1
|
Zeng J, Zou J, Yi H, He J, Zhao J, Zhu S, Li B, Dudu OE, Zhang L, Gong P. Localization and antigenicity reduction of immunodominant conformational IgE epitopes on αs1-casein. Int J Biol Macromol 2025; 285:138278. [PMID: 39631588 DOI: 10.1016/j.ijbiomac.2024.138278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
αs1-Casein (αs1-CN) is the major allergen in cow milk; however, the understanding of its conformational epitopes remains limited due to the absence of a well-defined three-dimensional structure, which has impeded efforts to effectively reduce its antigenicity. This study employed molecular dynamics simulations (MD), ELISA, cell assays and peptidomes analysis to investigate the critical conformational epitopes of αs1-Casein. MD and immunological analyses identified a dominant conformational epitope encompassing the regions S55-E75 & Y154-T174 & F179-W199, which exhibited strong binding affinity to IgE and triggered the releasing of β-hexosaminidase, histamine and IL-6 in KU812 cells, thereby inducing allergic responses. Notably, the segments Y154-T174 and F179-W199 were particularly impactful. Furthermore, the presence of helical structures within the epitopes enhanced their binding to IgE to a certain extent. Peptidomes analysis further revealed that papain efficiently disrupted the key epitope (Y154-T174) by selectively cleaving the hotspot amino acid residues (Y154 and Y165), thereby significantly reducing the antigenicity of αs1-CN, decreasing IgE and IgG binding to 7.28 % and 10.39 %, respectively. These findings enhance the understanding of αs1-CN's antigenic epitopes and provides a theoretical and technical foundation for the targeted reduction of its antigenicity.
Collapse
Affiliation(s)
- Jianhua Zeng
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; School of Food Engineering, Anhui Science and Technology University, Fengyang 233100, China
| | - Junzhe Zou
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Jinlong Zhao
- School of Food Engineering, Anhui Science and Technology University, Fengyang 233100, China
| | - Shiye Zhu
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Baolei Li
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | | | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Pimin Gong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| |
Collapse
|
2
|
Vogel M, Engeroff P. A Comparison of Natural and Therapeutic Anti-IgE Antibodies. Antibodies (Basel) 2024; 13:58. [PMID: 39051334 PMCID: PMC11270207 DOI: 10.3390/antib13030058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/17/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Immunoglobulin E (IgE) plays a critical role for the immune system, fighting against parasites, toxins, and cancer. However, when it reacts to allergens without proper regulation, it can cause allergic reactions, including anaphylaxis, through a process initiated by effector cells such as basophils and mast cells. These cells display IgE on their surface, bound to the high-affinity IgE receptor FcεRI. A cross-linking antigen then triggers degranulation and the release of inflammatory mediators from the cells. Therapeutic monoclonal anti-IgE antibodies such as omalizumab, disrupt this process and are used to manage IgE-related conditions such as severe allergic asthma and chronic spontaneous urticaria. Interestingly, naturally occurring anti-IgE autoantibodies circulate at surprisingly high levels in healthy humans and mice and may thus be instrumental in regulating IgE activity. Although many open questions remain, recent studies have shed new light on their role as IgE regulators and their mechanism of action. Here, we summarize the latest insights on natural anti-IgE autoantibodies, and we compare their functional features to therapeutic monoclonal anti-IgE autoantibodies.
Collapse
Affiliation(s)
- Monique Vogel
- Department of Rheumatology and Immunology, University Hosptial of Bern, 3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
| | - Paul Engeroff
- Department of Rheumatology and Immunology, University Hosptial of Bern, 3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
3
|
Oylumlu E, Uzel G, Durmus L, Ciraci C. IgE Immune Complexes Mitigate Eosinophilic Immune Responses through NLRC4 Inflammasome. Mediators Inflamm 2023; 2023:3224708. [PMID: 37885469 PMCID: PMC10599938 DOI: 10.1155/2023/3224708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/25/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Immune complexes (ICs) skew immune responses toward either a pro- or anti-inflammatory direction based on the type of stimulation. Immunoglobulin E (IgE) is associated with Th2 immune responses and known to activate innate immune cells. However, roles of antigen (Ag)-specific-IgE ICs in regulating human eosinophil responses remain elusive; therefore, this study builts upon the mechanism of which ovalbumin (Ova)-IgE ICs affects eosinophilic responses utilizing human EoL-1 cell line as a model. Eosinophils are granulocytes functioning through pattern recognition receptors (PRRs) and destructive granule contents in allergic inflammation and parasitic infections. One of the PRRs that eosinophils express is NLRC4, a member of the CARD domain containing nucleotide-binding oligomerization (NOD)-like receptor (NLR) family. Upon recognition of its specific ligand flagellin, NLRC4 inflammasome is formed and leads to the release of interleukin-1β (IL-1β). We exhibited that Ova-IgE ICs induced the NLRC4-inflammasome components, including NLRC4, caspase-1, intracellular IL-1β, and secretion of IL-1β, as well as the granule contents MMP9, TIMP1, and TIMP2 proteins via TLR2 signaling; these responses were suppressed, when NLRC4 inflammasome got actived in the presence of ICs. Furthermore, Ova-IgE ICs induced mRNA expressions of MMP9, TIMP2, and ECP and protein expressions of MMP9 and TIMP2 in EoL-1 through FcɛRII. Interestingly, TLR2 ligand and Ova-IgE ICs costimulation elevated the number of CD63+ cells, a degranulation marker, as compared to the native IgE. Collectively, our findings provide a mechanism for the impacts of Ova-IgE ICs on eosinophilic responses via NLRC4-inflammasome and may help understand eosinophil-associated diseases, including chronic eosinophilic pneumonia, eosinophilic esophagitis, eosinophilic granulomatosis, parasitic infections, allergy, and asthma.
