PD-1 promoted production of Abs with low affinity for an antigen in the hapten model. CONCLUSION Blockade of the pathway between PD-1 and its ligand is protective against allergic immune responses. The direct interaction between Tfh cells and B cells may play a pivotal role in controlling Ab quality and clinical manifestation of allergic diseases.
Collapse
Affiliation(s)
- Jyoti K. Lama
- Department of Immunology, Mayo Clinic Rochester, Rochester, MN 55905 and Mayo Clinic Arizona, Scottsdale, AZ 85259,Immunology Program, Mayo Graduate School of Biomedical Sciences, Rochester, MN 55905 and Scottsdale, AZ 85259
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology and Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259
| | - Takao Kobayashi
- Division of Allergy, Asthma and Clinical Immunology and Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259
| | - Hirohito Kita
- Department of Immunology, Mayo Clinic Rochester, Rochester, MN 55905 and Mayo Clinic Arizona, Scottsdale, AZ 85259,Division of Allergy, Asthma and Clinical Immunology and Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259
| |
Collapse
|
15
|
Luo X, Chen J, Yang H, Hu X, Alphonse MP, Shen Y, Kawakami Y, Zhou X, Tu W, Kawakami T, Wan M, Archer NK, Wang H, Gao P. Dendritic cell immunoreceptor drives atopic dermatitis by modulating oxidized CaMKII-involved mast cell activation. JCI Insight 2022; 7:152559. [PMID: 35113811 PMCID: PMC8983143 DOI: 10.1172/jci.insight.152559] [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: 06/21/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Allergens have been identified as potential triggers in patients with atopic dermatitis (AD). AD patients are highly sensitive to cockroach allergen. The underlying mechanism, however, remains undetermined. Here, we established a cockroach allergen-induced AD-like mouse model and demonstrated that repeated exposure to cockroach allergen led to aggravated mouse skin inflammation, characterized by increased type 2 immunity, type 2 innate lymphoid cells (ILC2s), and mast cells. Increased skin mast cells were also observed in AD patients. AD mice with mast cell-deficient mice (kitW-sh/W-sh) showed diminished skin inflammation, suggesting that mast cells are required in allergen-induced skin inflammation. Furthermore, dendritic cell immuno-receptor (DCIR) is up-regulated in skin mast cells of AD patients and mediates allergen binding and uptake. DCIR-/- mice or reconstituted kitW-sh/W-sh mice with DCIR-/- mast cells showed a significant reduction in AD-like inflammation. Both in vitro and in vivo analyses demonstrated that DCIR-/- mast cells had reduced IgE-mediated mast cell activation and passive cutaneous anaphylaxis. Mechanistically, DCIR regulates allergen-induced IgE-mediated mast cell ROS generation and oxidation of calmodulin kinase II (ox-CaMKII). ROS-resistant CaMKII (MM-VVδ) prevents allergen-induced mast cell activation and inflammatory mediator release. Our study reveals a previously unrecognized DCIR-ROS-CaMKII axis that controls allergen-induced mast cell activation and AD-like inflammation.
Collapse
Affiliation(s)
- Xiaoyan Luo
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Jingsi Chen
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Huan Yang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yuko Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Xiaoying Zhou
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Mei Wan
- Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Hua Wang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| |
Collapse
|
16
|
Smeekens JM, Kulis MD. Mouse Models of Food Allergy in the Pursuit of Novel Treatment Modalities. FRONTIERS IN ALLERGY 2021; 2:810067. [PMID: 35387036 PMCID: PMC8974753 DOI: 10.3389/falgy.2021.810067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The prevalence of IgE-mediated food allergies has increased dramatically in the past three decades, now affecting up to 10% of the US population. IgE-mediated food allergy is an immunologic disease, involving a variety of cells, including B and T cells, mast cells, basophils, ILC2s, and epithelial cells. Mouse models of food allergy mimic the overall immunologic processes known to exist in humans. Due to the limitations of invasive sampling of human tissue and the similarities of the human and mouse immune systems, comprehensive pathogenesis studies of food allergy have been performed in mouse models. Mouse models have been effective in elucidating the roles of non-oral routes of sensitization and identifying key cells and molecules involved in allergic sensitization. Furthermore, the development of novel therapeutic approaches for food allergy has been accelerated through the use of pre-clinical mouse models. Despite the groundbreaking findings stemming from research in mice, there are continued efforts to improve the translational utility of these models. Here, we highlight the achievements in understanding food allergy development and efforts to bring novel treatment approaches into clinical trials.
Collapse
Affiliation(s)
- Johanna M. Smeekens
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
- *Correspondence: Johanna M. Smeekens
| | - Michael D. Kulis
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Alberca RW, Gomes E, Moretti EH, Russo M, Steiner AA. Naturally occurring hypothermia promotes survival in severe anaphylaxis. Immunol Lett 2021; 237:27-32. [PMID: 34245741 DOI: 10.1016/j.imlet.2021.07.002] [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: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 11/24/2022]
Abstract
Although hypothermia has received substantial attention as an indicator of severity in anaphylaxis, it has been neglected from the perspective of whether it could act as a disease-modifying factor in this condition. Here, the impact of naturally occurring (spontaneous) hypothermia on anaphylaxis was evaluated in a murine model of ovalbumin (OVA)-induced allergy. Nonextreme changes in the ambient temperature (Ta) were used to modulate the magnitude of spontaneous hypothermia. At a Ta of 24°C, challenge with OVA intraperitoneally or intravenously resulted in a rapid, transient fall in body core temperature, which reached its nadir 4-6°C below baseline in 30 min. This hypothermic response was largely attenuated when the mice were kept at a Ta of 34°C. The Ta-dependent attenuation of hypothermia resulted in a survival rate of only 30%, as opposed to survival of 100% in the condition that favored the development of hypothermia. The protective effect of hypothermia did not involve changes in the rate of mast cell degranulation, as assessed by the concentration of mast cell protease-1 in bodily fluids. On the other hand, hypothermia improved oxygenation of the brain and kidneys, as indicated by higher NAD+/NADH ratios. Therefore, it is plausible to propose that naturally occurring hypothermia makes organs more resistant to the anaphylactic insult.
Collapse
Affiliation(s)
- Ricardo W Alberca
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508, Brazil
| | - Eliane Gomes
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508, Brazil
| | - Eduardo H Moretti
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508, Brazil
| | - Momtchilo Russo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508, Brazil
| | - Alexandre A Steiner
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508, Brazil.
| |
Collapse
|
18
|
Cianferoni A. Non-IgE-mediated anaphylaxis. J Allergy Clin Immunol 2021; 147:1123-1131. [PMID: 33832694 DOI: 10.1016/j.jaci.2021.02.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022]
Abstract
Anaphylaxis is a rapidly evolving, acute, life-threatening reaction that occurs rapidly on contact with a trigger. Anaphylaxis is classically defined as an allergen-driven process that induces specific IgE and the activation of mast cells and basophils through the cross-linking of IgE receptors. However, it is clear that non-IgE-mediated pathways can induce symptoms indistinguishable from those of classic anaphylaxis, and their activation could explain the severity of IgE-mediated anaphylaxis. Indeed, mast cells and basophils can be activated by antibodies against IgE or their receptors, by molecules such as anaphylatoxins, or through G-coupled receptors. Some other allergens can induce antibodies of class IgG that can activate neutrophils to produce a molecule similar to histamine to induce anaphylaxis. Finally, some inflammatory mediators such as bradykinin or prostaglandin can also modulate mast cell and basophil activation as well as directly cause vasodilation and bronchoconstriction, resulting in anaphylaxis-like reactions.
Collapse
Affiliation(s)
- Antonella Cianferoni
- Perelman School of Medicine, University of Pennsylvania, Allergy and Immunology Division, The Children's Hospital of Philadelphia, Philadelphia, Pa.
| |
Collapse
|
19
|
Grayson MH. Evidence from mice that immunoglobulin E deficiency does not drive spontaneous malignancy. Ann Allergy Asthma Immunol 2021; 126:309. [PMID: 33608070 DOI: 10.1016/j.anai.2021.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Mitchell H Grayson
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio.
| |
Collapse
|
20
|
Michelet M, Balbino B, Guilleminault L, Reber LL. IgE in the pathophysiology and therapy of food allergy. Eur J Immunol 2021; 51:531-543. [PMID: 33527384 DOI: 10.1002/eji.202048833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022]
Abstract
Food allergy is becoming a major public health issue, with no regulatory approved therapy to date. Food allergy symptoms range from skin rash and gastrointestinal symptoms to anaphylaxis, a potentially fatal systemic allergic shock reaction. IgE antibodies are thought to contribute importantly to key features of food allergy and anaphylaxis, and measurement of allergen-specific IgE is fundamental in diagnosing food allergy. This review will discuss recent advances in the regulation of IgE production and IgE repertoires in food allergy. We will describe the current understanding of the role of IgE and its high-affinity receptor FcεRI in food allergy and anaphylaxis, by reviewing insights gained from analyses of mouse models. Finally, we will review data derived from clinical studies of the effect of anti-IgE therapeutic monoclonal antibodies (mAbs) in food allergy, and recent insight on the efficiency and mechanisms through which these mAbs block IgE effector functions.
