1
|
Dejoux A, Zhu Q, Ganneau C, Goff ORL, Godon O, Lemaitre J, Relouzat F, Huetz F, Sokal A, Vandenberghe A, Pecalvel C, Hunault L, Derenne T, Gillis CM, Iannascoli B, Wang Y, Rose T, Mertens C, Nicaise-Roland P, England P, Mahévas M, de Chaisemartin L, Le Grand R, Letscher H, Saul F, Pissis C, Haouz A, Reber LL, Chappert P, Jönsson F, Ebo DG, Millot GA, Bay S, Chollet-Martin S, Gouel-Chéron A, Bruhns P. Rocuronium-specific antibodies drive perioperative anaphylaxis but can also function as reversal agents in preclinical models. Sci Transl Med 2024; 16:eado4463. [PMID: 39259810 DOI: 10.1126/scitranslmed.ado4463] [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: 02/02/2024] [Revised: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
Neuromuscular blocking agents (NMBAs) relax skeletal muscles to facilitate surgeries and ease intubation but can lead to adverse reactions, including complications because of postoperative residual neuromuscular blockade (rNMB) and, in rare cases, anaphylaxis. Both adverse reactions vary between types of NMBAs, with rocuronium, a widely used nondepolarizing NMBA, inducing one of the longest rNMB durations and highest anaphylaxis incidences. rNMB induced by rocuronium can be reversed by the synthetic γ-cyclodextrin sugammadex. However, in rare cases, sugammadex can provoke anaphylaxis. Thus, additional therapeutic options are needed. Rocuronium-induced anaphylaxis is proposed to rely on preexisting rocuronium-binding antibodies. To understand the pathogenesis of rocuronium-induced anaphylaxis and to identify potential therapeutics, we investigated the memory B cell antibody repertoire of patients with suspected hypersensitivity to rocuronium. We identified polyclonal antibody repertoires with a high diversity among V(D)J genes without evidence of clonal groups. When recombinantly expressed, these antibodies demonstrated specificity and low affinity for rocuronium without cross-reactivity for other NMBAs. Moreover, when these antibodies were expressed as human immunoglobulin E (IgE), they triggered human mast cell activation and passive systemic anaphylaxis in transgenic mice, although their affinities were insufficient to serve as reversal agents. Rocuronium-specific, high-affinity antibodies were thus isolated from rocuronium-immunized mice. The highest-affinity antibody was able to reverse rocuronium-induced neuromuscular blockade in nonhuman primates with kinetics comparable to that of sugammadex. Together, these data support the hypothesis that antibodies cause anaphylactic reactions to rocuronium and pave the way for improved diagnostics and neuromuscular blockade reversal agents.
Collapse
Affiliation(s)
- Alice Dejoux
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Qianqian Zhu
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
| | - Christelle Ganneau
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Chimie des Biomolécules, 75015 Paris, France
| | - Odile Richard-Le Goff
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Ophélie Godon
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Julien Lemaitre
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - François Huetz
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Aurélien Sokal
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
- Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris Cité, 92110 Clichy, France
| | - Alexis Vandenberghe
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Cyprien Pecalvel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31000 Toulouse, France
| | - Lise Hunault
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Thomas Derenne
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Caitlin M Gillis
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Bruno Iannascoli
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Yidan Wang
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Thierry Rose
- Institut Pasteur, Université Paris Cité, INSERM UMR1224, Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, 75015 Paris, France
| | - Christel Mertens
- Faculty of Medicine and Health Science, Department of Immunology-Allergology-Rheumatology, Antwerp University Hospital and the Infla-Med Center of Excellence, University of Antwerp, Antwerp, Belgium; Department of Immunology and Allergology, AZ Jan Palfijn Ghent, 9000 Ghent, Belgium
| | - Pascale Nicaise-Roland
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Patrick England
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Molecular Biophysics Core Facility, 75015 Paris, France
| | - Matthieu Mahévas
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Luc de Chaisemartin
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Hélène Letscher
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Frederick Saul
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Cédric Pissis
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31000 Toulouse, France
| | - Pascal Chappert
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Friederike Jönsson
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- CNRS, F-75015 Paris, France
| | - Didier G Ebo
- Faculty of Medicine and Health Science, Department of Immunology-Allergology-Rheumatology, Antwerp University Hospital and the Infla-Med Center of Excellence, University of Antwerp, Antwerp, Belgium; Department of Immunology and Allergology, AZ Jan Palfijn Ghent, 9000 Ghent, Belgium
| | - Gaël A Millot
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015 Paris, France
| | - Sylvie Bay
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Chimie des Biomolécules, 75015 Paris, France
| | - Sylvie Chollet-Martin
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Aurélie Gouel-Chéron
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Anaesthesiology and Critical Care Medicine Department, DMU Parabol, Bichat-Claude Bernard Hospital, AP-HP, 75018 Paris, France
- Université Paris Cité, 75010 Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- INSERM 1152, DHU FIRE, Labex Inflamex, Université Paris Diderot Paris 7, 75018 Paris, France
| |
Collapse
|
2
|
Mucha J, Cho A, Weijler AM, Muckenhuber M, Hofmann AG, Wahrmann M, Heinzel A, Linhart B, Gattinger P, Valenta R, Berlakovich G, Zuckermann A, Jaksch P, Oberbauer R, Wekerle T. Prospective assessment of pre-existing and de novo anti-HLA IgE in kidney, liver, lung and heart transplantation. Front Immunol 2023; 14:1179036. [PMID: 37731514 PMCID: PMC10507692 DOI: 10.3389/fimmu.2023.1179036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction Antibody mediated rejection (ABMR) is a major factor limiting outcome after organ transplantation. Anti-HLA donor-specific antibodies (DSA) of the IgG isotype are mainly responsible for ABMR. Recently DSA of the IgE isotype were demonstrated in murine models as well as in a small cohort of sensitized transplant recipients. In the present study, we aimed to determine the frequency of pre-existing and de novo anti-HLA IgE antibodies in a cohort of 105 solid organ transplant recipients. Methods We prospectively measured anti-HLA IgE antibodies in a cohort of kidney (n=60), liver, heart and lung (n=15 each) transplant recipients before and within one-year after transplantation, employing a single-antigen bead assay for HLA class I and class II antigens. Functional activity of anti-HLA IgE antibodies was assessed by an in vitro mediator release assay. Antibodies of the IgG1-4 subclasses and Th1 and Th2 cytokines were measured in anti-HLA IgE positive patients. Results Pre-existing anti-HLA IgE antibodies were detected in 10% of renal recipients (including 3.3% IgE-DSA) and in 4.4% of non-renal solid organ transplant recipients (heart, liver and lung cohort). Anti-HLA IgE occurred only in patients that were positive for anti-HLA IgG, and most IgE positive patients had had a previous transplant. Only a small fraction of patients developed de novo anti-HLA IgE antibodies (1.7% of kidney recipients and 4.4% of non-renal recipients), whereas no de novo IgE-DSA was detected. IgG subclass antibodies showed a distinct pattern in patients who were positive for anti-HLA IgE. Moreover, patients with anti-HLA IgE showed elevated Th2 and also Th1 cytokine levels. Serum from IgE positive recipients led to degranulation of basophils in vitro, demonstrating functionality of anti-HLA IgE. Discussion These data demonstrate that anti-HLA IgE antibodies occur at low frequency in kidney, liver, heart and lung transplant recipients. Anti-HLA IgE development is associated with sensitization at the IgG level, in particular through previous transplants and distinct IgG subclasses. Taken together, HLA specific IgE sensitization is a new phenomenon in solid organ transplant recipients whose potential relevance for allograft injury requires further investigation.
Collapse
Affiliation(s)
- Jasmin Mucha
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Ara Cho
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Anna Marianne Weijler
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Moritz Muckenhuber
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Amun Georg Hofmann
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Markus Wahrmann
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Birgit Linhart
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Pia Gattinger
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Gabriela Berlakovich
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Olivera-Ardid S, Bello-Gil D, Tuzikov A, Araujo RN, Ferrero-Alves Y, García Figueroa BE, Labrador-Horrillo M, García-Pérez AL, Bovin N, Mañez R. Poly-L-Lysine-Based αGal-Glycoconjugates for Treating Anti-αGal IgE-Mediated Diseases. Front Immunol 2022; 13:873019. [PMID: 35432370 PMCID: PMC9009260 DOI: 10.3389/fimmu.2022.873019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/02/2022] [Indexed: 11/25/2022] Open
Abstract
Anti-αGal IgE antibodies mediate a spreading allergic condition known as αGal-syndrome (AGS). People exposed to hard tick bites are sensitized to αGal, producing elevated levels of anti-αGal IgE, which are responsible for AGS. This work presents an immunotherapy based on polymeric αGal-glycoconjugates for potentially treating allergic disorders by selectively inhibiting anti-αGal IgE antibodies. We synthesized a set of αGal-glycoconjugates, based on poly-L-lysine of different degrees of polymerization (DP1000, DP600, and DP100), to specifically inhibit in vitro the anti-αGal IgE antibodies in the serum of αGal-sensitized patients (n=13). Moreover, an animal model for αGal sensitization in GalT-KO mice was developed by intradermal administration of hard tick' salivary gland extract, mimicking the sensitization mechanism postulated in humans. The in vitro exposure to all polymeric glycoconjugates (5-10-20-50-100 µg/mL) mainly inhibited anti-αGal IgE and IgM isotypes, with a lower inhibition effect on the IgA and IgG, respectively. We demonstrated a differential anti-αGal isotype inhibition as a function of the length of the poly-L-lysine and the number of αGal residues exposed in the glycoconjugates. These results defined a minimum of 27 αGal residues to inhibit most of the induced anti-αGal IgE in vitro. Furthermore, the αGal-glycoconjugate DP1000-RA0118 (10 mg/kg sc.) showed a high capacity to remove the anti-αGal IgE antibodies (≥75% on average) induced in GalT-KO mice, together with similar inhibition for circulating anti-αGal IgG and IgM. Our study suggests the potential clinical use of poly-L-lysine-based αGal-glycoconjugates for treating allergic disorders mediated by anti-αGal IgE antibodies.