Collapse
Affiliation(s)
- Ece Oylumlu
- Molecular Biology and Genetics Department, Istanbul Technical University, Istanbul 34469, Turkey
| | - Goksu Uzel
- Molecular Biology and Genetics Department, Istanbul Technical University, Istanbul 34469, Turkey
| | - Lubeyne Durmus
- Molecular Biology and Genetics Department, Istanbul Technical University, Istanbul 34469, Turkey
| | - Ceren Ciraci
- Molecular Biology and Genetics Department, Istanbul Technical University, Istanbul 34469, Turkey
| |
Collapse
|
4
|
Falcon RMG, Caoili SEC. Immunologic, genetic, and ecological interplay of factors involved in allergic diseases. FRONTIERS IN ALLERGY 2023; 4:1215616. [PMID: 37601647 PMCID: PMC10435091 DOI: 10.3389/falgy.2023.1215616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
An allergic or type I hypersensitivity reaction involves a misdirected immune overreaction to innocuous environmental and dietary antigens called allergens. The genetic predisposition to allergic disease, referred to as atopy, can be expressed as a variety of manifestations-e.g., allergic rhinitis, allergic conjunctivitis, atopic dermatitis, allergic asthma, anaphylaxis. Globally, allergic diseases are one the most common types of chronic conditions. Several factors have been identified to contribute to the pathogenesis and progression of the disease, leading to distinctively variable clinical symptoms. The factors which can attenuate or exacerbate allergic reactions can range from genetic heterozygosity, the prominence of various comorbid infections, and other factors such as pollution, climate, and interactions with other organisms and organism-derived products, and the surrounding environment. As a result, the effective prevention and control of allergies remains to be one of the most prominent public health problems. Therefore, to contextualize the current knowledge about allergic reactions, this review paper attempts to synthesize different aspects of an allergic response to describe its significance in the global health scheme. Specifically, the review shall characterize the biomolecular mechanisms of the pathophysiology of the disease based on underlying disease theories and current findings on ecologic interactions and describe prevention and control strategies being utilized. An integrated perspective that considers the underlying genetic, immunologic, and ecologic aspects of the disease would enable the development of more effective and targeted diagnostic tools and therapeutic strategies for the management and control of allergic diseases.
Collapse
Affiliation(s)
- Robbi Miguel G. Falcon
- Biomedical Innovations Research for Translational Health Science Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | | |
Collapse
|
5
|
McDonnell JM, Dhaliwal B, Sutton BJ, Gould HJ. IgE, IgE Receptors and Anti-IgE Biologics: Protein Structures and Mechanisms of Action. Annu Rev Immunol 2023; 41:255-275. [PMID: 36737596 DOI: 10.1146/annurev-immunol-061020-053712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The evolution of IgE in mammals added an extra layer of immune protection at body surfaces to provide a rapid and local response against antigens from the environment. The IgE immune response employs potent expulsive and inflammatory forces against local antigen provocation, at the risk of damaging host tissues and causing allergic disease. Two well-known IgE receptors, the high-affinity FcεRI and low-affinity CD23, mediate the activities of IgE. Unlike other known antibody receptors, CD23 also regulates IgE expression, maintaining IgE homeostasis. This mechanism evolved by adapting the function of the complement receptor CD21. Recent insights into the dynamic character of IgE structure, its resultant capacity for allosteric modulation, and the potential for ligand-induced dissociation have revealed previously unappreciated mechanisms for regulation of IgE and IgE complexes. We describe recent research, highlighting structural studies of the IgE network of proteins to analyze the uniquely versatile activities of IgE and anti-IgE biologics.