Collapse
Affiliation(s)
- Marine Michelet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Pediatric Pneumo-allergology Department, Children's Hospital, University Hospital Centre of Toulouse, Toulouse, France
| | - Bianca Balbino
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERM, Paris, France
| | - Laurent Guilleminault
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Department of Respiratory Medicine and Allergic Diseases, University Hospital Centre of Toulouse, Toulouse, France
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERM, Paris, France
| |
Collapse
|
21
|
Aguilera-Lizarraga J, Florens MV, Viola MF, Jain P, Decraecker L, Appeltans I, Cuende-Estevez M, Fabre N, Van Beek K, Perna E, Balemans D, Stakenborg N, Theofanous S, Bosmans G, Mondelaers SU, Matteoli G, Ibiza Martínez S, Lopez-Lopez C, Jaramillo-Polanco J, Talavera K, Alpizar YA, Feyerabend TB, Rodewald HR, Farre R, Redegeld FA, Si J, Raes J, Breynaert C, Schrijvers R, Bosteels C, Lambrecht BN, Boyd SD, Hoh RA, Cabooter D, Nelis M, Augustijns P, Hendrix S, Strid J, Bisschops R, Reed DE, Vanner SJ, Denadai-Souza A, Wouters MM, Boeckxstaens GE. Local immune response to food antigens drives meal-induced abdominal pain. Nature 2021; 590:151-156. [PMID: 33442055 DOI: 10.1038/s41586-020-03118-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022]
Abstract
Up to 20% of people worldwide develop gastrointestinal symptoms following a meal1, leading to decreased quality of life, substantial morbidity and high medical costs. Although the interest of both the scientific and lay communities in this issue has increased markedly in recent years, with the worldwide introduction of gluten-free and other diets, the underlying mechanisms of food-induced abdominal complaints remain largely unknown. Here we show that a bacterial infection and bacterial toxins can trigger an immune response that leads to the production of dietary-antigen-specific IgE antibodies in mice, which are limited to the intestine. Following subsequent oral ingestion of the respective dietary antigen, an IgE- and mast-cell-dependent mechanism induced increased visceral pain. This aberrant pain signalling resulted from histamine receptor H1-mediated sensitization of visceral afferents. Moreover, injection of food antigens (gluten, wheat, soy and milk) into the rectosigmoid mucosa of patients with irritable bowel syndrome induced local oedema and mast cell activation. Our results identify and characterize a peripheral mechanism that underlies food-induced abdominal pain, thereby creating new possibilities for the treatment of irritable bowel syndrome and related abdominal pain disorders.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Morgane V Florens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Maria Francesca Viola
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Piyush Jain
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Lisse Decraecker
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Iris Appeltans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Maria Cuende-Estevez
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Naomi Fabre
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Kim Van Beek
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Eluisa Perna
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Dafne Balemans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Stavroula Theofanous
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Goele Bosmans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Stéphanie U Mondelaers
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Sales Ibiza Martínez
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Cintya Lopez-Lopez
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Karel Talavera
- Laboratory for Ion Channel Research, VIB Center for Brain and Disease Research, KU Leuven Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - Yeranddy A Alpizar
- Neuroscience Research group, BIOMED, Hasselt University, Hasselt, Belgium
| | | | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Ricard Farre
- Mucosal Permeability Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Jiyeon Si
- KU Leuven Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Leuven, Belgium.,VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | - Jeroen Raes
- KU Leuven Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Leuven, Belgium.,VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | - Christine Breynaert
- Allergy and Clinical Immunology Research Group, KU Leuven Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Rik Schrijvers
- Allergy and Clinical Immunology Research Group, KU Leuven Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Cédric Bosteels
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramona A Hoh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deirdre Cabooter
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Maxim Nelis
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Patrick Augustijns
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Raf Bisschops
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alexandre Denadai-Souza
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Mira M Wouters
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Guy E Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium.
| |
Collapse
|
22
|
Lamri Y, Vibhushan S, Pacreau E, Boedec E, Saidoune F, Mailleux A, Crestani B, Blank U, Benhamou M, Papo T, Daugas E, Sacré K, Charles N. Basophils and IgE contribute to mixed connective tissue disease development. J Allergy Clin Immunol 2020; 147:1478-1489.e11. [PMID: 33338538 DOI: 10.1016/j.jaci.2020.12.622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/20/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Mixed connective tissue disease (MCTD) is a rare and complex autoimmune disease that presents mixed features with other connective tissue diseases, such as systemic lupus erythematosus, systemic sclerosis, and myositis. It is characterized by high levels of anti-U1 small nuclear ribonucleoprotein 70k autoantibodies and a high incidence of life-threatening pulmonary involvement. The pathophysiology of MCTD is not well understood, and no specific treatment is yet available for the patients. Basophils and IgE play a role in the development of systemic lupus erythematosus and thus represent new therapeutic targets for systemic lupus erythematosus and other diseases involving basophils and IgE in their pathogenesis. OBJECTIVE We sought to investigate the role of basophils and IgE in the pathophysiology of MCTD. METHODS Basophil activation status and the presence of autoreactive IgE were assessed in peripheral blood of a cohort of patients with MCTD and in an MCTD-like mouse model. Basophil depletion and IgE-deficient animals were used to investigate the contribution of basophils and IgE in the lung pathology development of this mouse model. RESULTS Patients with MCTD have a peripheral basopenia and activated blood basophils overexpressing C-C chemokine receptor 3. Autoreactive IgE raised against the main MCTD autoantigen U1 small nuclear ribonucleoprotein 70k were found in nearly 80% of the patients from the cohort. Basophil activation and IgE anti-U1 small nuclear ribonucleoprotein 70k were also observed in the MCTD-like mouse model along with basophil accumulation in lymph nodes and lungs. Basophil depletion dampened lung pathology, and IgE deficiency prevented its development. CONCLUSIONS Basophils and IgE contribute to MCTD pathophysiology and represent new candidate therapeutic targets for patients with MCTD.
Collapse
Affiliation(s)
- Yasmine Lamri
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Shamila Vibhushan
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Emeline Pacreau
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Erwan Boedec
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Fanny Saidoune
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Arnaud Mailleux
- Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Université de Paris, INSERM UMR1152, Faculté de Médecine site Bichat, Paris, France
| | - Bruno Crestani
- Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Université de Paris, INSERM UMR1152, Faculté de Médecine site Bichat, Paris, France; Department of Pulmonology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Ulrich Blank
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Marc Benhamou
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Thomas Papo
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Eric Daugas
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Nephrology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Karim Sacré
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Nicolas Charles
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France.