Collapse
Affiliation(s)
- Sara Olivera-Ardid
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Daniel Bello-Gil
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Alexander Tuzikov
- Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Ricardo N. Araujo
- Laboratório de Artrópodes Hematófagos, Departamento de Parasitologia, ICB/UFMG, Belo Horizonte, Brazil
| | - Yara Ferrero-Alves
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Blanca Esther García Figueroa
- MEGA: Asthma Inception and Progression Mechanisms, Complejo Hospitalario de Navarra (CHN), Pamplona, Spain
- Instituto de investigación sanitaria de Navarra (IdiSNA), Pamplona, Spain
- ARADyAL Research Network, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Moisés Labrador-Horrillo
- ARADyAL Research Network, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Allergy Section, Internal Medicine Department, Hospital Universitari Vall d’Hebron (HUVH), Barcelona, Spain
- Immunomediated Diseases and Innovative Therapies, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Ana L. García-Pérez
- Departamento de Sanidad Animal, Instituto Vasco de Investigación de Desarrollo Agrario (NEIKER), Derio, Spain
| | - Nicolai Bovin
- Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Rafael Mañez
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
- Hospital Universitari de Bellvitge, Servicio de Medicina Intensiva, Hospitalet de Llobregat, Barcelona, Spain
- Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Grupo Inmunidad Innata y Patología del Paciente Crítico, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
4
|
Zografou C, Vakrakou AG, Stathopoulos P. Short- and Long-Lived Autoantibody-Secreting Cells in Autoimmune Neurological Disorders. Front Immunol 2021; 12:686466. [PMID: 34220839 PMCID: PMC8248361 DOI: 10.3389/fimmu.2021.686466] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022] Open
Abstract
As B cells differentiate into antibody-secreting cells (ASCs), short-lived plasmablasts (SLPBs) are produced by a primary extrafollicular response, followed by the generation of memory B cells and long-lived plasma cells (LLPCs) in germinal centers (GCs). Generation of IgG4 antibodies is T helper type 2 (Th2) and IL-4, -13, and -10-driven and can occur parallel to IgE, in response to chronic stimulation by allergens and helminths. Although IgG4 antibodies are non-crosslinking and have limited ability to mobilize complement and cellular cytotoxicity, when self-tolerance is lost, they can disrupt ligand-receptor binding and cause a wide range of autoimmune disorders including neurological autoimmunity. In myasthenia gravis with predominantly IgG4 autoantibodies against muscle-specific kinase (MuSK), it has been observed that one-time CD20+ B cell depletion with rituximab commonly leads to long-term remission and a marked reduction in autoantibody titer, pointing to a short-lived nature of autoantibody-secreting cells. This is also observed in other predominantly IgG4 autoantibody-mediated neurological disorders, such as chronic inflammatory demyelinating polyneuropathy and autoimmune encephalitis with autoantibodies against the Ranvier paranode and juxtaparanode, respectively, and extends beyond neurological autoimmunity as well. Although IgG1 autoantibody-mediated neurological disorders can also respond well to rituximab induction therapy in combination with an autoantibody titer drop, remission tends to be less long-lasting and cases where titers are refractory tend to occur more often than in IgG4 autoimmunity. Moreover, presence of GC-like structures in the thymus of myasthenic patients with predominantly IgG1 autoantibodies against the acetylcholine receptor and in ovarian teratomas of autoimmune encephalitis patients with predominantly IgG1 autoantibodies against the N‐methyl‐d‐aspartate receptor (NMDAR) confers increased the ability to generate LLPCs. Here, we review available information on the short-and long-lived nature of ASCs in IgG1 and IgG4 autoantibody-mediated neurological disorders and highlight common mechanisms as well as differences, all of which can inform therapeutic strategies and personalized medical approaches.
Collapse
Affiliation(s)
- C Zografou
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - A G Vakrakou
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - P Stathopoulos
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
5
|
Aalberse RC, Hoekzema R, Grayson MH. Do germinal centers protect most of us from becoming allergic? Ann Allergy Asthma Immunol 2021; 127:301-305. [PMID: 34102303 DOI: 10.1016/j.anai.2021.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To review the literature and discuss a hypothesis as to why most people do not have allergy. This hypothesis is dependent on the following 3 main components: (1) airborne allergens (eg, from pollen or mites) are weak antigens that induce a B-cell response only in immunologically most reactive subjects (ie, with atopy); (2) a roadblock to production of immunoglobulin E (IgE) is the T helper 2/interleukin 4 requirement for class switch to IgE; (3) activated germinal centers prevent the formation of mature IgE-switched B-cells, creating a second roadblock to IgE production. DATA SOURCES Transgenic reporter mice and a cross-sectional human cohort. STUDY SELECTIONS From the mouse studies, we selected the data on histology and tissue-derived cell suspensions published by several groups in 2011 to 2014. From the human cohort, we selected our published microarray data on the levels of allergen-specific IgE and IgG in serum. RESULTS The immune response to airborne atopic allergens entails both IgE and IgG antibodies rather than just an IgG or IgE response. However, as expected for an immune response without mature germinal centers, the specific IgG levels will be very low, typically in the ng/ml range. CONCLUSION Control of IgE production is not just through the T helper 2/interleukin 4-mediated class switch. Recent studies suggest that mature germinal centers are likely to provide protection against the development of allergy to airborne allergens, as well. This may explain why allergen exposure does not induce allergen-specific IgE in everyone.
Collapse
Affiliation(s)
- Rob C Aalberse
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands.
| | - Rick Hoekzema
- Department of Dermatology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Mitchell H Grayson
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio; Center for Clinical and Translational Research, the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
6
|
Bruhns P, Chollet-Martin S. Mechanisms of human drug-induced anaphylaxis. J Allergy Clin Immunol 2021; 147:1133-1142. [PMID: 33832695 DOI: 10.1016/j.jaci.2021.02.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/11/2021] [Indexed: 02/08/2023]
Abstract
Drug-induced anaphylaxis is a hyperacute reaction affecting multiple organs that can be of fatal consequence. Its incidence is increasing, consistent with a global increased sensitization to various allergens and drugs in the population. Few risk factors and mechanisms have been identified from human studies due to the rarity of anaphylactic events and their unpredictability. This systemic reaction is caused by the rapid release of a large range of functionally diverse mediators, including histamine and platelet-activating factor as the main drivers identified. Mechanisms defined from models of experimental anaphylaxis identify drug-specific antibodies of the IgE and IgG class that link the drug to antibody receptors on multiple cell types, causing their activation and mediator release. In the case of drugs with peculiar chemical structures, antibodies may not be necessary because drug-binding receptors, such as Mas-related G protein-coupled receptor member X2, have been identified. This review describes the complex reaction leading to drug-induced anaphylaxis that can involve various antibody classes, various cell types-including mast cells, neutrophils, platelets, basophils, macrophages, and monocytes-and their mediators and receptors that, importantly, can be activated alone or in association to participate in the severity of the reaction.
Collapse
Affiliation(s)
- Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, UMR 1222 INSERM, Institut Pasteur, Paris, France; DHU FIRE, Labex Inflamex, Université Paris Diderot Paris 7, Paris, France.
| | - Sylvie Chollet-Martin
- Department "Auto-immunité et Hypersensibilités," DMU BioGeM, APHP, Hôpital Bichat, Paris, France; "Inflammation, Microbiome and Immunosurveillance" INSERM UMR 996, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France.
| |
Collapse
|
7
|
Kariyawasam HH, James LK. Chronic Rhinosinusitis with Nasal Polyps: Targeting IgE with Anti-IgE Omalizumab Therapy. Drug Des Devel Ther 2020; 14:5483-5494. [PMID: 33328726 PMCID: PMC7735718 DOI: 10.2147/dddt.s226575] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a complex, clinically heterogeneous and persistent inflammatory disorder of the upper airway. Detailed mechanistic insights into disease pathogenesis are lacking, but it is now accepted that local tissue IgE driven T2-high inflammatory pathways are critical to disease. The recent CRSwNP Phase 3 POLYP1 and POLYP2 replicate studies of blocking IgE with omalizumab confirmed rapid improvements in all clinical parameters of sinonasal disease, confirming a pivotal role for IgE driven inflammatory pathways in CRSwNP. This review summarises the biology of IgE in relation to CRSwNP. Insight into how IgE may drive CRSwNP is evaluated in the context of clinical improvements seen with omalizumab. The need for further studies using a broader patient and biomarker specific groups to aid more precise drug-patient selection alongside more detailed mechanistic studies of omalizumab in CRSwNP is highlighted.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| | - Louisa K James
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Chandrasekhar JL, Cox KM, Erickson LD. B Cell Responses in the Development of Mammalian Meat Allergy. Front Immunol 2020; 11:1532. [PMID: 32765532 PMCID: PMC7379154 DOI: 10.3389/fimmu.2020.01532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Studies of meat allergic patients have shown that eating meat poses a serious acute health risk that can induce severe cutaneous, gastrointestinal, and respiratory reactions. Allergic reactions in affected individuals following meat consumption are mediated predominantly by IgE antibodies specific for galactose-α-1,3-galactose (α-gal), a blood group antigen of non-primate mammals and therefore present in dietary meat. α-gal is also found within certain tick species and tick bites are strongly linked to meat allergy. Thus, it is thought that exposure to tick bites promotes cutaneous sensitization to tick antigens such as α-gal, leading to the development of IgE-mediated meat allergy. The underlying immune mechanisms by which skin exposure to ticks leads to the production of α-gal-specific IgE are poorly understood and are key to identifying novel treatments for this disease. In this review, we summarize the evidence of cutaneous exposure to tick bites and the development of mammalian meat allergy. We then provide recent insights into the role of B cells in IgE production in human patients with mammalian meat allergy and in a novel mouse model of meat allergy. Finally, we discuss existing data more generally focused on tick-mediated immunomodulation, and highlight possible mechanisms for how cutaneous exposure to tick bites might affect B cell responses in the skin and gut that contribute to loss of oral tolerance.
Collapse
Affiliation(s)
- Jessica L Chandrasekhar
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kelly M Cox
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Loren D Erickson
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
9
|
Robinson MJ, Webster RH, Tarlinton DM. How intrinsic and extrinsic regulators of plasma cell survival might intersect for durable humoral immunity. Immunol Rev 2020; 296:87-103. [DOI: 10.1111/imr.12895] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Marcus J. Robinson
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - Rosela H. Webster
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - David M. Tarlinton
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| |
Collapse
|
10
|
Dhondalay GK, Rael E, Acharya S, Zhang W, Sampath V, Galli SJ, Tibshirani R, Boyd SD, Maecker H, Nadeau KC, Andorf S. Food allergy and omics. J Allergy Clin Immunol 2019; 141:20-29. [PMID: 29307411 DOI: 10.1016/j.jaci.2017.11.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 01/06/2023]
Abstract
Food allergy (FA) prevalence has been increasing over the last few decades and is now a global health concern. Current diagnostic methods for FA result in a high number of false-positive results, and the standard of care is either allergen avoidance or use of epinephrine on accidental exposure, although currently with no other approved treatments. The increasing prevalence of FA, lack of robust biomarkers, and inadequate treatments warrants further research into the mechanism underlying food allergies. Recent technological advances have made it possible to move beyond traditional biological techniques to more sophisticated high-throughput approaches. These technologies have created the burgeoning field of omics sciences, which permit a more systematic investigation of biological problems. Omics sciences, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, microbiomics, and exposomics, have enabled the construction of regulatory networks and biological pathway models. Parallel advances in bioinformatics and computational techniques have enabled the integration, analysis, and interpretation of these exponentially growing data sets and opens the possibility of personalized or precision medicine for FA.