Collapse
Affiliation(s)
- J M McDonnell
- Randall Centre for Cell and Molecular Biophysics and School of Basic and Medical Biosciences, King's College London, London, UK; , ,
| | | | - B J Sutton
- Randall Centre for Cell and Molecular Biophysics and School of Basic and Medical Biosciences, King's College London, London, UK; , ,
| | - H J Gould
- Randall Centre for Cell and Molecular Biophysics and School of Basic and Medical Biosciences, King's College London, London, UK; , ,
| |
Collapse
|
6
|
Kuo BS, Li CH, Chen JB, Shiung YY, Chu CY, Lee CH, Liu YJ, Kuo JH, Hsu C, Su HW, Li YF, Lai A, Ho YF, Cheng YN, Huang HX, Lung MC, Wu MS, Yang FH, Lin CH, Tseng W, Yang J, Lin CY, Tsai PH, Chang HK, Wang YJ, Chen T, Lynn S, Liao MJ, Wang CY. IgE-neutralizing UB-221 mAb, distinct from omalizumab and ligelizumab, exhibits CD23-mediated IgE downregulation and relieves urticaria symptoms. J Clin Invest 2022; 132:157765. [PMID: 35912861 PMCID: PMC9337824 DOI: 10.1172/jci157765] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/14/2022] [Indexed: 01/08/2023] Open
Abstract
Over the last 2 decades, omalizumab is the only anti-IgE antibody that has been approved for asthma and chronic spontaneous urticaria (CSU). Ligelizumab, a higher-affinity anti-IgE mAb and the only rival viable candidate in late-stage clinical trials, showed anti-CSU efficacy superior to that of omalizumab in phase IIb but not in phase III. This report features the antigenic-functional characteristics of UB-221, an anti-IgE mAb of a newer class that is distinct from omalizumab and ligelizumab. UB-221, in free form, bound abundantly to CD23-occupied IgE and, in oligomeric mAb-IgE complex forms, freely engaged CD23, while ligelizumab reacted limitedly and omalizumab stayed inert toward CD23; these observations are consistent with UB-221 outperforming ligelizumab and omalizumab in CD23-mediated downregulation of IgE production. UB-221 bound IgE with a strong affinity to prevent FcԑRI-mediated basophil activation and degranulation, exhibiting superior IgE-neutralizing activity to that of omalizumab. UB-221 and ligelizumab bound cellular IgE and effectively neutralized IgE in sera of patients with atopic dermatitis with equal strength, while omalizumab lagged behind. A single UB-221 dose administered to cynomolgus macaques and human IgE (ε, κ)-knockin mice could induce rapid, pronounced serum-IgE reduction. A single UB-221 dose administered to patients with CSU in a first-in-human trial exhibited durable disease symptom relief in parallel with a rapid reduction in serum free-IgE level.
Collapse
Affiliation(s)
- Be-Sheng Kuo
- United BioPharma, Inc., Hsinchu, Taiwan.,UBI Asia, Hsinchu, Taiwan.,United Biomedical, Inc., Hauppauge, New York, USA
| | | | | | | | - Chia-Yu Chu
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | - Cindy Hsu
- United BioPharma, Inc., Hsinchu, Taiwan
| | | | | | - Annie Lai
- United BioPharma, Inc., Hsinchu, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Chang Yi Wang
- United BioPharma, Inc., Hsinchu, Taiwan.,UBI Asia, Hsinchu, Taiwan.,United Biomedical, Inc., Hauppauge, New York, USA
| |
Collapse
|
7
|
Votto M, Achilli G, De Filippo M, Licari A, Marseglia A, Moiraghi A, Di Sabatino A, Marseglia GL. Pediatric chronic spontaneous urticaria: a brief clinician's guide. Expert Rev Clin Immunol 2022; 18:889-899. [PMID: 35833360 DOI: 10.1080/1744666x.2022.2101999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic urticaria (CU) appears with daily or intermittent/recurrent wheals with/without angioedema for more than six weeks. When no specific eliciting factors are found, chronic urticaria is defined as spontaneous (CSU). Up to 50% of patients with CSU do not respond to therapy, leading to a prolonged disease course and the need for expensive therapies, impacting the quality of life (QoL) and healthcare resources. AREAS COVERED : Diagnosis of CSU is made when other potential causes of chronic urticaria are excluded. CSU therapy aims to achieve complete control of symptoms and normalization of QoL. Current treatment options for urticaria aim to target mast cell mediators such as histamine, or activators, such as autoantibodies. Guidelines recommend starting with second generation antihistamines (sgAHs) and adding omalizumab therapy if symptoms are not controlled. This review aims to provide a practical guide for CSU in the pediatric population. EXPERT OPINION : Treatment options for pediatric CSU are primarily based on adult data that have been extrapolated for children. Current guidelines should be re-evaluated based on pediatric data, new biological treatments, and the COVID-19 pandemic. Future research is needed to investigate strategies to personalize current treatments and identify potential predictive biomarkers.