| |
Collapse
|
23
|
Kanagaratham C, El Ansari YS, Lewis OL, Oettgen HC. IgE and IgG Antibodies as Regulators of Mast Cell and Basophil Functions in Food Allergy. Front Immunol 2020; 11:603050. [PMID: 33362785 PMCID: PMC7759531 DOI: 10.3389/fimmu.2020.603050] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Food allergy is a major health issue, affecting the lives of 8% of U.S. children and their families. There is an urgent need to identify the environmental and endogenous signals that induce and sustain allergic responses to ingested allergens. Acute reactions to foods are triggered by the activation of mast cells and basophils, both of which release inflammatory mediators that lead to a range of clinical manifestations, including gastrointestinal, cutaneous, and respiratory reactions as well as systemic anaphylaxis. Both of these innate effector cell types express the high affinity IgE receptor, FcϵRI, on their surface and are armed for adaptive antigen recognition by very-tightly bound IgE antibodies which, when cross-linked by polyvalent allergen, trigger degranulation. These cells also express inhibitory receptors, including the IgG Fc receptor, FcγRIIb, that suppress their IgE-mediated activation. Recent studies have shown that natural resolution of food allergies is associated with increasing food-specific IgG levels. Furthermore, oral immunotherapy, the sequential administration of incrementally increasing doses of food allergen, is accompanied by the strong induction of allergen-specific IgG antibodies in both human subjects and murine models. These can deliver inhibitory signals via FcγRIIb that block IgE-induced immediate food reactions. In addition to their role in mediating immediate hypersensitivity reactions, mast cells and basophils serve separate but critical functions as adjuvants for type 2 immunity in food allergy. Mast cells and basophils, activated by IgE, are key sources of IL-4 that tilts the immune balance away from tolerance and towards type 2 immunity by promoting the induction of Th2 cells along with the innate effectors of type 2 immunity, ILC2s, while suppressing the development of regulatory T cells and driving their subversion to a pathogenic pro-Th2 phenotype. This adjuvant effect of mast cells and basophils is suppressed when inhibitory signals are delivered by IgG antibodies signaling via FcγRIIb. This review summarizes current understanding of the immunoregulatory effects of mast cells and basophils and how these functions are modulated by IgE and IgG antibodies. Understanding these pathways could provide important insights into innovative strategies for preventing and/or reversing food allergy in patients.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Owen L. Lewis
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - 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
| |
Collapse
|
24
|
Bruton K, Koenig JFE, Phelps A, Jordana M. Perturbations to Homeostasis in Experimental Models Revealed Innate Pathways Driving Food Allergy. Front Immunol 2020; 11:603272. [PMID: 33362786 PMCID: PMC7758527 DOI: 10.3389/fimmu.2020.603272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
While type 2 immunity has been conventionally viewed as beneficial against helminths, venoms, and poisons, and harmful in allergy, contemporary research has uncovered its critical role in the maintenance of homeostasis. The initiation of a type 2 immune response involves an intricate crosstalk between structural and immune cells. Structural cells react to physical and chemical tissue perturbations by secreting alarmins, which signal the innate immune system to restore homeostasis. This pathway acts autonomously in the context of sterile injury and in the presence of foreign antigen initiates an adaptive Th2 response that is beneficial in the context of venoms, toxins, and helminths, but not food allergens. The investigation of the triggers and mechanisms underlying food allergic sensitization in humans is elusive because sensitization is a silent process. Therefore, the central construct driving food allergy modeling is based on introducing perturbations of tissue homeostasis along with an allergen which will result in an immunological and clinical phenotype that is consistent with that observed in humans. The collective evidence from multiple models has revealed the pre-eminent role of innate cells and molecules in the elicitation of allergic sensitization. We posit that, with the expanding use of technologies capable of producing formidable datasets, models of food allergy will continue to have an indispensable role to delineate mechanisms and establish causal relationships.
Collapse
Affiliation(s)
| | | | | | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton ON, Canada
| |
Collapse
|
25
|
El Ansari YS, Kanagaratham C, Lewis OL, Oettgen HC. IgE and mast cells: The endogenous adjuvant. Adv Immunol 2020; 148:93-153. [PMID: 33190734 DOI: 10.1016/bs.ai.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are most familiar as the effectors of type I hypersensitivity reactions including anaphylaxis. It is becoming clear however that this pair has important immunomodulatory effects on innate and adaptive cells of the immune system. In this purview, they act as endogenous adjuvants to ignite evolving immune responses, promote the transition of allergic disease into chronic illness and disrupt the development of active mechanisms of tolerance to ingested foods. Suppression of IgE-mediated mast cell activation can be exerted by molecules targeting IgE, FcɛRI or signaling kinases including Syk, or by IgG antibodies acting via inhibitory Fcγ receptors. In 2015 we reviewed the evidence for the adjuvant functions of mast cells. This update includes the original text, incorporates some important developments in the field over the past five years and discusses how interventions targeting these pathways might have promise in the development of strategies to treat allergic disease.
Collapse
Affiliation(s)
- Yasmeen S El Ansari
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Owen L Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
26
|
IgE Effector Mechanisms, in Concert with Mast Cells, Contribute to Acquired Host Defense against Staphylococcusaureus. Immunity 2020; 53:793-804.e9. [PMID: 32910906 DOI: 10.1016/j.immuni.2020.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 06/02/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
Allergies are considered to represent mal-directed type 2 immune responses against mostly innocuous exogenous compounds. Immunoglobulin E (IgE) antibodies are a characteristic feature of allergies and mediate hypersensitivity against allergens through activation of effector cells, particularly mast cells (MCs). Although the physiological functions of this dangerous branch of immunity have remained enigmatic, recent evidence shows that allergic immune reactions can help to protect against the toxicity of venoms. Because bacteria are a potent alternative source of toxins, we assessed the possible role of allergy-like type 2 immunity in antibacterial host defense. We discovered that the adaptive immune response against Staphylococcus aureus (SA) skin infection substantially improved systemic host defense against secondary SA infections in mice. Moreover, this acquired protection depended on IgE effector mechanisms and MCs. Importantly, our results reveal a previously unknown physiological function of allergic immune responses, IgE antibodies, and MCs in host defense against a pathogenic bacterium.
Collapse
|
27
|
Schmitt ME, Lutz J, Haase P, Bösl MR, Wienands J, Engels N, Voehringer D. The B-cell antigen receptor of IgE-switched plasma cells regulates memory IgE responses. J Allergy Clin Immunol 2020; 146:642-651.e5. [DOI: 10.1016/j.jaci.2020.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/25/2022]
|
28
|
Paranjape A, Haque TT, Kiwanuka KN, Qayum AA, Barnstein BO, Finkelman FD, Nigrovic PA, Ryan JJ. The Fyn-Stat5 cascade is required for Fcγ receptor-mediated mast cell function. Cell Immunol 2020; 356:104134. [PMID: 32862025 DOI: 10.1016/j.cellimm.2020.104134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023]
Abstract
Mast cells, well established effectors in allergic disease, can be activated by numerous stimuli. We previously found that the Fyn-Stat5B pathway is critical for FcεRI-stimulated mast cell function. Because IgG receptors employ similar signaling pathways, we investigated Fyn-Stat5B function downstream of FcγR. We report that FcγR elicits Fyn-dependent Stat5B tyrosine phosphorylation in mast cells. As we previously found for Fyn kinase, Stat5B is indispensable for IgG-mediated mast cell cytokine expression and secretion. However, Stat5B KO macrophages responded normally to FcγR signaling, indicating a lineage-restricted role for Stat5B. This was consistent in vivo, since passive FcγR activation induced anaphylaxis in a macrophage-dominated response even when Stat5B was deleted. We further investigated this lineage restriction using the K/BxN model of inflammatory arthritis. This model exhibits a rapid and transient mast cell-dependent joint inflammation followed days later by a macrophage- and neutrophil-dependent response. Consistent with our hypothesis, Fyn or Stat5B deficiency did not protect mice from late joint swelling, but greatly reduced the early mast cell-dependent response. This was associated with decreased joint and plasma histamine. We conclude that Fyn-Stat5B is a linage-restricted pathway critical for IgG-mediated mast cell responses.
Collapse
Affiliation(s)
- Anuya Paranjape
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Tamara T Haque
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Kasalina N Kiwanuka
- Department of Biochemistry, Virginia Commonwealth University, Box 980614, Richmond, VA 23298-0614, USA
| | - Amina Abdul Qayum
- Department of Microbiology and Immunology, Virginia Commonwealth University, Box 980678, Richmond, VA 23298-0678, USA
| | - Brian O Barnstein
- Department of Biology, Virginia Commonwealth University, Box 842012, Richmond, VA 23284-2012, USA
| | - Fred D Finkelman
- Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Peter A Nigrovic
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Box 842012, Richmond, VA 23284-2012, USA.
| |
Collapse
|
29
|
Patel M, Narke D, Kurade M, Frey KM, Rajalingam S, Siddiquee A, Mustafa SJ, Ledent C, Ponnoth DS. Limonene-induced activation of A 2A adenosine receptors reduces airway inflammation and reactivity in a mouse model of asthma. Purinergic Signal 2020; 16:415-426. [PMID: 32789792 DOI: 10.1007/s11302-020-09697-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/19/2020] [Indexed: 02/02/2023] Open
Abstract
Animal models of asthma have shown that limonene, a naturally occurring terpene in citrus fruits, can reduce inflammation and airway reactivity. However, the mechanism of these effects is unknown. We first performed computational and molecular docking analyses that showed limonene could bind to both A2A and A2B receptors. The pharmacological studies were carried out with A2A adenosine receptor knock-out (A2AKO) and wild-type (WT) mice using ovalbumin (OVA) to generate the asthma phenotype. We investigated the effects of limonene on lung inflammation and airway responsiveness to methacholine (MCh) and NECA (nonselective adenosine analog) by administering limonene as an inhalation prior to OVA aerosol challenges in one group of allergic mice for both WT and KO. In whole-body plethysmography studies, we observed that airway responsiveness to MCh in WT SEN group was significantly lowered upon limonene treatment but no effect was observed in A2AKO. Limonene also attenuated NECA-induced airway responsiveness in WT allergic mice with no effect being observed in A2AKO groups. Differential BAL analysis showed that limonene reduced levels of eosinophils in allergic WT mice but not in A2AKO. However, limonene reduced neutrophils in sensitized A2AKO mice, suggesting that it may activate A2B receptors as well. These data indicate that limonene-induced reduction in airway inflammation and airway reactivity occurs mainly via activation of A2AAR but A2B receptors may also play a supporting role.