Collapse
Affiliation(s)
- Gopal Krishna Dhondalay
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Efren Rael
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Swati Acharya
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Wenming Zhang
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, Calif
| | - Robert Tibshirani
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, Calif
| | - Scott D Boyd
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Holden Maecker
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Kari Christine Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif.
| | - Sandra Andorf
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, Calif
| |
Collapse
|
11
|
Shah HB, Smith K, Wren JD, Webb CF, Ballard JD, Bourn RL, James JA, Lang ML. Insights From Analysis of Human Antigen-Specific Memory B Cell Repertoires. Front Immunol 2019; 9:3064. [PMID: 30697210 PMCID: PMC6340933 DOI: 10.3389/fimmu.2018.03064] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022] Open
Abstract
Memory B cells that are generated during an infection or following vaccination act as sentinels to guard against future infections. Upon repeat antigen exposure memory B cells differentiate into new antibody-secreting plasma cells to provide rapid and sustained protection. Some pathogens evade or suppress the humoral immune system, or induce memory B cells with a diminished ability to differentiate into new plasma cells. This leaves the host vulnerable to chronic or recurrent infections. Single cell approaches coupled with next generation antibody gene sequencing facilitate a detailed analysis of the pathogen-specific memory B cell repertoire. Monoclonal antibodies that are generated from antibody gene sequences allow a functional analysis of the repertoire. This review discusses what has been learned thus far from analysis of diverse pathogen-specific memory B cell compartments and describes major differences in their repertoires. Such information may illuminate ways to advance the goal of improving vaccine and therapeutic antibody design.
Collapse
Affiliation(s)
- Hemangi B Shah
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kenneth Smith
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jonathan D Wren
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States.,Department of Biochemistry and Molecular Biology and Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Carol F Webb
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Division of Rheumatology, Immunology and Allergy, Department of Cell Biology and Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jimmy D Ballard
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rebecka L Bourn
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Judith A James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States.,Department of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Mark L Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
12
|
Aalberse RC, Lupinek C, Siroux V, Nadif R, Just J, Bousquet J, Valenta R, Platts-Mills TA. sIgE and sIgG to airborne atopic allergens: Coupled rather than inversely related responses. Allergy 2018; 73:2239-2242. [PMID: 29987842 DOI: 10.1111/all.13548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Rob C. Aalberse
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Christian Lupinek
- The Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Valérie Siroux
- Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health; Inserm; CNRS; Institute for Advanced Biosciences (IAB); U1209 Joint Research Center; University Grenoble Alpes; Grenoble France
| | - Rachel Nadif
- INSERM U1168; VIMA: Aging and Chronic Diseases. Epidemiological and Public Health Approaches; Villejuif France
- UMR-S 1168; Univ Versailles St-Quentin-en-Yvelines; Montigny le Bretonneux France
| | - Jocelyne Just
- Assistance Publique-Hôpitaux de Paris; Allergology Department; Hôpital Armand-Trousseau; Paris France
- Université Paris; Paris France
| | - Jean Bousquet
- INSERM U1168; VIMA: Aging and Chronic Diseases. Epidemiological and Public Health Approaches; Villejuif France
- UMR-S 1168; Univ Versailles St-Quentin-en-Yvelines; Montigny le Bretonneux France
- University Hospital; Montpellier France
| | - Rudolf Valenta
- The Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Thomas A. Platts-Mills
- Asthma and Allergic Diseases Center; University of Virginia Health System; Charlottesville Virginia
| |
Collapse
|
13
|
Ehlers AM, Blankestijn MA, Knulst AC, Klinge M, Otten HG. Can alternative epitope mapping approaches increase the impact of B-cell epitopes in food allergy diagnostics? Clin Exp Allergy 2018; 49:17-26. [PMID: 30294841 PMCID: PMC7380004 DOI: 10.1111/cea.13291] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 01/08/2023]
Abstract
In vitro allergy diagnostics are currently based on the detection of specific IgE binding on intact allergens or a mixture thereof. This approach has drawbacks as it may yield false‐negative and/or false‐positive results. Thus, we reviewed the impact of known B‐cell epitopes of food allergens to predict transience or persistence, tolerance or allergy and the severity of an allergic reaction and to examine new epitope mapping strategies meant to improve serum‐based allergy diagnostics. Recent epitope mapping approaches have been worthwhile in epitope identification and may increase the specificity of allergy diagnostics by using epitopes predominately recognized by allergic patients in some cases. However, these approaches did not lead to discrimination between clinically relevant and irrelevant epitopes so far, since the polyclonal serum IgE‐binding epitope spectrum seems to be too individual, independent of the disease status of the patients. New epitope mapping strategies are necessary to overcome these obstacles. The use of patient‐derived monoclonal antibodies instead of patient sera for functional characterization of clinically relevant and irrelevant epitope combinations, distinguished by their ability to induce degranulation, might be a promising approach to gain more insight into the allergic reaction and to improve serum‐based allergy diagnostics.
Collapse
Affiliation(s)
- Anna M Ehlers
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mark A Blankestijn
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Andre C Knulst
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Henny G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Matricardi PM, Hofmaier S, Perna S, Huang X, Keil T, Lau S. Reply to: "Allergen-specific IgG responses preceding allergic sensitization". Allergy 2018; 73:1926-1928. [PMID: 29897104 DOI: 10.1111/all.13503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- P M Matricardi
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S Hofmaier
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S Perna
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - X Huang
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Pediatrics, Shengzhou People's Hospital, Shengzhou, China
| | - T Keil
- Institute for Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S Lau
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15
|
Heeringa JJ, Rijvers L, Arends NJ, Driessen GJ, Pasmans SG, Dongen JJM, Jongste JC, Zelm MC. IgE-expressing memory B cells and plasmablasts are increased in blood of children with asthma, food allergy, and atopic dermatitis. Allergy 2018; 73:1331-1336. [PMID: 29380876 DOI: 10.1111/all.13421] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Despite the critical role of soluble IgE in the pathology of IgE-mediated allergic disease, little is known about abnormalities in the memory B cells and plasma cells that produce IgE in allergic patients. We here applied a flow cytometric approach to cross-sectionally study blood IgE+ memory B cells and plasmablasts in 149 children with atopic dermatitis, food allergy, and/or asthma and correlated these to helper T(h)2 cells and eosinophils. Children with allergic disease had increased numbers of IgE+CD27- and IgE+CD27+ memory B cells and IgE+ plasmablasts, as well as increased numbers of eosinophils and Th2 cells. IgE+ plasmablast numbers correlated positively with Th2 cell numbers. These findings open new possibilities for diagnosis and monitoring of treatment in patients with allergic diseases.
Collapse
Affiliation(s)
- J. J. Heeringa
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
- Department of Pediatrics Erasmus MC University Medical Center Rotterdam The Netherlands
| | - L. Rijvers
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
| | - N. J. Arends
- Department of Allergy Erasmus MC University Medical Center Rotterdam The Netherlands
| | - G. J. Driessen
- Department of Pediatrics Erasmus MC University Medical Center Rotterdam The Netherlands
- Department of Pediatrics Haga Teaching Hospital Juliana Children's Hospital The Hague The Netherlands
| | - S. G. Pasmans
- Department of Pediatric Dermatology Erasmus MC University Medical Center Rotterdam The Netherlands
| | - J. J. M. Dongen
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
- Department of Immunology Leiden University Medical Center Leiden The Netherlands
| | - J. C. Jongste
- Department of Pediatrics Division of Respiratory Medicine Erasmus MC University Medical Center Rotterdam The Netherlands
| | - M. C. Zelm
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
- Department of Immunology and Pathology Central Clinical School Monash University and Alfred Hospital Melbourne VIC Australia
| |
Collapse
|
16
|
A murine model of atopic dermatitis can be generated by painting the dorsal skin with hapten twice 14 days apart. Sci Rep 2018; 8:5988. [PMID: 29662233 PMCID: PMC5902631 DOI: 10.1038/s41598-018-24363-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 03/20/2018] [Indexed: 01/28/2023] Open
Abstract
Drug development involves pharmacometric experiments in animals. Such experiments should limit animal pain and stress. Conventional murine models of atopic dermatitis (AD) used in drug development are generated by weekly painting of hapten on dorsal skin for 5 weeks. The present study aimed to develop a protocol that involves less animal distress. The experiments focused on serum total IgE levels, which are a marker of AD. The conventional protocol induced ever rising IgE levels. Experiments with extended intervals between sensitizations showed that IgE peaked ~5 days after the second sensitization, after which it returned to the control level within 12-19 days. An additional third sensitization on day 28 further increased the serum IgE level. In the 4-5 days after the second sensitization, the dorsal skin exhibited typical AD-like lesions with edema, scabs, epithelial-cell hypertrophy, marked mast-cell and lymphocyte infiltration of dermis, and increased IL-4, IL-6, IL-10, IL-1β, IL-17A, IFN-γ and TNF-α expression. Thus, two 2,4-dinitrofluorobenzene sensitizations yield a murine AD model in less than 20 days. This study shows that animal model protocols used in drug development can be fine-tuned so that they remain effective yet cause animals less stress and pain.
Collapse
|
17
|
Kirik U, Persson H, Levander F, Greiff L, Ohlin M. Antibody Heavy Chain Variable Domains of Different Germline Gene Origins Diversify through Different Paths. Front Immunol 2017; 8:1433. [PMID: 29180996 PMCID: PMC5694033 DOI: 10.3389/fimmu.2017.01433] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/16/2017] [Indexed: 02/04/2023] Open
Abstract
B cells produce antibodies, key effector molecules in health and disease. They mature their properties, including their affinity for antigen, through hypermutation events; processes that involve, e.g., base substitution, codon insertion and deletion, often in association with an isotype switch. Investigations of antibody evolution define modes whereby particular antibody responses are able to form, and such studies provide insight important for instance for development of efficient vaccines. Antibody evolution is also used in vitro for the design of antibodies with improved properties. To better understand the basic concepts of antibody evolution, we analyzed the mutational paths, both in terms of amino acid substitution and insertions and deletions, taken by antibodies of the IgG isotype. The analysis focused on the evolution of the heavy chain variable domain of sets of antibodies, each with an origin in 1 of 11 different germline genes representing six human heavy chain germline gene subgroups. Investigated genes were isolated from cells of human bone marrow, a major site of antibody production, and characterized by next-generation sequencing and an in-house bioinformatics pipeline. Apart from substitutions within the complementarity determining regions, multiple framework residues including those in protein cores were targets of extensive diversification. Diversity, both in terms of substitutions, and insertions and deletions, in antibodies is focused to different positions in the sequence in a germline gene-unique manner. Altogether, our findings create a framework for understanding patterns of evolution of antibodies from defined germline genes.