Collapse
Affiliation(s)
- Martina Votto
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy
| | - Giovanna Achilli
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Maria De Filippo
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy.,Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Amelia Licari
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy.,Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessia Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy
| | - Alice Moiraghi
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Italy.,Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
8
|
Agache I, Akdis CA, Akdis M, Brockow K, Chivato T, Giacco S, Eiwegger T, Eyerich K, Giménez‐Arnau A, Gutermuth J, Guttman‐Yassky E, Maurer M, Ogg G, Ong PY, O’Mahony L, Schwarze J, Warner A, Werfel T, Palomares O, Jutel M. EAACI Biologicals Guidelines-Omalizumab for the treatment of chronic spontaneous urticaria in adults and in the paediatric population 12-17 years old. Allergy 2022; 77:17-38. [PMID: 34324716 DOI: 10.1111/all.15030] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/21/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022]
Abstract
Chronic spontaneous urticaria (CSU) imposes a significant burden on patients, families and healthcare systems. Management is difficult, due to disease heterogeneity and insufficient efficacy of classical drugs such as H1 R-antihistamines. Better understanding of the mechanisms has enabled a stratified approach to the management of CSU, supporting the use of targeted treatment with omalizumab. However, many practical issues including selection of responders, the definition of response, strategies to enhance the responder rate, the duration of treatment and its regimen (in the clinic or home-based) and its cost-effectiveness still require further clarification. The EAACI Guidelines on the use of omalizumab in CSU follow the GRADE approach in formulating recommendations for each outcome. In addition, future therapeutic approaches and perspectives as well as research priorities are discussed.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐CARE Davos Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Knut Brockow
- Department of Dermatology and Allergology Biederstein School of Medicine Technical University of Munich Munich Germany
| | - Tomas Chivato
- School of Medicine University CEU San Pablo Madrid Spain
| | - Stefano Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - Thomas Eiwegger
- Translational Medicine Program, Research InstituteHospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Karl Landsteiner University of Health Sciences Krems Austria
- Department of Paediatrics University Hospital St. Pölten Pölten Austria
| | - Kilian Eyerich
- Department of Dermatology and Allergy Biederstein Technical University of Munich Munich Germany
| | - Ana Giménez‐Arnau
- Department of Dermatology Hospital del Mar‐ Institut Mar d'Investigacions Mèdiques Universitat Autònoma de Barcelona Barcelona Spain
| | - Jan Gutermuth
- Department of Dermatology Universitair Ziekenhuis BrusselVrije Universiteit Brussel (VUB Brussels Belgium
| | - Emma Guttman‐Yassky
- Department of DermatologyIcahn School of Medicine at Mount Sinai New York New York USA
| | - Marcus Maurer
- Dermatological Allergology Allergie‐Centrum‐Charité Department of Dermatology and Allergy Charité ‐ Universitätsmedizin Berlin Berlin Germany
| | - Graham Ogg
- MRC Human Immunology Unit MRC Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre Radcliffe Department of Medicine University of Oxford Oxford UK
| | - Peck Y. Ong
- Division of Clinical Immunology & Allergy Children’s Hospital Los Angeles Keck School of Medicine University of Southern California Los Angeles California USA
| | - Liam O’Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland University College Cork Cork Ireland
| | - Jürgen Schwarze
- Centre for Inflammation Research, Child Life and Health The University of Edinburgh Edinburgh UK
| | | | - Thomas Werfel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology Chemistry School Complutense University of Madrid Madrid Spain
| | - Marek Jutel
- Department of Clinical Immunology Wrocław Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wroclaw Poland
| |
Collapse
|
9
|
Rodak A, Stadlmayr G, Stadlbauer K, Lichtscheidl D, Bobbili MR, Rüker F, Wozniak-Knopp G. Bispecific T-Cell Engagers Targeting Membrane-Bound IgE. Biomedicines 2021; 9:1568. [PMID: 34829798 PMCID: PMC8615095 DOI: 10.3390/biomedicines9111568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022] Open
Abstract
The increased incidence of allergies and asthma has sparked interest in IgE, the central player in the allergic response. Interaction with its high-affinity receptor FcεRI leads to sensitization and allergen presentation, extracellular membrane-proximal domain in membrane IgE can act as an antigen receptor on B cells, and the interaction with low-affinity IgE receptor CD23 additionally influences its homeostatic range. Therapeutic anti-IgE antibodies act by the inhibition of IgE functions by interfering with its receptor binding or by the obliteration of IgE-B cells, causing a reduction of serum IgE levels. Fusion proteins of antibody fragments that can act as bispecific T-cell engagers have proven very potent in eliciting cytotoxic T-lymphocyte-mediated killing. We have tested five anti-IgE Fc antibodies, recognizing different epitopes on the membrane-expressed IgE, for the ability to elicit specific T-cell activation when expressed as single-chain Fv fragments fused with anti-CD3ε single-chain antibody. All candidates could specifically stain the cell line, expressing the membrane-bound IgE-Fc and bind to CD3-positive Jurkat cells, and the specific activation of engineered CD3-overexpressing Jurkat cells and non-stimulated CD8-positive cells was demonstrated for 8D6- and ligelizumab-based bispecific antibodies. Thus, such anti-IgE antibodies have the potential to be developed into agents that reduce the serum IgE concentration by lowering the numbers of IgE-secreting cells.