Collapse
Affiliation(s)
- Mehaben Patel
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Deven Narke
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Mangesh Kurade
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Kathleen M Frey
- Fairleigh Dickinson University School of Pharmacy and Health Sciences, Teaneck, NJ, USA
| | - Sahith Rajalingam
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Armaan Siddiquee
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | | | - Dovenia S Ponnoth
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Island University, Brooklyn, NY, USA. .,Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, WV, Lewisburg, USA.
| |
Collapse
|
30
|
Desheva Y, Mamontov A, Petkova N, Karev V, Nazarov P. Mast cell degranulation and histamine release during A/H5N1 influenza infection in influenza-sensitized mice. Life Sci 2020; 258:118230. [PMID: 32777303 PMCID: PMC7413848 DOI: 10.1016/j.lfs.2020.118230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022]
Abstract
Here we evaluate the role of mast cells in infection with influenza A/H5N1 virus in immunized mice. CBA mice were immunized intramuscularly with formalin-inactivated A/Vietnam/1194/2004 (H5N1)NIBRG-14 (H5N1). Serum samples were obtained on days 7, 12, 14, 21 after immunization. At day 14, the mice were infected intranasally with the A/Indonesia/5/2005 (H5N1)IDCDC-RG2 (H5N1) influenza virus with half of the animals receiving a mixture of the antihistamines. 67% of the vaccinated mice were protected from the lethality compared to 43% in the PBS-immunized group. Administration of antihistamines increased survival up to 85%–95%. Immunohistochemical examination using CD117 staining of the lungs demonstrated a larger quantity of activated mast cells after infection of immunized mice compared to mock-immunized mice. This was correlated to increased histamine level in the lungs and blood. Our experimental results suggest the involvement of mast cells and the histamine they produce in the pathogenesis of influenza infection in case of incomplete formation of the immune response to vaccination and mismatch of the vaccine and infection influenza viruses.
Collapse
Affiliation(s)
- Yulia Desheva
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", Acad. Pavlov's str., 12, 197376 Saint Petersburg, Russian Federation.
| | - Andrey Mamontov
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", Acad. Pavlov's str., 12, 197376 Saint Petersburg, Russian Federation
| | - Nadezhda Petkova
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", Acad. Pavlov's str., 12, 197376 Saint Petersburg, Russian Federation
| | - Vadim Karev
- Federal State Budgetary Institution "Research institute of children's diseases", 9 Professor Popov's Str., 197022 Saint Petersburg, Russian Federation
| | - Peter Nazarov
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", Acad. Pavlov's str., 12, 197376 Saint Petersburg, Russian Federation
| |
Collapse
|
31
|
Castells MC, Li JT. Anaphylaxis: Parts Unknown. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:1216-1218. [PMID: 32276690 DOI: 10.1016/j.jaip.2020.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 06/11/2023]
Affiliation(s)
- Mariana C Castells
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
| | - James T Li
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minn
| |
Collapse
|
32
|
Galli SJ, Metz M, Starkl P, Marichal T, Tsai M. Mast cells and IgE in defense against lethality of venoms: Possible "benefit" of allergy[]. ALLERGO JOURNAL INTERNATIONAL 2020; 29:46-62. [PMID: 33224714 PMCID: PMC7673288 DOI: 10.1007/s40629-020-00118-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/20/2019] [Indexed: 01/15/2023]
Abstract
Physicians think of mast cells and IgE primarily in the context of allergic disorders, including fatal anaphylaxis. This 'bad side' of mast cells and IgE is so well accepted that it can be difficult to think of them in other contexts, particularly those in which they may have beneficial functions. However, there is evidence that mast cells and IgE, as well as basophils (circulating granulocytes whose functions partially overlap with those of mast cells), can contribute to host defense as components of adaptive type 2 immune responses to helminths, ticks and certain other parasites. Accordingly, allergies often are conceptualized as "misdirected" type 2 immune responses, in which IgE antibodies are produced against any of a diverse group of apparently harmless antigens, and against components of animal venoms. Indeed, certain unfortunate patients who have become sensitized to venoms develop severe IgE-associated allergic reactions, including fatal anaphylaxis, upon subsequent venom exposure. In this review, we will describe evidence that mast cells can enhance innate resistance, and survival, to challenge with reptile or arthropod venoms during a first exposure to such venoms. We also will discuss findings indicating that, in mice surviving an initial encounter with venom, acquired type 2 immune responses, IgE antibodies, the high affinity IgE receptor (FcεRI), and mast cells can contribute to acquired resistance to the lethal effects of both honeybee venom and Russell's viper venom. These findings support the hypothesis that mast cells and IgE can help protect the host against venoms and perhaps other noxious substances.
Collapse
Affiliation(s)
- Stephen J. Galli
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, 94305; USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Martin Metz
- Department of Dermatology and Allergy, Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Department of Medicine 1, Laboratory of Infection Biology, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Mindy Tsai
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, 94305; USA
| |
Collapse
|
33
|
Galli SJ, Metz M, Starkl P, Marichal T, Tsai M. Mast cells and IgE in defense against lethality of venoms: Possible "benefit" of allergy*. ALLERGO JOURNAL 2020. [DOI: 10.1007/s15007-020-0746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
34
|
Jiménez‐Saiz R. Drug-induced IgG-neutrophil-mediated anaphylaxis in humans: Uncovered! Allergy 2020; 75:484-485. [PMID: 31736087 DOI: 10.1111/all.14118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Rodrigo Jiménez‐Saiz
- Department of Immunology & Oncology Centro Nacional de Biotecnología (CNB)‐CSIC Madrid Spain
- McMaster Immunology Research Centre (MIRC) Department of Pathology & Molecular Medicine McMaster University Hamilton ON Canada
| |
Collapse
|
35
|
Zhang X, Li J, Luo S, Wang M, Huang Q, Deng Z, de Febbo C, Daoui A, Liew PX, Sukhova GK, Metso J, Jauhiainen M, Shi GP, Guo J. IgE Contributes to Atherosclerosis and Obesity by Affecting Macrophage Polarization, Macrophage Protein Network, and Foam Cell Formation. Arterioscler Thromb Vasc Biol 2020; 40:597-610. [PMID: 31996021 DOI: 10.1161/atvbaha.119.313744] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE By binding to its high-affinity receptor FcεR1, IgE activates mast cells, macrophages, and other inflammatory and vascular cells. Recent studies support an essential role of IgE in cardiometabolic diseases. Plasma IgE level is an independent predictor of human coronary heart disease. Yet, a direct role of IgE and its mechanisms in cardiometabolic diseases remain incompletely understood. Approach and Results: Using atherosclerosis prone Apoe-/- mice and IgE-deficient Ige-/- mice, we demonstrated that IgE deficiency reduced atherosclerosis lesion burden, lesion lipid deposition, smooth muscle cell and endothelial cell contents, chemokine MCP (monocyte chemoattractant protein)-1 expression and macrophage accumulation. IgE deficiency also reduced bodyweight gain and increased glucose and insulin sensitivities with significantly reduced plasma cholesterol, triglyceride, insulin, and inflammatory cytokines and chemokines, including IL (interleukin)-6, IFN (interferon)-γ, and MCP-1. From atherosclerotic lesions and peritoneal macrophages from Apoe-/-Ige-/- mice that consumed an atherogenic diet, we detected reduced expression of M1 macrophage markers (CD68, MCP-1, TNF [tumor necrosis factor]-α, IL-6, and iNOS [inducible nitric oxide synthase]) but increased expression of M2 macrophage markers (Arg [arginase]-1 and IL-10) and macrophage-sterol-responsive-network molecules (complement C3, lipoprotein lipase, LDLR [low-density lipoprotein receptor]-related protein 1, and TFR [transferrin]) that suppress macrophage foam cell formation. These IgE activities can be reproduced in bone marrow-derived macrophages from wild-type mice, but muted in cells from FcεR1-deficient mice, or blocked by anti-IgE antibody or complement C3 deficiency. CONCLUSIONS IgE deficiency protects mice from diet-induced atherosclerosis, obesity, glucose tolerance, and insulin resistance by regulating macrophage polarization, macrophage-sterol-responsive-network gene expression, and foam cell formation.