Collapse
Affiliation(s)
- Ufuk Kirik
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Helena Persson
- Science for Life Laboratory, Drug Discovery and Development Platform, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden.,National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Immunotechnology, Lund University, Lund, Sweden
| | - Lennart Greiff
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Otorhinolaryngology, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden.,Science for Life Laboratory, Drug Discovery and Development Platform, Human Antibody Therapeutics, Lund University, Lund, Sweden.,U-READ, Lund School of Technology, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Kim YW, Singh A, Shannon CP, Thiele J, Steacy LM, Ellis AK, Neighbour H, Gliddon DR, Hickey PLC, Larché M, Tebbutt SJ. Investigating Immune Gene Signatures in Peripheral Blood from Subjects with Allergic Rhinitis Undergoing Nasal Allergen Challenge. THE JOURNAL OF IMMUNOLOGY 2017; 199:3395-3405. [DOI: 10.4049/jimmunol.1700378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/14/2017] [Indexed: 12/31/2022]
|
19
|
Li C, Fu Y, Wang Y, Kong Y, Li M, Ma D, Zhai W, Wang H, Lin Y, Liu S, Ren F, Li J, Wang Y. Mesenchymal stromal cells ameliorate acute allergic rhinitis in rats. Cell Biochem Funct 2017; 35:420-425. [PMID: 28940415 PMCID: PMC5698748 DOI: 10.1002/cbf.3291] [Citation(s) in RCA: 16] [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: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been extensively investigated as a potential antiinflammatory treatment in many inflammatory‐related diseases; however, it remains unclear whether MSCs could be used to treat acute allergic rhinitis. A rat model of allergic rhinitis was treated with MSCs. The effect of MSCs on the inflammation of allergic rhinitis was evaluated by sneezing, nose rubbing, the pathology of the nasal mucosa, and the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum of rats. Also, the population of MSCs isolated from umbilical cords of humans was evaluated to determine if they could inhibit the symptoms and inflammation of acute allergic rhinitis in a rat model. We observed that this population of cells inhibited sneezing, nose rubbing, and changes in the pathology of the nasal mucosa. Intriguingly, we observed that MSCs reduced the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum. Furthermore, MSCs reduced the expression of histamine and the recruitment of macrophages in the nasal mucosa of allergic rhinitis rats. We reasoned that the effect of MSCs on allergic rhinitis might be through its regulation of the secretion of related cytokines from macrophages during the process of acute allergic rhinitis. This work suggested that MSCs from the umbilical cords of humans could be used as a positive clinical therapy for the human disease.
Collapse
Affiliation(s)
- Chunlei Li
- Graduate School of Beijing University of Chinese Medicine, Clinical Medical School of Beijing University of Chinese MedicineBeijing University of Chinese MedicineBeijingChina
- National Clinical Research Center of Respiratory Diseases, Center for Respiratory Diseases China‐Japan Friendship Hospital, The 2nd Pulmonary Department of TCM, The Key Institute of State Administration of Traditional Chinese Medicine (Pneumonopathy Chronic Cough and Dyspnea), Beijing Key Laboratory (no BZ0321)China‐Japan Friendship HospitalBeijingChina
| | - Yanxia Fu
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Yanhua Kong
- National Clinical Research Center of Respiratory Diseases, Center for Respiratory Diseases China‐Japan Friendship Hospital, The 2nd Pulmonary Department of TCM, The Key Institute of State Administration of Traditional Chinese Medicine (Pneumonopathy Chronic Cough and Dyspnea), Beijing Key Laboratory (no BZ0321)China‐Japan Friendship HospitalBeijingChina
| | - Mengdi Li
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Danhui Ma
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Wanli Zhai
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Hao Wang
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Yuting Lin
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Sihan Liu
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Jun Li
- Institute of Immunology, Medical SchoolThird Military Medical UniversityChongqingChina
| | - Yi Wang
- State Key Laboratory of Membrane Biology, School of MedicineTsinghua UniversityBeijingChina
| |
Collapse
|
20
|
Sullivan KM, Enoch SJ, Ezendam J, Sewald K, Roggen EL, Cochrane S. An Adverse Outcome Pathway for Sensitization of the Respiratory Tract by Low-Molecular-Weight Chemicals: Building Evidence to Support the Utility ofIn VitroandIn SilicoMethods in a Regulatory Context. ACTA ACUST UNITED AC 2017. [DOI: 10.1089/aivt.2017.0010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Kristie M. Sullivan
- Physicians Committee for Responsible Medicine, Washington, District of Columbia
| | - Steven J. Enoch
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, England
| | - Janine Ezendam
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, The Netherlands
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Erwin L. Roggen
- 3Rs Management & Consulting ApS (3RsMC ApS), Lyngby, Denmark
| | | |
Collapse
|
21
|
Jiménez-Saiz R, Chu DK, Mandur TS, Walker TD, Gordon ME, Chaudhary R, Koenig J, Saliba S, Galipeau HJ, Utley A, King IL, Lee K, Ettinger R, Waserman S, Kolbeck R, Jordana M. Lifelong memory responses perpetuate humoral T H2 immunity and anaphylaxis in food allergy. J Allergy Clin Immunol 2017; 140:1604-1615.e5. [PMID: 28216433 DOI: 10.1016/j.jaci.2017.01.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/10/2017] [Accepted: 01/26/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND A number of food allergies (eg, fish, shellfish, and nuts) are lifelong, without any disease-transforming therapies, and unclear in their underlying immunology. Clinical manifestations of food allergy are largely mediated by IgE. Although persistent IgE titers have been attributed conventionally to long-lived IgE+ plasma cells (PCs), this has not been directly and comprehensively tested. OBJECTIVE We sought to evaluate mechanisms underlying persistent IgE and allergic responses to food allergens. METHODS We used a model of peanut allergy and anaphylaxis, various knockout mice, adoptive transfer experiments, and in vitro assays to identify mechanisms underlying persistent IgE humoral immunity over almost the entire lifespan of the mouse (18-20 months). RESULTS Contrary to conventional paradigms, our data show that clinically relevant lifelong IgE titers are not sustained by long-lived IgE+ PCs. Instead, lifelong reactivity is conferred by allergen-specific long-lived memory B cells that replenish the IgE+ PC compartment. B-cell reactivation requires allergen re-exposure and IL-4 production by CD4 T cells. We define the half-lives of antigen-specific germinal centers (23.3 days), IgE+ and IgG1+ PCs (60 and 234.4 days, respectively), and clinically relevant cell-bound IgE (67.3 days). CONCLUSIONS These findings can explain lifelong food allergies observed in human subjects as the consequence of allergen exposures that recurrently activate memory B cells and identify these as a therapeutic target with disease-transforming potential.
Collapse
Affiliation(s)
- Rodrigo Jiménez-Saiz
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Derek K Chu
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Talveer S Mandur
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Tina D Walker
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Melissa E Gordon
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Roopali Chaudhary
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joshua Koenig
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Saliba
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Heather J Galipeau
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Adam Utley
- Departments of Immunology and Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Irah L King
- Department of Microbiology & Immunology, McGill University, Montreal, Quebec, Canada
| | - Kelvin Lee
- Departments of Immunology and Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Rachel Ettinger
- Department of Respiratory, Inflammation & Autoimmunity, MedImmune, Gaithersburg, Md
| | - Susan Waserman
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Roland Kolbeck
- Department of Respiratory, Inflammation & Autoimmunity, MedImmune, Gaithersburg, Md
| | - Manel Jordana
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
22
|
Initiation, Persistence and Exacerbation of Food Allergy. BIRKHÄUSER ADVANCES IN INFECTIOUS DISEASES 2017. [DOI: 10.1007/978-3-319-69968-4_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
23
|
Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Mechanisms of Aeroallergen Immunotherapy: Subcutaneous Immunotherapy and Sublingual Immunotherapy. Immunol Allergy Clin North Am 2016; 36:71-86. [PMID: 26617228 DOI: 10.1016/j.iac.2015.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Allergen immunotherapy (AIT) is an effective way to treat allergic disorders, targeting the underlying mechanisms and altering the disease course by inducing a long-lasting clinical and immune tolerance to allergens. Although sublingual and subcutaneous routes are used in daily practice, many novel ways to decrease side effects and duration and increase efficacy have been pursued. Further studies are needed to develop biomarkers for the identification of AIT responder patients and also to use the developed knowledge in allergy prevention studies. Future directions in AIT include treatments for autoimmune diseases, chronic infections, organ transplantation, and breaking immune tolerance to cancer cells.
Collapse
Affiliation(s)
- Cevdet Ozdemir
- Department of Pediatric Allergy, Memorial Atasehir Hospital, Memorial Health Group, Vedat Gunyol Cad. 28-30, Istanbul 34758, Turkey
| | - Umut Can Kucuksezer
- Department of Immunology, Institute of Experimental Medicine (DETAE), Vakif Gureba Cad, Istanbul University, Istanbul 34093, Turkey
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Obere Strasse, CH-7270, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Obere Strasse, CH-7270, Davos, Switzerland.
| |
Collapse
|
24
|
Affiliation(s)
- A. O. Eifan
- Allergy and Clinical Immunology; Faculty of Medicine; National Heart and Lung Institute; Imperial College London; London UK
| | - S. R. Durham
- Allergy and Clinical Immunology; Faculty of Medicine; National Heart and Lung Institute; Imperial College London; London UK
| |
Collapse
|
25
|
Aalberse RC, Rispens T. A Tale of 2 Tails: The Interpretation of Changes in Allergen-Specific IgE Following Incidental Allergen Exposure. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:246-7. [PMID: 26968962 DOI: 10.1016/j.jaip.2016.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 12/31/2015] [Accepted: 01/05/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Rob C Aalberse
- Division of Research, Department of Immunopathology, Sanquin Blood Supply, Amsterdam, The Netherlands; Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Theo Rispens
- Division of Research, Department of Immunopathology, Sanquin Blood Supply, Amsterdam, The Netherlands; Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Noble A, Zhao J. Follicular helper T cells are responsible for IgE responses to Der p 1 following house dust mite sensitization in mice. Clin Exp Allergy 2016; 46:1075-82. [PMID: 27138589 DOI: 10.1111/cea.12750] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 02/18/2016] [Accepted: 04/28/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Th2 cells have long been considered responsible for the switching of B cells to production of IgE during cognate interaction, primarily due to their expression of CD40L and secretion of IL-4. This concept has been challenged by the more recent definition of follicular helper T cells (Tfh) as the key T cell subset in B cell isotype switching, due to their physical location at the boundary of T cell:B cell areas in lymphoid follicles and ability to express IL-4 and CD40L. OBJECTIVE To determine whether Tfh cells are responsible for IgE responses to Der p 1 allergen after house dust mite (HDM)-induced allergic sensitization. METHODS Mice deficient in Tfh cells were sensitized to HDM and Der p 1-specific IgE measured by ELISA. RESULTS Mice with a mutation in T cell-expressed IL-6R were unable to expand Tfh populations after HDM sensitization, and their anti-Der p 1 IgE, IgG1 and total IgE responses were reduced by 80-90% compared with wild-type mice. These animals displayed unaltered lung Th2 and eosinophilic responses after intranasal HDM challenge and normal IL-4 production, but B cell infiltration of the airways was abrogated. CONCLUSIONS AND CLINICAL RELEVANCE Our data indicate that Tfh cells are largely responsible for switching B cells to IgE synthesis, most likely via an IgG1(+) intermediate. However, Th2 cells are the major source of IL-4 during HDM sensitization and this might contribute to IgE synthesis at a stage distal to Tfh-mediated isotype switching. The IL-6/follicular helper T cell pathway is a potential therapeutic target in allergic disease.