Collapse
Affiliation(s)
- Xian Zhang
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Jie Li
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.).,Department of Geriatrics, National Key Clinic Specialty, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China (J.L.)
| | - Songyuan Luo
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Minjie Wang
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Qin Huang
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Zhiyong Deng
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Caroline de Febbo
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Aida Daoui
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Pei Xiong Liew
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Galina K Sukhova
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Jari Metso
- Minerva Foundation Institute for Medical Research, National Institute for Health and Welfare, Genomics and Biobank Unit, Biomedicum 2U, Helsinki, Finland (J.M., M.J.)
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, National Institute for Health and Welfare, Genomics and Biobank Unit, Biomedicum 2U, Helsinki, Finland (J.M., M.J.)
| | - Guo-Ping Shi
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.)
| | - Junli Guo
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (X.Z., J.L., S.L., M.W., Q.H., Z.D., C.d.F., A.D., P.X.L., G.K.S., G.-P.S., J.G.).,Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou 571199, China (J.G.)
| |
Collapse
|
36
|
Hayes MD, Ward S, Crawford G, Seoane RC, Jackson WD, Kipling D, Voehringer D, Dunn-Walters D, Strid J. Inflammation-induced IgE promotes epithelial hyperplasia and tumour growth. eLife 2020; 9:e51862. [PMID: 31931959 PMCID: PMC6959995 DOI: 10.7554/elife.51862] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 12/21/2022] Open
Abstract
IgE is the least abundant circulating antibody class but is constitutively present in healthy tissues bound to resident cells via its high-affinity receptor, FcεRI. The physiological role of endogenous IgE antibodies is unclear but it has been suggested that they provide host protection against a variety of noxious environmental substances and parasitic infections at epithelial barrier surfaces. Here we show, in mice, that skin inflammation enhances levels of IgE antibodies that have natural specificities and a repertoire, VDJ rearrangements and CDRH3 characteristics similar to those of IgE antibodies in healthy tissue. IgE-bearing basophils are recruited to inflamed skin via CXCL12 and thymic stromal lymphopoietin (TSLP)/IL-3-dependent upregulation of CXCR4. In the inflamed skin, IgE/FcεRI-signalling in basophils promotes epithelial cell growth and differentiation, partly through histamine engagement of H1R and H4R. Furthermore, this IgE response strongly drives tumour outgrowth of epithelial cells harbouring oncogenic mutation. These findings indicate that natural IgE antibodies support skin barrier defences, but that during chronic tissue inflammation this role may be subverted to promote tumour growth.
Collapse
Affiliation(s)
- Mark David Hayes
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| | - Sophie Ward
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| | - Greg Crawford
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| | - Rocio Castro Seoane
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| | - William David Jackson
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| | - David Kipling
- Division of Cancer and Genetics, School of MedicineCardiff UniversityCardiffUnited Kingdom
| | - David Voehringer
- Department of Infection BiologyUniversity Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU)ErlangenGermany
| | - Deborah Dunn-Walters
- Faculty of Health and Medical Sciences, School of Biosciences and MedicineUniversity of SurreyGuildfordUnited Kingdom
| | - Jessica Strid
- Department of Immunology and InflammationImperial College LondonLondonUnited Kingdom
| |
Collapse
|
37
|
Yu Z, Shibazaki M, Otsuka H, Takada H, Nakamura M, Endo Y. Dynamics of Platelet Behaviors as Defenders and Guardians: Accumulations in Liver, Lung, and Spleen in Mice. Biol Pharm Bull 2020; 42:1253-1267. [PMID: 31366863 DOI: 10.1248/bpb.b18-00975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic platelet behaviors in experimental animals are often assessed by infusion of isotope-labeled platelets and measuring them under anesthesia. However, such procedures alter, therefore may not reveal, real-life platelet behaviors. 5-Hydroxytryptamine (5HT or serotonin) is present within limited cell-types, including platelets. In our studies, by measuring 5HT as a platelet-marker in non-anesthetized mice, we identified stimulation- and time-dependent accumulations in liver, lung, and/or spleen as important systemic platelet behaviors. For example, intravenous, intraperitoneal, or intragingival injection of lipopolysaccharide (LPS, a cell-wall component of Gram-negative bacteria), interleukin (IL)-1, or tumor necrosis factor (TNF)-α induced hepatic platelet accumulation (HPA) and platelet translocation into the sinusoidal and perisinusoidal spaces or hepatocytes themselves. These events occurred "within a few hours" of the injection, caused hypoglycemia, and exhibited protective or causal effects on hepatitis. Intravenous injection of larger doses of LPS into normal mice, or intravenous antigen-challenge to sensitized mice, induced pulmonary platelet accumulation (PPA), as well as HPA. These reactions occurred "within a few min" of the LPS injection or antigen challenge and resulted in shock. Intravenous injection of 5HT or a catecholamine induced a rapid PPA "within 6 s." Intravenous LPS injection, within a minute, increased the pulmonary catecholamines that mediate the LPS-induced PPA. Macrophage-depletion from liver and spleen induced "day-scale" splenic platelet accumulation, suggesting the spleen is involved in clearing senescent platelets. These findings indicate the usefulness of 5HT as a marker of platelet behaviors, and provide a basis for a discussion of the roles of platelets as both "defenders" and "guardians."
Collapse
Affiliation(s)
- Zhiqian Yu
- Department of Disaster Psychiatry, International Research Institute for Disaster Science, Tohoku University
| | - Masahiro Shibazaki
- Department of Tumor Biology, Institute of Biomedical Sciences, Iwate Medical University
| | - Hirotada Otsuka
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University
| | - Haruhiko Takada
- Department of Microbiology and Immunology, Graduate School of Dentistry, Tohoku University
| | - Masanori Nakamura
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University
| | - Yasuo Endo
- Division of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Tohoku University
| |
Collapse
|
38
|
Ariza A, Torres MJ, Moreno-Aguilar C, Fernández-Santamaría R, Fernández TD. Early Biomarkers for Severe Drug Hypersensitivity Reactions. Curr Pharm Des 2019; 25:3829-3839. [DOI: 10.2174/1381612825666191107105440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023]
Abstract
Drug hypersensitivity reactions (DHRs) are typically classified into immediate and delayed reactions
based on the time interval between drug exposure and onset of symptoms. Clinical manifestations range from
mild to severe and life-threatening reactions. The most severe clinical entities are anaphylaxis and anaphylactic
shock for immediate reactions, and severe cutaneous adverse reactions such as Steven Johnson Syndrome and
Toxic Epidermal Necrolysis for delayed reactions. The diagnosis is complex and challenging, as drug provocation
tests and even skin tests can be very risky procedures, which makes them not recommended. Therefore, it is necessary
to search for useful early biomarkers to manage the diagnosis of these reactions. These biomarkers could
be useful to determine the clinical entity, but not to identify the culprit drug. Some of the currently available
biomarkers are few genetic associations of drug allergy with polymorphisms of human leukocyte antigen (HLA),
the detection of inflammatory and lipid mediators in serum, or the detection of cytokines, chemokines, and cytotoxic
markers in skin biopsies. In this literature review, it has been summarize the immunological mechanisms
involved in severe reactions, both immediate and delayed, and different early biomarkers: those currently used for
the diagnosis of these reactions as well as possible early biomarkers that could be useful with further studies to
standardize their clinical use.
Collapse
Affiliation(s)
- Adriana Ariza
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| | - Maria J. Torres
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| | - Carmen Moreno-Aguilar
- Immunology and Allergy Unit, IMIBICHospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Tahia D. Fernández
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| |
Collapse
|
39
|
Granger V, Peyneau M, Chollet-Martin S, de Chaisemartin L. Neutrophil Extracellular Traps in Autoimmunity and Allergy: Immune Complexes at Work. Front Immunol 2019; 10:2824. [PMID: 31849989 PMCID: PMC6901596 DOI: 10.3389/fimmu.2019.02824] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022] Open
Abstract
Neutrophil extracellular traps (NETs) have been initially described as main actors in host defense owing to their ability to immobilize and sometimes kill microorganisms. Subsequent studies have demonstrated their implication in the pathophysiology of various diseases, due to the toxic effects of their main components on surrounding tissues. Several distinct NETosis pathways have been described in response to various triggers. Among these triggers, IgG immune complexes (IC) play an important role since they induce robust NET release upon binding to activating FcγRs on neutrophils. Few in vitro studies have documented the mechanisms of IC-induced NET release and evidence about the partners involved is controversial. In vivo, animal models and clinical studies have strongly suggested the importance of IgG IC-induced NET release for autoimmunity and anaphylaxis. In this review, we will focus on two autoimmune diseases in which NETs are undoubtedly major players, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA). We will also discuss anaphylaxis as another example of disease recently associated with IC-induced NET release. Understanding the role of IC-induced NETs in these settings will pave the way for new diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Vanessa Granger
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Marine Peyneau
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Sylvie Chollet-Martin
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Luc de Chaisemartin
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
40
|
Eosinophils Mediate Basophil-Dependent Allergic Skin Inflammation in Mice. J Invest Dermatol 2019; 139:1957-1965.e2. [DOI: 10.1016/j.jid.2019.03.1129] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/28/2019] [Accepted: 03/13/2019] [Indexed: 01/22/2023]
|
41
|
MacDonald SM. History of Histamine-Releasing Factor (HRF)/Translationally Controlled Tumor Protein (TCTP) Including a Potential Therapeutic Target in Asthma and Allergy. Results Probl Cell Differ 2019; 64:291-308. [PMID: 29149416 DOI: 10.1007/978-3-319-67591-6_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histamine-releasing factor (HRF) also known as translationally controlled tumor protein (TCTP) is a highly conserved, ubiquitous protein that has both intracellular and extracellular functions. Here we will highlight the subcloning of the molecule, its clinical implications, as well as an inducible-transgenic mouse. Particular attention will be paid to its extracellular functioning and its potential role as a therapeutic target in asthma and allergy. The cells and the cytokines that are produced when stimulated or primed by HRF/TCTP will be detailed as well as the downstream signaling pathway that HRF/TCTP elicits. While it was originally thought that HRF/TCTP interacted with IgE, the finding that cells not binding IgE also respond to HRF/TCTP called this interaction into question. HRF/TCTP or at least its mouse counterpart appears to interact with some, but not all IgE and IgG molecules. HRF/TCTP has been shown to activate multiple human cells including basophils, eosinophils, T cells, and B cells. Since many of the cells that are activated by HRF/TCTP participate in the allergic response, the extracellular functions of HRF/TCTP could exacerbate the allergic, inflammatory cascade. Particularly exciting is that small molecule agonists of the phosphatase SHIP-1 have been shown to modulate the P13 kinase/AKT pathway and may control inflammatory disorders. This review discusses this possibility in light of HRF/TCTP.
Collapse
Affiliation(s)
- Susan M MacDonald
- The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3B.69, Baltimore, MD, 21224, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
42
|
Jönsson F, de Chaisemartin L, Granger V, Gouel-Chéron A, Gillis CM, Zhu Q, Dib F, Nicaise-Roland P, Ganneau C, Hurtado-Nedelec M, Paugam-Burtz C, Necib S, Keita-Meyer H, Le Dorze M, Cholley B, Langeron O, Jacob L, Plaud B, Fischler M, Sauvan C, Guinnepain MT, Montravers P, Aubier M, Bay S, Neukirch C, Tubach F, Longrois D, Chollet-Martin S, Bruhns P. An IgG-induced neutrophil activation pathway contributes to human drug-induced anaphylaxis. Sci Transl Med 2019; 11:11/500/eaat1479. [DOI: 10.1126/scitranslmed.aat1479] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 12/21/2018] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Anaphylaxis is a systemic acute hypersensitivity reaction that is considered to depend on allergen-specific immunoglobulin E (IgE) antibodies and histamine release by mast cells and basophils. Nevertheless, allergen-specific IgG antibodies have been proposed to contribute when the allergen is an abundant circulating large molecule, e.g., after infusions of therapeutic antibodies or dextran. Data from animal models demonstrate a pathway involving platelet-activating factor (PAF) release by monocytes/macrophages and neutrophils activated via their Fc gamma receptors (FcγRs). We hypothesized that such a pathway may also apply to small drugs and could be responsible for non–IgE-mediated anaphylaxis and influence anaphylaxis severity in humans. We prospectively conducted a multicentric study of 86 patients with suspected anaphylaxis to neuromuscular-blocking agents (NMBAs) during general anesthesia and 86 matched controls. We found that concentrations of anti-NMBA IgG and markers of FcγR activation, PAF release, and neutrophil activation correlated with anaphylaxis severity. Neutrophils underwent degranulation and NETosis early after anaphylaxis onset, and plasma-purified anti-NMBA IgG triggered neutrophil activation ex vivo in the presence of NMBA. Neutrophil activation could also be observed in patients lacking evidence of classical IgE-dependent anaphylaxis. This study supports the existence of an IgG-neutrophil pathway in human NMBA-induced anaphylaxis, which may aggravate anaphylaxis in combination with the IgE pathway or underlie anaphylaxis in the absence of specific IgE. These results reconcile clinical and experimental data on the role of antibody classes in anaphylaxis and could inform diagnostic approaches to NMBA-induced acute hypersensitivity reactions.
Collapse
|
43
|
Chiang SR, Lin CY, Chen DY, Tsai HF, Lin XC, Hsu TC, Tzang BS. The effects of human parvovirus VP1 unique region in a mouse model of allergic asthma. PLoS One 2019; 14:e0216799. [PMID: 31086415 PMCID: PMC6516678 DOI: 10.1371/journal.pone.0216799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/29/2019] [Indexed: 01/22/2023] Open
Abstract
Evidence has indicated that viral infection increases the risk of developing asthma. Although the association of human parvovirus B19 (B19V) or human bocavirus (HBoV) with respiratory diseases has been reported, little is known about the influence of the B19V-VP1u and HBoV-VP1u proteins on the symptoms of asthma. Herein, we investigated the systemic influence of subcutaneously injected B19V-VP1u and HBoV-VP1u recombinant proteins in an OVA-sensitized asthmatic mouse model. A significantly higher Penh ratio and IgE level were detected in the serum, bronchoalveolar lavage fluid (BALF) and the supernatant of a lymphocyte culture from mice treated with HBoV-VP1u or B19V-VP1u than in a lymphocyte culture from OVA-sensitized mice. Significantly higher levels of serum and BALF IgE, total IgG, IgG1, OVA-specific IgE and OVA-specific IgG1 were detected in mice treated with HBoV-VP1u or B19V-VP1u than in OVA-sensitized mice. Conversely, a significantly lower IgG2a level was detected in mice from the HBoV-VP1u or B19V-VP1u groups than in mice from the OVA group. The mice treated with HBoV-VP1u or B19V-VP1u exhibited more significant lung inflammatory indices, including elevated serum and BALF IL-4, IL-5, IL-10 and IL-13 levels; BALF lymphocyte, neutrophil and eosinophil counts, MMP-9 and MMP-2 activity; and the amount of lymphocyte infiltration, relative to those in the control mice or in those sensitized with OVA. These findings demonstrate that the subcutaneous injection of HBoV-VP1u or B19V-VP1u proteins in OVA-sensitized mice result in elevated asthmatic indices and suggest that human parvoviruses may increase the risk of developing airway inflammation in a mouse model of asthma.
Collapse
Affiliation(s)
- Shyh-Ren Chiang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
- Department of General Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, R.O.C
| | - Chia-Yun Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
- Rheumatic Diseases Research Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
- School of Medicine, China Medical University, Taichung, Taiwan, R.O.C
| | - Hui-Fang Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taiwan, R.O.C
| | - Xin-Ci Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Tsai-Ching Hsu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
- Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- * E-mail: (BST); (TCH)
| | - Bor-Show Tzang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
- Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- * E-mail: (BST); (TCH)
| |
Collapse
|
44
|
Ayakannu R, Abdullah NA, Radhakrishnan AK, Lechimi Raj V, Liam CK. Relationship between various cytokines implicated in asthma. Hum Immunol 2019; 80:755-763. [PMID: 31054782 DOI: 10.1016/j.humimm.2019.04.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023]
Abstract
Asthma is a complex disorder involving immunologic, environmental, genetic and other factors. Today, asthma is the most common disease encountered in clinical medicine in both children and adults worldwide. Asthma is characterized by increased responsiveness of the tracheobronchial tree resulting in chronic swelling and inflammation of the airways recognized to be controlled by the T-helper 2 (Th2) lymphocytes, which secrete cytokines to increase the production of IgE by B cells. There are many cytokines implicated in the development of the chronic inflammatory processes that are often observed in asthma. Ultimately, these cytokines cause the release of mediators such as histamine and leukotrienes (LT), which in turn promote airway remodeling, bronchial hyperresponsiveness and bronchoconstriction. The CD4+ T-lymphocytes from the airways of asthmatics express a panel of cytokines that represent the Th2 cells. The knowledge derived from numerous experimental and clinical studies have allowed physicians and scientists to understand the normal functions of these cytokines and their roles in the pathogenesis of asthma. The main focus of this review is to accentuate the relationship between various cytokines implicated in human asthma. However, some key findings from animal models will be highlighted to support the discoveries from clinical studies.