Collapse
Affiliation(s)
- A Noble
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - J Zhao
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| |
Collapse
|
27
|
Aalberse RC, Platts-Mills TA, Rispens T. The Developmental History of IgE and IgG4 Antibodies in Relation to Atopy, Eosinophilic Esophagitis, and the Modified TH2 Response. Curr Allergy Asthma Rep 2016; 16:45. [PMID: 27221343 PMCID: PMC5026408 DOI: 10.1007/s11882-016-0621-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A common reaction from anyone confronted with allergy is the question: what prevents universal allergy? We will discuss recent findings in the mouse system that have provided us with clues on why allergy is not more common. We will also address one crucial aspect of atopic allergy in humans, which is absent in most mouse model systems, an IgG/IgE ratio <10. We consider the typical mouse IgE response to be more closely related to the "modified TH2" response in humans. We will discuss the similarities and differences between the IgE and IgG4 response to allergens and an update on the IgG4 B cell, partly derived from studies on eosinophilic esophagitis and IgG4-related diseases.
Collapse
Affiliation(s)
- Rob C Aalberse
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, P.O. Box 9190, 1006 AD, Amsterdam, The Netherlands.
| | - Thomas A Platts-Mills
- Division of Allergy and Immunology, University of Virginia, Charlottesville, VA, USA
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, P.O. Box 9190, 1006 AD, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Stentzel S, Teufelberger A, Nordengrün M, Kolata J, Schmidt F, van Crombruggen K, Michalik S, Kumpfmüller J, Tischer S, Schweder T, Hecker M, Engelmann S, Völker U, Krysko O, Bachert C, Bröker BM. Staphylococcal serine protease-like proteins are pacemakers of allergic airway reactions to Staphylococcus aureus. J Allergy Clin Immunol 2016; 139:492-500.e8. [PMID: 27315768 DOI: 10.1016/j.jaci.2016.03.045] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/15/2016] [Accepted: 03/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND A substantial subgroup of asthmatic patients have "nonallergic" or idiopathic asthma, which often takes a severe course and is difficult to treat. The cause might be allergic reactions to the gram-positive pathogen Staphylococcus aureus, a frequent colonizer of the upper airways. However, the driving allergens of S aureus have remained elusive. OBJECTIVE We sought to search for potentially allergenic S aureus proteins and characterize the immune response directed against them. METHODS S aureus extracellular proteins targeted by human serum IgG4 were identified by means of immunoblotting to screen for potential bacterial allergens. Candidate antigens were expressed as recombinant proteins and used to analyze the established cellular and humoral immune responses in healthy adults and asthmatic patients. The ability to induce a type 2 immune response in vivo was tested in a mouse asthma model. RESULTS We identified staphylococcal serine protease-like proteins (Spls) as dominant IgG4-binding S aureus proteins. SplA through SplF are extracellular proteases of unknown function expressed by S aureus in vivo. Spls elicited IgE antibody responses in most asthmatic patients. In healthy S aureus carriers and noncarriers, peripheral blood T cells elaborated TH2 cytokines after stimulation with Spls, as is typical for allergens. In contrast, TH1/TH17 cytokines, which dominated the response to S aureus α-hemolysin, were of low concentration or absent. In mice inhalation of SplD without adjuvant induced lung inflammation characterized by TH2 cytokines and eosinophil infiltration. CONCLUSION We identify Spls as triggering allergens released by S aureus, opening prospects for diagnosis and causal therapy of asthma.
Collapse
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Julia Kolata
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany; Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank Schmidt
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | | | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | - Jana Kumpfmüller
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, HKI, Jena, Germany
| | - Sebastian Tischer
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Schweder
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Michael Hecker
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Susanne Engelmann
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Institute for Microbiology, University of Braunschweig, Braunschweig, Germany; Helmholtz Center for Infection Research, Microbial Proteomics, Braunschweig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Olga Krysko
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium; Division of Ear, Nose, and Throat Diseases, Clintec, Karolinska Institute, Stockholm, Sweden
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
29
|
Persistence of the IgE repertoire in birch pollen allergy. J Allergy Clin Immunol 2016; 137:1884-1887.e8. [PMID: 27001158 DOI: 10.1016/j.jaci.2015.12.1333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/15/2015] [Accepted: 12/29/2015] [Indexed: 01/04/2023]
|
30
|
Looney TJ, Lee JY, Roskin KM, Hoh RA, King J, Glanville J, Liu Y, Pham TD, Dekker CL, Davis MM, Boyd SD. Human B-cell isotype switching origins of IgE. J Allergy Clin Immunol 2016; 137:579-586.e7. [PMID: 26309181 PMCID: PMC4747810 DOI: 10.1016/j.jaci.2015.07.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND B cells expressing IgE contribute to immunity against parasites and venoms and are the source of antigen specificity in allergic patients, yet the developmental pathways producing these B cells in human subjects remain a subject of debate. Much of our knowledge of IgE lineage development derives from model studies in mice rather than from human subjects. OBJECTIVE We evaluate models for isotype switching to IgE in human subjects using immunoglobulin heavy chain (IGH) mutational lineage data. METHODS We analyzed IGH repertoires in 9 allergic and 24 healthy adults using high-throughput DNA sequencing of 15,843,270 IGH rearrangements to identify clonal lineages of B cells containing members expressing IgE. Somatic mutations in IGH inherited from common ancestors within the clonal lineage are used to infer the relationships between B cells. RESULTS Data from 613,641 multi-isotype B-cell clonal lineages, of which 592 include an IgE member, are consistent with indirect switching to IgE from IgG- or IgA-expressing lineage members in human subjects. We also find that these inferred isotype switching frequencies are similar in healthy and allergic subjects. CONCLUSIONS We found evidence that secondary isotype switching of mutated IgG1-expressing B cells is the primary source of IgE in human subjects, with lesser contributions from precursors expressing other switched isotypes and rarely IgM or IgD, suggesting that IgE is derived from previously antigen-experienced B cells rather than naive B cells that typically express low-affinity unmutated antibodies. These data provide a basis from which to evaluate allergen-specific human antibody repertoires in healthy and diseased subjects.
Collapse
Affiliation(s)
- Timothy J Looney
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Ji-Yeun Lee
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Krishna M Roskin
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Ramona A Hoh
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Jasmine King
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif; Department of Biology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Jacob Glanville
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif; Program in Immunology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Yi Liu
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif; Biomedical Informatics Training Program, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Tho D Pham
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Cornelia L Dekker
- Department of Pediatrics, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Mark M Davis
- Department of Microbiology & Immunology, Transplantation and Infection, Stanford University, Stanford, Calif; Howard Hughes Medical Institute, Transplantation and Infection, Stanford University, Stanford, Calif; Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, Calif
| | - Scott D Boyd
- Department of Pathology, Transplantation and Infection, Stanford University, Stanford, Calif.
| |
Collapse
|
31
|
Ramadani F, Upton N, Hobson P, Chan YC, Mzinza D, Bowen H, Kerridge C, Sutton BJ, Fear DJ, Gould HJ. Intrinsic properties of germinal center-derived B cells promote their enhanced class switching to IgE. Allergy 2015; 70:1269-77. [PMID: 26109279 PMCID: PMC4744720 DOI: 10.1111/all.12679] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND Research on the origins and development of human IgE-expressing (IgE(+) ) cells is required for understanding the pathogenesis of allergy and asthma. These studies have been thwarted by the rarity of IgE(+) cells in vivo and the low frequency of class switch recombination (CSR) to IgE ex vivo. To determine the main source of IgE(+) cells, we investigated the relation between the phenotypic composition of tonsil B cells and the CSR to IgE ex vivo. METHODS Human tonsil B cells were analyzed by flow cytometry (FACS) and cultured with IL-4 and anti-CD40 to induce CSR to IgE. Naïve, germinal center (GC), early GC (eGC), and memory tonsil B cells were isolated by FACS, and their capacities for IL-4 and anti-CD40 signaling, cell proliferation, and de novo class switching to IgE were analyzed by RT-PCR and FACS. RESULTS B cells from different tonsils exhibited varying capacities for CSR to IgE ex vivo. This was correlated with the percentage of eGC B cells in the tonsil at the outset of the culture. Despite relatively poor cell viability, eGC and GC B-cell cultures produced the highest yields of IgE(+) cells compared to naïve and memory B-cell cultures. The main factors accounting for this result were the strength of IL-4R and CD40 signaling and relative rates of cell proliferation. CONCLUSIONS This study shows that the maturation state of tonsil B cells determines their capacity to undergo class switching to IgE ex vivo, with the GC-derived B cells yielding the highest percentage of IgE(+) cells.
Collapse
Affiliation(s)
- F Ramadani
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - N Upton
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - P Hobson
- Division of Asthma, Allergy and Lung Biology, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - Y-C Chan
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - D Mzinza
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - H Bowen
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - C Kerridge
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - B J Sutton
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - D J Fear
- Division of Asthma, Allergy and Lung Biology, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| | - H J Gould
- Randall Division of Cell and Molecular Biohphysics, King's College London, London, UK
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, UK
| |
Collapse
|
32
|
Abstract
Lipocalins are one of the most important groups of inhalant animal allergens. The analysis of structural features of these proteins is important to get insights into their allergenicity. We have determined two different dimeric crystal structures for bovine dander lipocalin Bos d 2, which was earlier described as a monomeric allergen. The crystal structure analysis of all other determined lipocalin allergens also revealed oligomeric structures which broadly utilize inherent structural features of the β-sheet in dimer formation. According to the moderate size of monomer-monomer interfaces, most of these dimers would be transient in solution. Native mass spectrometry was employed to characterize quantitatively transient dimerization of two lipocalin allergens, Bos d 2 and Bos d 5, in solution.