Collapse
Affiliation(s)
- Rathimalar Ayakannu
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - N A Abdullah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine, Monash University Malaysia, Jalan Lagoon, 47500 Bandar Sunway, Selangor, Malaysia
| | - Vijaya Lechimi Raj
- Department of Pharmacology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Selangor, Malaysia
| | - C K Liam
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Shilovskiy IP, Sundukova MS, Babakhin АА, Gaisina AR, Maerle AV, Sergeev IV, Nikolskiy AA, Barvinckaya ED, Kovchina VI, Kudlay DA, Nikonova AA, Khaitov MR. Experimental protocol for development of adjuvant-free murine chronic model of allergic asthma. J Immunol Methods 2019; 468:10-19. [PMID: 30880263 DOI: 10.1016/j.jim.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 03/01/2019] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mouse models of allergic asthma play a crucial role in exploring of asthma pathogenesis and testing of novel anti-inflammatory drugs. Widely used acute asthma models usually developed with adjuvant (aluminum hydroxide (alum)) do not reproduce one of the main asthma feature - airway remodeling while chronic asthma model mimic the pathophysiology of human disease. Moreover, the use of alum causes distress in experimental animals and impedes the test of adjuvant-containing drugs. In this study, we aimed to develop a chronic adjuvant-free asthma model with pronounced asthmatic phenotype. METHODS Female BALB/c mice were divided into 3 groups. The first group was sensitized with intraperitoneal injections of ovalbumin (OVA) emulsified in aluminum hydroxide on days 0, 14, 28 followed by two stages of intranasally challenge with OVA on days 41-43 and 62-64. The second group was subcutaneously sensitized with the same dose of OVA without adjuvant and challenged on the same days. The third group (negative control) included mice which did not received any kind of treatment (i.e. sensitization and challenge). Serum levels of OVA-specific IgE, IgG2a and IgG1 antibodies were detected by ELISA. Airway hyper-responsiveness was measured by non-invasive plethysmography on days 44 and 65. Bronchoalveolar lavage fluids (BALF) sampled in all groups on days 45 and 66 were analyzed by light microscopy. The left lung was removed for histological analysis. The IL-4 and IFNγ mRNA expression in BALF cells was evaluated by RT-PCR. RESULTS The OVA-specific IgE antibody response was two-fold increased in mice from adjuvant-free group compared to the adjuvant group that reflects reorientation of immune response towards Th2 phenotype. At the same time, the level of OVA-specific IgG1 and IgG2a antibodies was increased in the adjuvant group. Airway hyperresponsiveness to methacholine in mice of both experimental groups was two-fold higher than in control. Analysis of cell composition in BAL has shown a significant increase in eosinophil count in both experimental groups that indicate the development of allergic inflammation. Lung histology revealed airway remodeling in both experimental groups including goblet cell hyperplasia/metaplasia, thickening of airway walls, collagen deposition in the wall of distal airways. Additionally, the tendency to develop hypertrophy of bronchial smooth muscle layer was observed. Study of gene expression in BAL cells revealed the increase of IL-4 level in both adjuvant and adjuvant-free groups while IFNγ expression in both experimental groups was similar to control group. CONCLUSION We have developed a chronic adjuvant-free mouse asthma model which possesses all necessary features of the disease including airway remodeling and is more suitable for pre-clinical evaluation of novel therapeutic approaches including adjuvant-containing drugs.
Collapse
Affiliation(s)
- I P Shilovskiy
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia.
| | - M S Sundukova
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - А А Babakhin
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - A R Gaisina
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - A V Maerle
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - I V Sergeev
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - A A Nikolskiy
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - E D Barvinckaya
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - V I Kovchina
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - D A Kudlay
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| | - A A Nikonova
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia; Mechnikov Research Institute for vaccines and sera, 105064, 5A, M. Kazenny Per, Moscow, Russia
| | - M R Khaitov
- National Research Center - Institute of immunology of Federal Medico-Biological Agency, 115478, 24, Kashirskoye Shosse, Moscow, Russia
| |
Collapse
|
46
|
Kow ASF, Chik A, Soo KM, Khoo LW, Abas F, Tham CL. Identification of Soluble Mediators in IgG-Mediated Anaphylaxis via Fcγ Receptor: A Meta-Analysis. Front Immunol 2019; 10:190. [PMID: 30809224 PMCID: PMC6379333 DOI: 10.3389/fimmu.2019.00190] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/22/2019] [Indexed: 01/14/2023] Open
Abstract
Background: Anaphylaxis is an acute and life-threatening allergic response. Classically and most commonly, it can be mediated by the crosslinking of allergens to immunoglobulin E (IgE)- high affinity IgE receptor (FcεRI) complex found mostly on mast cells. However, there is another pathway of anaphylaxis that is less well-studied. This pathway known as the alternative pathway is mediated by IgG and its Fc gamma receptor (Fcγ). Though it was not documented in human anaphylaxis, a few studies have found that IgG-mediated anaphylaxis can happen as demonstrated in rodent models of anaphylaxis. In these studies, a variety of soluble mediators were being evaluated and they differ from each study which causes confusion in the suitability, and reliability of choice of soluble mediators to be analyzed for diagnosis or therapeutic purposes. Hence, the objective of this meta-analysis is to identify the potential soluble mediators that are involved in an IgG-mediated anaphylaxis reaction. Methods: Studies related to IgG-mediated anaphylaxis were sourced from five search engines namely PubMed, Scopus, Ovid, Cochrane Library, and Center for Agricultural Bioscience International (CABI) regardless of publication year. Relevant studies were then reviewed based on specific inclusion factors. The means and standard deviations of each soluble mediator studied were then extracted using ImageJ or Get Data Graph Digitiser software and the data were subjected to meta-analysis. Results: From our findings, we found that histamine, serotonin, platelet activating factor (PAF), β-hexosaminidase, leukotriene C4 (LTC4), mucosal mast cell protease-1 (MMCP-1), interleukins (IL)-4,−6, and−13; tumor necrosis factor alpha (TNF-α), and macrophage inflammatory protein-1α (MIP-1α) were often being analyzed. Out of these soluble mediators, histamine, PAF, β-hexosaminidase, IL-6, and−13, MIP-1α and TNF-α were more significant with positive effect size and p < 0.001. As study effect was relatively small, we performed publication bias and found that there was publication bias and this could be due to the small sample size studied. Conclusion: As such, we proposed that through meta-analysis, the potential soluble mediators involved in rodent IgG-mediated anaphylaxis to be histamine, PAF, β-hexosaminidase, IL-6 and−13 and MIP-1α, and TNF-α but will require further studies with larger sample size.
Collapse
Affiliation(s)
- Audrey Siew Foong Kow
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Azirah Chik
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Kuan-Meng Soo
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Leng Wei Khoo
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia.,Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
47
|
Moñino-Romero S, Erkert L, Schmidthaler K, Diesner SC, Sallis BF, Pennington L, Jardetzky T, Oettgen HC, Bohle B, Fiebiger E, Szépfalusi Z. The soluble isoform of human FcɛRI is an endogenous inhibitor of IgE-mediated mast cell responses. Allergy 2019; 74:236-245. [PMID: 30030936 DOI: 10.1111/all.13567] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/01/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The soluble isoform of FcɛRI, the high-affinity IgE receptor (sFcεRI), is a protein of the IgE network with poorly defined functions. OBJECTIVE To define cellular sources and signals that result in the production of human sFcεRI and study its in vivo functions. METHODS FcεRI-transfected human cell lines (MelJuso), human monocyte-derived dendritic cells (moDCs), and murine bone marrow-derived mast cells (MC) were stimulated by FcεRI cross-linking and release of sFcεRI was analyzed (ELISA, Western Blot). Lysosomal-associated membrane protein 1 degranulation assays and human basophil activation tests (BATs) were used to study IgE-dependent activation. Recombinant sFcεRI (rsFcεRI) was used to assess its role in murine models of anaphylaxis with WT (wild-type) and IgE-/- (IgE-deficient) mice. RESULTS Antigen-specific cross-linking of IgE-loaded FcɛRI on MelJuso cells that express the trimeric or tetrameric receptor isoform induced the production of sFcεRI. Using MCs and moDCs, we confirmed that IgE/FcɛRI activation induces sFcɛRI release. We demonstrated that generation of sFcɛRI requires Src phosphorylation and endo/lysosomal acidification. In experimental mouse models, sFcɛRI diminishes the severity of IgE-mediated anaphylaxis. BATs confirmed that, comparable to the anti-IgE monoclonal antibody omalizumab, sFcɛRI is an inhibitor of the human innate IgE effector axis, implying that sFcɛRI and omalizumab potentially inhibit each other in vivo. CONCLUSION sFcɛRI is produced after antigen-specific IgE/FcɛRI-mediated activation signals and functions as an endogenous inhibitor of IgE loading to FcɛRI and IgE-mediated activation. Our results imply, therefore, that sFcɛRI contributes to a negative regulatory feedback loop that aims at preventing overshooting responses after IgE-mediated immune activation.
Collapse
Affiliation(s)
- S. Moñino-Romero
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - L. Erkert
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - K. Schmidthaler
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| | - S. C. Diesner
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| | - B. F. Sallis
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| | - L. Pennington
- Department of Structural Biology; School of Medicine; Stanford University; Stanford California
| | - T. Jardetzky
- Department of Structural Biology; School of Medicine; Stanford University; Stanford California
| | - H. C. Oettgen
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Division of Immunology; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - B. Bohle
- Department of Pathophysiology and Allergy Research; Medical University of Vienna; Vienna Austria
| | - E. Fiebiger
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| | - Z. Szépfalusi
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| |
Collapse
|
48
|
Jiménez‐Saiz R, Ellenbogen Y, Koenig JFE, Gordon ME, Walker TD, Rosace D, Spill P, Bruton K, Kong J, Monteiro K, Wen J, Tuomanen EI, Kolbeck R, Chu DK, Waserman S, Jordana M. IgG1 + B-cell immunity predates IgE responses in epicutaneous sensitization to foods. Allergy 2019; 74:165-175. [PMID: 29790165 DOI: 10.1111/all.13481] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND The generation of IgE-mediated food allergy in humans is silent and only diagnosed upon manifestation of clinical symptoms. While experimental models have been used to investigate some mechanisms of allergic sensitization, the generation of humoral immunity and memory remains to be elucidated. Here, we defined the evolution of allergen-specific B-cell responses during epicutaneous sensitization to foods. METHODS Wild-type and genetic knockout animals, and drug or antibody strategies for cell depletion and immunoglobulin signaling blockade were used to investigate epicutaneous sensitization and disease progression; we analyzed allergen-specific germinal centers and IgG1+ memory B cells by flow cytometry, evaluated humoral responses, and determined clinical reactivity (anaphylaxis). RESULTS Epicutaneous sensitization caused microscopic skin damage, inflammation, and recruitment of activated dendritic cells to the draining lymph nodes. This process generated allergen-specific IgG1+ germinal center B cells, serum IgG1, and anaphylaxis that was mediated by the alternative pathway. Whether we used peanut and/or ovalbumin from the egg white for sensitization, the allergen-specific IgG1+ memory compartment predominantly exhibited an immature, pro-germinal center phenotype (PDL-2- CD80- CD35+ CD73+ ). Subsequent subclinical exposures to the allergen induced IgE+ germinal center B cells, serum IgE, and likely activated the classical pathway of anaphylaxis. CONCLUSIONS Our data demonstrate that IgG1+ B-cell immunity against food allergens in epicutaneous sensitization precedes the generation of IgE responses. Therefore, the assessment of allergen-specific cellular and humoral IgG1+ immunity may help to identify individuals at risk of developing IgE-mediated food allergy and hence provide a window for therapeutic interventions.
Collapse
Affiliation(s)
- R. Jiménez‐Saiz
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - Y. Ellenbogen
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - J. F. E. Koenig
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - M. E. Gordon
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - T. D. Walker
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - D. Rosace
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - P. Spill
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - K. Bruton
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - J. Kong
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - K. Monteiro
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - J. Wen
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| | - E. I. Tuomanen
- Department of Infectious Diseases St. Jude Children's Research Hospital Memphis TN USA
| | - R. Kolbeck
- Department of Respiratory, Inflammation & Autoimmunity MedImmune LLC Gaithersburg MA USA
| | - D. K. Chu
- Department of Medicine McMaster University Hamilton ON Canada
| | - S. Waserman
- Department of Medicine McMaster University Hamilton ON Canada
| | - M. Jordana
- Department of Pathology & Molecular Medicine McMaster Immunology Research Centre (MIRC) McMaster University Hamilton ON Canada
| |
Collapse
|
49
|
Mompó SM, González-Fernández Á. Antigen-Specific Human Monoclonal Antibodies from Transgenic Mice. Methods Mol Biol 2018; 1904:253-291. [PMID: 30539474 DOI: 10.1007/978-1-4939-8958-4_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Due to the difficulties found when generating fully human monoclonal antibodies (mAbs) by the traditional method, several efforts have attempted to overcome these problems, with varying levels of success. One approach has been the development of transgenic mice carrying immunoglobulin (Ig) genes in germline configuration. The engineered mouse genome can undergo productive rearrangement in the B-cell population, with the generation of mouse B lymphocytes expressing human Ig (hIg) chains. To avoid the expression of mouse heavy or light chains, the endogenous mouse Ig (mIg) loci must be silenced by gene-targeting techniques. Subsequently, to obtain antigen-specific mAbs, conventional immunization protocols can be followed and the mAb technique used (fusion of activated B cells with mouse myeloma cells, screening, cloning, freezing, and testing) with these animThis chapter summarizes the most common chromatographic mAb andals expressing human Ig genes. This chapter describes the type of transgenic-knockout mice generated for various research groups, provides examples of human mAbs developed by research groups and companies, and includes protocols of immunization, generation, production, and purification of human mAbs from such mice. In addition, it also addresses the problems detected, and includes some of the methods that can be used to analyze functional activities with human mAbs.
Collapse
Affiliation(s)
- Susana Magadán Mompó
- Immunology, Centro de Investigaciones Biomédicas (CINBIO), Centro de Investigación Singular de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Vigo, Spain
| | - África González-Fernández
- Immunology, Centro de Investigaciones Biomédicas (CINBIO), Centro de Investigación Singular de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Vigo, Spain.
| |
Collapse
|
50
|
Khosravi M, Boroun F, Nemati M. The anti-penicillin antibodies levels in sensitive and insensitive people to intradermal skin test. Hum Antibodies 2018; 27:63-68. [PMID: 30223394 DOI: 10.3233/hab-180349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The hypersensitivity reaction to penicillin is a public health problem. Immunological responses to penicillin and other beta-lactam antibiotics can be classified into immediate and non-immediate responses. The immediate hypersensitivity is mediated by IgE; however, the non-immediate sensitivity is facilitated by other isotypes of antibody or T lymphocytes. OBJECTIVE This research detected the non-IgE antibody value against penicillin in allergic and normal people. METHODS Thirty-eight samples from patients with positive or negative intradermal skin testing results of penicillin allergy were included in this study. The total antibody and IgM levels against penicillin G were defined by in-house ELISA test. RESULTS The results showed a significant (P< 0.05) elevation in total immunoglobulin and non-IgM anti-penicillin antibody of sensitive groups; however, the anti-penicillin IgM was significantly greater in non-sensitive peoples. CONCLUSIONS Although the sensitized people to penicillin cannot be certainly detected with the total antibody, specific IgG and IgM value against penicillin, these values are good indicators for prediction of immediate and late response of the immune system to penicillin.
Collapse
Affiliation(s)
- Mohammad Khosravi
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Fatemeh Boroun
- Graduated Student of Microbiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Nemati
- Department of Venomous Animals and Toxins, Razi Vaccine and Serum Research Institute, Ahvaz Branch, Agriculture Research Education and Extension Organization (AREEO), Iran
| |
Collapse
|