Collapse
|
33
|
Nony E, Timbrell V, Hrabina M, Boutron M, Solley G, Moingeon P, Davies JM. Specific IgE recognition of pollen allergens from subtropic grasses in patients from the subtropics. Ann Allergy Asthma Immunol 2015; 114:214-220.e2. [PMID: 25744907 DOI: 10.1016/j.anai.2014.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 11/17/2014] [Accepted: 12/08/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND Pollens of subtropical grasses, Bahia (Paspalum notatum), Johnson (Sorghum halepense), and Bermuda (Cynodon dactylon), are common causes of respiratory allergies in subtropical regions worldwide. OBJECTIVE To evaluate IgE cross-reactivity of grass pollen (GP) found in subtropical and temperate areas. METHODS Case and control serum samples from 83 individuals from the subtropical region of Queensland were tested for IgE reactivity with GP extracts by enzyme-linked immunosorbent assay. A randomly sampled subset of 21 serum samples from patients with subtropical GP allergy were examined by ImmunoCAP and cross-inhibition assays. RESULTS Fifty-four patients with allergic rhinitis and GP allergy had higher IgE reactivity with P notatum and C dactylon than with a mixture of 5 temperate GPs. For 90% of 21 GP allergic serum samples, P notatum, S halepense, or C dactylon specific IgE concentrations were higher than temperate GP specific IgE, and GP specific IgE had higher correlations of subtropical GP (r = 0.771-0.950) than temperate GP (r = 0.317-0.677). In most patients (71%-100%), IgE with P notatum, S halepense, or C dactylon GPs was inhibited better by subtropical GP than temperate GP. When the temperate GP mixture achieved 50% inhibition of IgE with subtropical GP, there was a 39- to 67-fold difference in concentrations giving 50% inhibition and significant differences in maximum inhibition for S halepense and P notatum GP relative to temperate GP. CONCLUSION Patients living in a subtropical region had species specific IgE recognition of subtropical GP. Most GP allergic patients in Queensland would benefit from allergen specific immunotherapy with a standardized content of subtropical GP allergens.
Collapse
Affiliation(s)
| | - Victoria Timbrell
- The University of Queensland, School of Medicine, Brisbane, Australia
| | | | | | | | | | - Janet M Davies
- The University of Queensland, School of Medicine, Brisbane, Australia
| |
Collapse
|
34
|
Wu YCB, James LK, Vander Heiden JA, Uduman M, Durham SR, Kleinstein SH, Kipling D, Gould HJ. Influence of seasonal exposure to grass pollen on local and peripheral blood IgE repertoires in patients with allergic rhinitis. J Allergy Clin Immunol 2015; 134:604-12. [PMID: 25171866 PMCID: PMC4151999 DOI: 10.1016/j.jaci.2014.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 04/11/2014] [Accepted: 04/25/2014] [Indexed: 11/18/2022]
Abstract
Background Previous studies of immunoglobulin gene sequences in patients with allergic diseases using low-throughput Sanger sequencing have limited the analytic depth for characterization of IgE repertoires. Objectives We used a high-throughput, next-generation sequencing approach to characterize immunoglobulin heavy-chain gene (IGH) repertoires in patients with seasonal allergic rhinitis (AR) with the aim of better understanding the underlying disease mechanisms. Methods IGH sequences in matched peripheral blood and nasal biopsy specimens from nonallergic healthy control subjects (n = 3) and patients with grass pollen–related AR taken in season (n = 3) or out of season (n = 4) were amplified and pyrosequenced on the 454 GS FLX+ System. Results A total of 97,610 IGH (including 8,135 IgE) sequences were analyzed. Use of immunoglobulin heavy-chain variable region gene families 1 (IGHV1) and 5 (IGHV5) was higher in IgE clonotypic repertoires compared with other antibody classes independent of atopic status. IgE repertoires measured inside the grass pollen season were more diverse and more mutated (particularly in the biopsy specimens) and had more evidence of antigen-driven selection compared with those taken outside of the pollen season or from healthy control subjects. Clonal relatedness was observed for IgE between the blood and nasal biopsy specimens. Furthermore in patients with AR, but not healthy control subjects, we found clonal relatedness between IgE and IgG classes. Conclusion This is the first report that exploits next-generation sequencing to determine local and peripheral blood IGH repertoires in patients with respiratory allergic disease. We demonstrate that natural pollen exposure was associated with changes in IgE repertoires that were suggestive of ongoing germinal center reactions. Furthermore, these changes were more often apparent in nasal biopsy specimens compared with peripheral blood and in patients with AR compared with healthy control subjects.
Collapse
Affiliation(s)
- Yu-Chang B Wu
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, United Kingdom.
| | - Louisa K James
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, United Kingdom
| | - Jason A Vander Heiden
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, Conn
| | - Mohamed Uduman
- Department of Pathology, Yale School of Medicine, New Haven, Conn
| | - Stephen R Durham
- Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, United Kingdom; Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Steven H Kleinstein
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, Conn; Department of Pathology, Yale School of Medicine, New Haven, Conn
| | - David Kipling
- Institute of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Hannah J Gould
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; Medical Research Council and Asthma UK Centre, Allergic Mechanisms in Asthma, London, United Kingdom
| |
Collapse
|
35
|
Wong KJ, Timbrell V, Xi Y, Upham JW, Collins AM, Davies JM. IgE+ B cells are scarce, but allergen-specific B cells with a memory phenotype circulate in patients with allergic rhinitis. Allergy 2015; 70:420-8. [PMID: 25556717 DOI: 10.1111/all.12563] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND Despite the critical role of immunoglobulin E (IgE) in allergy, circulating IgE+ B cells are scarce. Here, we describe in patients with allergic rhinitis B cells with a memory phenotype responding to a prototypic aeroallergen. METHODS Fifteen allergic rhinitis patients with grass pollen allergy and 13 control subjects were examined. Blood mononuclear cells stained with carboxyfluorescein diacetate succinimidyl ester (CFSE) were cultured with Bahia grass pollen. Proliferation and phenotype were assessed by multicolour flow cytometry. RESULTS In blood of allergic rhinitis patients with high serum IgE to grass pollen, most IgE(hi) cells were CD123+ HLA-DR(-) basophils, with IgE for the major pollen allergen (Pas n 1). Both B and T cells from pollen-allergic donors showed higher proliferation to grass pollen than nonallergic donors (P = 0.002, and 0.010, respectively), whereas responses to vaccine antigens and mitogen did not differ between groups. Allergen-driven B cells that divided rapidly (CD19(mid) CD3(-) CFSE(lo) ) showed higher CD27 (P = 0.008) and lower CD19 (P = 0.004) and CD20 (P = 0.004) expression than B cells that were slow to respond to allergen (CD19(hi) CD3(-) CFSE(mid) ). Moreover, rapidly dividing allergen-driven B cells (CD19(mid) CFSE(lo) CD27(hi) ) showed higher expression of the plasmablast marker CD38 compared with B cells (CD19(hi) CFSE(mid) CD27(lo) ) that were slow to divide. CONCLUSION Patients with pollen allergy but not control donors have a population of circulating allergen-specific B cells with the phenotype and functional properties of adaptive memory B-cell responses. These cells could provide precursors for allergen-specific IgE production upon allergen re-exposure.
Collapse
Affiliation(s)
- K. J. Wong
- Lung and Allergy Research Centre; School of Medicine; The University of Queensland; Woolloongabba Qld Australia
| | - V. Timbrell
- Lung and Allergy Research Centre; School of Medicine; The University of Queensland; Woolloongabba Qld Australia
| | - Y. Xi
- Lung and Allergy Research Centre; School of Medicine; The University of Queensland; Woolloongabba Qld Australia
| | - J. W. Upham
- Lung and Allergy Research Centre; School of Medicine; The University of Queensland; Woolloongabba Qld Australia
- Department of Respiratory Medicine; Princess Alexandra Hospital; Woolloongabba Qld Australia
| | - A. M. Collins
- School of Biotechnology and Biomolecular Sciences; University of New South Wales; Kensington NSW Australia
| | - J. M. Davies
- Lung and Allergy Research Centre; School of Medicine; The University of Queensland; Woolloongabba Qld Australia
| |
Collapse
|
36
|
Tripathi A, Commins SP, Heymann PW, Platts-Mills TAE. Delayed anaphylaxis to red meat masquerading as idiopathic anaphylaxis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2015; 2:259-65. [PMID: 24811014 DOI: 10.1016/j.jaip.2014.02.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 11/24/2022]
Abstract
Anaphylaxis is traditionally recognized as a rapidly developing combination of symptoms that often includes hives and hypotension or respiratory symptoms. Furthermore, when a specific cause is identified, exposure to this cause is usually noted to have occurred within minutes to 2 hours before the onset of symptoms. This case is of a 79-year-old woman who developed a severe episode of anaphylaxis 3 hours after eating pork. Before 2012, she had not experienced any symptoms after ingestion of meat products. Delayed anaphylaxis to mammalian meat has many contrasting features to immediate food-induced anaphylaxis. The relevant IgE antibody is specific for the oligosaccharide galactose-alpha-1,3-galactose, a blood group substance of nonprimate mammals. There is evidence from Australia, Sweden, and the United States that the primary cause of this IgE antibody response is tick bites. These bites characteristically itch for 10 days or more. Diagnosis can be made by the presence of specific IgE to beef, pork, lamb, and milk, and the lack of IgE to chicken, turkey, and fish. Skin prick tests (but not intradermal tests) generally are negative. Management of these cases, now common across the southeastern United States, consists of education combined with avoidance of both ingestion of red meat and further tick bites.
Collapse
Affiliation(s)
- Anubha Tripathi
- Asthma and Allergic Diseases Center, University of Virginia Medical Center, Charlottesville, Va
| | - Scott P Commins
- Asthma and Allergic Diseases Center, University of Virginia Medical Center, Charlottesville, Va
| | - Peter W Heymann
- Asthma and Allergic Diseases Center, University of Virginia Medical Center, Charlottesville, Va
| | - Thomas A E Platts-Mills
- Asthma and Allergic Diseases Center, University of Virginia Medical Center, Charlottesville, Va.
| |
Collapse
|
37
|
Aalberse RC, Knol EF. Historic overview of allergy research in the Netherlands. Immunol Lett 2014; 162:163-72. [PMID: 25455604 DOI: 10.1016/j.imlet.2014.10.015] [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: 11/18/2022]
Abstract
Research in allergy has a long history in the Netherlands, although the relation with immunology has not always been appreciated. In many aspects Dutch researchers have made major contribution in allergy research. This ranges from the first characterization of house dust mite as an important allergen, the first characterization of human Th2 and Th1 T cell clones, to the development of diagnostic test systems. In this overview Aalberse and Knol have made an overview of the major contributions of Dutch immunologists in allergy.
Collapse
Affiliation(s)
- Rob C Aalberse
- Department of Immunopathology, Sanquin Blood Supply Foundation and Academic Medical Centre, Amsterdam, The Netherlands
| | - Edward F Knol
- Department of Immunology, University Medical Center Utrecht, The Netherlands; Department of Dermatology and Allergology, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
38
|
Cheng LE, Locksley RM. Allergic inflammation--innately homeostatic. Cold Spring Harb Perspect Biol 2014; 7:a016352. [PMID: 25414367 DOI: 10.1101/cshperspect.a016352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Allergic inflammation is associated closely with parasite infection but also asthma and other common allergic diseases. Despite the engagement of similar immunologic pathways, parasitized individuals often show no outward manifestations of allergic disease. In this perspective, we present the thesis that allergic inflammatory responses play a primary role in regulating circadian and environmental inputs involved with tissue homeostasis and metabolic needs. Parasites feed into these pathways and thus engage allergic inflammation to sustain aspects of the parasitic life cycle. In response to parasite infection, an adaptive and regulated immune response is layered on the host effector response, but in the setting of allergy, the effector response remains unregulated, thus leading to the cardinal features of disease. Further understanding of the homeostatic pressures driving allergic inflammation holds promise to further our understanding of human health and the treatment of these common afflictions.
Collapse
Affiliation(s)
- Laurence E Cheng
- Department of Pediatrics, University of California, San Francisco, San Francisco, California 94143
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, California 94143 Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143 Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
39
|
Rogosch T, Kerzel S, Dey F, Wagner JJ, Zhang Z, Maier RF, Zemlin M. IgG4 and IgE transcripts in childhood allergic asthma reflect divergent antigen-driven selection. THE JOURNAL OF IMMUNOLOGY 2014; 193:5801-8. [PMID: 25385824 DOI: 10.4049/jimmunol.1401409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The physiologic function of the "odd" Ab IgG4 remains enigmatic. IgG4 mediates immunotolerance, as, for example, during specific immunotherapy of allergies, but it mediates tissue damage in autoimmune pemphigus vulgaris and "IgG4-related disease." Approximately half of the circulating IgG4 molecules are bispecific owing to their unique ability to exchange half-molecules. Better understanding of the interrelation between IgG4 and IgE repertoires may yield insight into the pathogenesis of allergies and into potential novel therapies that modulate IgG4 responses. We aimed to compare the selective forces that forge the IgG4 and IgE repertoires in allergic asthma. Using an IgG4-specific RT-PCR, we amplified, cloned, and sequenced IgG4 H chain transcripts of PBMCs from 10 children with allergic asthma. We obtained 558 functional IgG4 sequences, of which 286 were unique. Compared with previously published unique IgE transcripts from the same blood samples, the somatic mutation rate was significantly enhanced in IgG4 transcripts (62 versus 83%; p < 0.001), whereas fewer IgG4 sequences displayed statistical evidence of Ag-driven selection (p < 0.001). On average, the hypervariable CDRH3 region was four nucleotides shorter in IgG4 than in IgE transcripts (p < 0.001). IgG4 transcripts in the circulation of children with allergic asthma reflect some characteristics of classical Ag-driven B2 immune responses but display less indication of Ag selection than do IgE transcripts. Although allergen-specific IgG4 can block IgE-mediated allergen presentation and degranulation of mast cells, key factors that influence the Ag-binding properties of the Ab differ between the overall repertoires of circulating IgG4- and IgE-expressing cells.
Collapse
Affiliation(s)
- Tobias Rogosch
- Department of Pediatrics, Philipps-University Marburg, D-35033 Marburg, Germany
| | - Sebastian Kerzel
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg, St. Hedwig Campus, 93053 Regensburg, Germany; and
| | - Friederike Dey
- Department of Pediatrics, Philipps-University Marburg, D-35033 Marburg, Germany
| | | | - Zhixin Zhang
- Department of Pathology and Microbiology, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198
| | - Rolf F Maier
- Department of Pediatrics, Philipps-University Marburg, D-35033 Marburg, Germany
| | - Michael Zemlin
- Department of Pediatrics, Philipps-University Marburg, D-35033 Marburg, Germany;
| |
Collapse
|
40
|
Berkowska MA, Heeringa JJ, Hajdarbegovic E, van der Burg M, Thio HB, van Hagen PM, Boon L, Orfao A, van Dongen JJ, van Zelm MC. Human IgE+ B cells are derived from T cell–dependent and T cell–independent pathways. J Allergy Clin Immunol 2014; 134:688-697.e6. [DOI: 10.1016/j.jaci.2014.03.036] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 03/14/2014] [Accepted: 03/25/2014] [Indexed: 12/11/2022]
|
41
|
Wu LC, Scheerens H. Targeting IgE production in mice and humans. Curr Opin Immunol 2014; 31:8-15. [PMID: 25156315 DOI: 10.1016/j.coi.2014.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 11/26/2022]
Abstract
Immunoglobulin E (IgE) is pathogenic in allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, and food allergy. Recent studies using genetically modified IgE reporter mice indicate that the majority of serum IgE in mice is produced by short-lived IgE plasma cells, with minor contributions from long-lived IgE plasma cells, and implicate IgG1 and IgE memory B cells as potential sources of IgE memory. Clinical studies using antibodies against IL-13 or the IL-4 and IL-13 receptor subunit IL-4Rα, as well as an antibody against the M1 prime domain of human membrane IgE, indicate that, similar to mice, a proportion of IgE in humans is derived from ongoing IgE immune responses and short-lived plasma cells. Targeting IgE production may lead to new therapies for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Lawren C Wu
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Heleen Scheerens
- Department of Pharmacodynamic Biomarkers, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
42
|
Erwin EA, Woodfolk JA, James HR, Satinover SM, Platts-Mills TAE. Changes in cat specific IgE and IgG antibodies with decreased cat exposure. Ann Allergy Asthma Immunol 2014; 112:545-550.e1. [PMID: 24726651 DOI: 10.1016/j.anai.2014.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/13/2014] [Accepted: 03/16/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Current understanding of the effects of reducing exposure to cat allergens is limited. It has also become clear that there are different forms of immune response to cat allergens. OBJECTIVE To investigate changes in skin tests and cat specific IgG and IgE antibodies when students from a home with a cat move to a college dormitory. METHODS Ninety-seven college students participated in a prospective study that consisted of allergy skin prick testing and serum measurement of IgE and IgG antibodies to cat at the beginning and end of one academic year in college. A subgroup returned for follow-up at the end of 2 years. RESULTS Among 97 students, 33% had IgG antibodies to Fel d 1 but no evidence of sensitization, 25% had positive skin test results and/or serum IgE antibodies, and 42% had negative skin test results and no detectable serum antibodies. Among the non-cat sensitized students with IgG antibodies, the titers decreased during 8 months (P = .002). Titers of IgG4 to Fel d 1 also decreased (P < .001). Among the sensitized students, no change in IgE antibodies to cat occurred in 8 months (P = .20), whereas Fel d 1 specific IgG antibodies decreased (P < .001). Thus, ratios of IgG to IgE decreased highly significantly (P = .007). Among the students with negative skin test results who returned for follow-up (n = 56), none developed positive skin test results or serum IgE antibodies. CONCLUSION Under conditions of marked decrease in exposure, no participants developed new-onset sensitization. Among the individuals sensitized at study entry, there were major decreases in the ratio of IgG to IgE.
Collapse
Affiliation(s)
- Elizabeth A Erwin
- Center for Innovation in Pediatric Practice, Nationwide Children's Hospital, Columbus, Ohio.
| | - Judith A Woodfolk
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, Virginia
| | - Hayley R James
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, Virginia
| | - Shama M Satinover
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, Virginia
| | - Thomas A E Platts-Mills
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
43
|
Wang Y, Jackson KJL, Davies J, Chen Z, Gaeta BA, Rimmer J, Sewell WA, Collins AM. IgE-associated IGHV genes from venom and peanut allergic individuals lack mutational evidence of antigen selection. PLoS One 2014; 9:e89730. [PMID: 24586993 PMCID: PMC3934916 DOI: 10.1371/journal.pone.0089730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 01/22/2014] [Indexed: 11/18/2022] Open
Abstract
Antigen selection of B cells within the germinal center reaction generally leads to the accumulation of replacement mutations in the complementarity-determining regions (CDRs) of immunoglobulin genes. Studies of mutations in IgE-associated VDJ gene sequences have cast doubt on the role of antigen selection in the evolution of the human IgE response, and it may be that selection for high affinity antibodies is a feature of some but not all allergic diseases. The severity of IgE-mediated anaphylaxis is such that it could result from higher affinity IgE antibodies. We therefore investigated IGHV mutations in IgE-associated sequences derived from ten individuals with a history of anaphylactic reactions to bee or wasp venom or peanut allergens. IgG sequences, which more certainly experience antigen selection, served as a control dataset. A total of 6025 unique IgE and 5396 unique IgG sequences were generated using high throughput 454 pyrosequencing. The proportion of replacement mutations seen in the CDRs of the IgG dataset was significantly higher than that of the IgE dataset, and the IgE sequences showed little evidence of antigen selection. To exclude the possibility that 454 errors had compromised analysis, rigorous filtering of the datasets led to datasets of 90 core IgE sequences and 411 IgG sequences. These sequences were present as both forward and reverse reads, and so were most unlikely to include sequencing errors. The filtered datasets confirmed that antigen selection plays a greater role in the evolution of IgG sequences than of IgE sequences derived from the study participants.
Collapse
Affiliation(s)
- Yan Wang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Katherine J. L. Jackson
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Janet Davies
- The Lung and Allergy Research Centre, School of Medicine, The University of Queensland, Woolloongabba, Australia
| | - Zhiliang Chen
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Bruno A. Gaeta
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | | | - William A. Sewell
- Institute of Laboratory Medicine, St Vincent's Hospital, Darlinghurst, Australia and St Vincent's Clinical School, University of New South Wales, Darlinghurst, Australia
| | - Andrew M. Collins
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
44
|
Matricardi PM. Allergen-specific immunoprophylaxis: toward secondary prevention of allergic rhinitis? Pediatr Allergy Immunol 2014; 25:15-8. [PMID: 24588481 DOI: 10.1111/pai.12200] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Paolo M Matricardi
- Department of Pediatric Pneumology and Immunology, Charité Medical School, Berlin, Germany; Social and Epidemiology Institute, Charité Medical School, Berlin, Germany.
| |
Collapse
|
45
|
Berg EA, Platts-Mills TAE, Commins SP. Drug allergens and food--the cetuximab and galactose-α-1,3-galactose story. Ann Allergy Asthma Immunol 2013; 112:97-101. [PMID: 24468247 DOI: 10.1016/j.anai.2013.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/12/2013] [Accepted: 11/17/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVE A novel form of food allergy has been described that initially became apparent from IgE reactivity with the drug cetuximab. Ongoing work regarding the etiology, distribution, clinical management, and cellular mechanisms of the IgE response to the oligosaccharide galactose-α-1,3-galactose (α-gal) is reviewed. DATA SOURCES Brief review of the relevant literature in peer-reviewed journals. STUDY SELECTION Studies on the clinical and immunologic features, pathogenesis, epidemiology, laboratory evaluation, and management of IgE to α-gal are included in this review. RESULTS Recent work has identified a novel IgE antibody response to the mammalian oligosaccharide epitope, α-gal, that has been associated with 2 distinct forms of anaphylaxis: (1) immediate-onset anaphylaxis during first exposure to intravenous cetuximab and (2) delayed-onset anaphylaxis 3 to 6 hours after ingestion of mammalian food products (eg, beef and pork). Study results have suggested that tick bites are a cause of IgE antibody responses to α-gal in the United States. Patients with IgE antibody to α-gal continue to emerge, and, increasingly, these cases involve children. Nevertheless, this IgE antibody response does not appear to pose a risk for asthma but may impair diagnostic testing in some situations. CONCLUSION The practicing physician should understand the symptoms, evaluation, and management when diagnosing delayed allergic reactions to mammalian meat from IgE to α-gal or when initiating treatment with cetuximab in patients who have developed an IgE antibody response to α-gal.
Collapse
Affiliation(s)
- Emily A Berg
- Department of Internal Medicine, Division of Allergy and Immunology, University of Virginia Health System, Charlottesville, Virginia
| | - Thomas A E Platts-Mills
- Department of Internal Medicine, Division of Allergy and Immunology, University of Virginia Health System, Charlottesville, Virginia
| | - Scott P Commins
- Department of Internal Medicine, Division of Allergy and Immunology, University of Virginia Health System, Charlottesville, Virginia; Department of Pediatrics, Division of Allergy and Immunology, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
46
|
Zakzuk J, Benedetti I, Fernández-Caldas E, Caraballo L. The influence of chitin on the immune response to the house dust mite allergen Blo T 12. Int Arch Allergy Immunol 2013; 163:119-29. [PMID: 24335274 DOI: 10.1159/000356482] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 10/16/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Information about the biological properties of Blomia tropicalis allergens is scarce. It is predicted that Blo t 12, an allergen with two described isoforms, contains a chitin-binding domain, similar to that found in peritrophins. Th2 adjuvant properties have been described for chitin. Therefore, it is feasible that binding to this carbohydrate influences its allergenicity. We aimed to evaluate the chitin-binding activity of Blo t 12 isoallergens and its effect on airway inflammation and antibody responses in a murine model of allergen sensitization. METHODS Chitin-binding assays were conducted with the recombinant isoallergens Blo t 12.0101 and Blo t 12.0102. BALB/c mice were sensitized via i.p. with any of the two isoforms (alone, with chitin or alum) and then challenged intranasally. Methacholine-induced bronchial hyperreactivity was tested by whole-body plethysmography and lung sections were stained with hematoxylin and eosin and periodic-acid Schiff. Total IgE and allergen-specific IgE, IgG1 and IgG2 levels were measured by ELISA. RESULTS The two isoforms bound chitin, but Blo t 12.0101 showed a stronger binding capacity. Both isoforms induced total and allergen-specific IgE, airway hyperreactivity, bronchial inflammation and mucus secretion without any adjuvant; however, when administered with chitin, Blo t 12.0101 induced higher total IgE levels. The IgG1/IgG2a ratio was significantly higher in mice immunized with Blo t 12.0101 than those immunized with Blo t 12.0102. As peritrophins, Blo t 12 was detected in mite feces. CONCLUSIONS Blo t 12 isoforms are chitin-binding proteins that induce airway inflammation and bronchial hyperreactivity. However, for Blo t 12.0101, chitin reinforces its effects on total IgE production.
Collapse
Affiliation(s)
- Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | | | | | | |
Collapse
|
47
|
Gadermaier E, Levin M, Flicker S, Ohlin M. The human IgE repertoire. Int Arch Allergy Immunol 2013; 163:77-91. [PMID: 24296690 DOI: 10.1159/000355947] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IgE is a key mediator in allergic diseases. However, in strong contrast to other antibody isotypes, many details of the composition of the human IgE repertoire are poorly defined. The low levels of human IgE in the circulation and the rarity of IgE-producing B cells are important reasons for this lack of knowledge. In this review, we summarize the current knowledge on these repertoires both in terms of their complexity and activity, i.e. knowledge which despite the difficulties encountered when studying the molecular details of human IgE has been acquired in recent years. We also take a look at likely future developments, for instance through improvements in sequencing technology and methodology that allow the isolation of additional allergen-specific human antibodies mimicking IgE, as this certainly will support our understanding of human IgE in the context of human disease in the years to come.
Collapse
Affiliation(s)
- Elisabeth Gadermaier
- Division of Immunopathology, Department of Pathophysiology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
48
|
Mudnakudu Nagaraju KK, Babina M, Worm M. Opposing effects on immune function and skin barrier regulation by the proteasome inhibitor bortezomib in an allergen-induced eczema model. Exp Dermatol 2013; 22:742-7. [DOI: 10.1111/exd.12261] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Kiran Kumar Mudnakudu Nagaraju
- Department of Dermatology und Allergology; Allergie-Centrum-Charité; CCM; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Magda Babina
- Department of Dermatology und Allergology; Allergie-Centrum-Charité; CCM; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Margitta Worm
- Department of Dermatology und Allergology; Allergie-Centrum-Charité; CCM; Charité - Universitätsmedizin Berlin; Berlin Germany
| |
Collapse
|
49
|
Pittaway CE, Lawson AL, Coles GC, Wilson AD. Systemic and mucosal IgE antibody responses of horses to infection with Anoplocephala perfoliata. Vet Parasitol 2013; 199:32-41. [PMID: 24183646 DOI: 10.1016/j.vetpar.2013.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/24/2013] [Accepted: 10/05/2013] [Indexed: 11/26/2022]
Abstract
Infection of horses with Anoplocephala perfoliata induces a severe inflammatory reaction of the caecal mucosa around the site of parasite attachment adjacent to the ileocecal valve. Lesions show epithelial erosion or ulceration of the mucosa with infiltration by eosinophils, lymphocytes and mast cells leading to oedema, gross thickening and fibrosis of the caecal wall. Despite this evidence of an inflammatory reaction to A. perfoliata within the mucosa of the caecum there is little information about the nature of the local immune response to A. perfoliata. An ELISA which assays serum IgG(T) antibodies to A. perfoliata excretory/secretory antigens has been developed as a diagnostic test. However, the specificity of the ELISA remains sub-optimal and the role of other isotypes in the immune response to A. perfoliata has not been reported. This study measured IgA, IgE and IgG(T) antibody responses to A. perfoliata excretory/secretory antigens in sera of 75 horses presented for slaughter. The prevalence of A. perfoliata infection, as confirmed by the presence of parasites in the terminal ileum, caecum or proximal colon, was 55%. A. perfoliata-specific IgG(T) and IgE antibodies were significantly elevated in infected horses compared to controls; IgA antibodies were also detected but did not differ between infected and control horses. Diagnosis by serum IgG(T) ELISA had a sensitivity of 78% and a specificity of 80%, by comparison the serum IgE ELISA had a sensitivity of just 44% with a specificity of 82% and therefore did not provide an improved diagnostic test. Western blots with sera from infected horses demonstrated IgE-binding to at least 10 separate components of excretory/secretory (E/S) antigens. A similar pattern was also found with IgG(T). Around 30% of horses had high levels of serum IgE which bound fucose-containing carbohydrate antigens on the parasite surface but this was unrelated to the presence of A. perfoliata infection. Immunoperoxidase staining detected numerous IgE-positive cells within lymphoid follicles in the caecal mucosa close to the site of A. perfoliata attachment and quantitative RT-PCR detected high levels of IgE transcription in the caecal mucosa of all horses. Mucosal synthesis of antibodies was confirmed by the demonstration of A. perfoliata-specific IgG(T) and IgE in the supernatant of lamina propria explant cultures that discriminated clearly between infected and uninfected horses. We conclude that there is an active immune response to A. perfoliata within the caecal mucosa involving local production of both IgG(T) and IgE antibody isotypes; but it remains unclear whether this immune response can reduce or eliminate parasite burden.
Collapse
Affiliation(s)
- Charles E Pittaway
- University of Bristol, School of Clinical Veterinary Sciences, Langford House, Bristol BS40 5DU, UK
| | | | | | | |
Collapse
|
50
|
Platts-Mills TAE, Commins SP. Emerging antigens involved in allergic responses. Curr Opin Immunol 2013; 25:769-74. [PMID: 24095162 DOI: 10.1016/j.coi.2013.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/04/2013] [Accepted: 09/06/2013] [Indexed: 10/26/2022]
Abstract
New allergic diseases can 'emerge' because of exposure to a novel antigen, because the immune responsiveness of the subject changes, or because of a change in the behavior of the population. Novel antigens have entered the environment as new pests in the home (e.g., Asian lady beetle or stink bugs), in the diet (e.g., prebiotics or wheat isolates), or because of the spread of a biting arthropod (e.g., ticks). Over the last few years, a significant new disease has been identified, which has changed the paradigm for food allergy. Bites of the tick, Amblyomma americanum, are capable of inducing IgE antibodies to galactose-alpha-1,3-galactose, which is associated with two novel forms of anaphylaxis. In a large area of the southeastern United States, the disease of delayed anaphylaxis to mammalian meat is now common. This disease challenges many previous rules about food allergy and provides a striking model of an emerging allergic disease.
Collapse
Affiliation(s)
- Thomas A E Platts-Mills
- Department of Medicine, Division of Allergy and Immunology, University of Virginia Health System, Charlottesville, VA, USA.
| | | |
Collapse
|