Collapse
Affiliation(s)
- Aleksandra Rodak
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| | - Gerhard Stadlmayr
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| | - Katharina Stadlbauer
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| | - Dominic Lichtscheidl
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| | - Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
- Ludwig Boltzmann Institute for Experimental, Clinical Traumatology in the AUVA Research Center, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Florian Rüker
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| | - Gordana Wozniak-Knopp
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria; (A.R.); (G.S.); (K.S.); (D.L.); (M.R.B.); (F.R.)
| |
Collapse
|
10
|
Engeroff P, Vogel M. The role of CD23 in the regulation of allergic responses. Allergy 2021; 76:1981-1989. [PMID: 33378583 PMCID: PMC8359454 DOI: 10.1111/all.14724] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
IgE, the key molecule in atopy has been shown to bind two receptors, FcεRI, the high‐affinity receptor, and FcεRII (CD23), binding IgE with lower affinity. Whereas cross‐linking of IgE on FcεRI expressed by mast cells and basophils triggers the allergic reaction, binding of IgE to CD23 on B cells plays an important role in both IgE regulation and presentation. Furthermore, IgE‐immune complexes (IgE‐ICs) bound by B cells enhance antibody and T cell responses in mice and humans. However, the mechanisms that regulate the targeting of the two receptors and the respective function of the two pathways in inflammation or homeostasis are still a matter of debate. Here, we focus on CD23 and discuss several mechanisms related to IgE binding, as well as the impact of the IgE/antigen‐binding on different immune cells expressing CD23. One recent paper has shown that free IgE preferentially binds to FcεRI whereas IgE‐ICs are preferentially captured by CD23. Binding of IgE‐ICs to CD23 on B cells can, on one hand, regulate serum IgE and prevent effector cell activation and on the other hand facilitate antigen presentation by delivering the antigen to dendritic cells. These data argue for a multifunctional role of CD23 for modulating IgE serum levels and immune responses.
Collapse
Affiliation(s)
- Paul Engeroff
- INSERM UMR_S 959 Immunology‐Immunopathology‐Immunotherapy (i3) Sorbonne Université Paris France
| | - Monique Vogel
- Center for Clinical Research Region Västmanland/Uppsala University, Västmanland hospital Västerås Sweden
- Department of BioMedical Research University of Bern Bern Switzerland
| |
Collapse
|
11
|
Wedi B, Traidl S. Anti-IgE for the Treatment of Chronic Urticaria. Immunotargets Ther 2021; 10:27-45. [PMID: 33628747 PMCID: PMC7898214 DOI: 10.2147/itt.s261416] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/22/2021] [Indexed: 12/27/2022] Open
Abstract
Urticaria and angioedema are very common. Management of chronic urticaria subtypes, which usually persist for many years, is challenging. Recent years have demonstrated that targeting IgE with antibodies provides a safe and efficient treatment approach. Whilst several anti-IgE antibodies have been developed, omalizumab is currently the only one approved for use. International and national guidelines recommend its use after failure of antihistamines at standard and increased dose. Whilst not yet approved, many new anti-IgE approaches are currently being investigated in pre-clinical studies or clinical trials. This non-systematic focused review summarizes current knowledge of omalizumab and other anti-IgE biologics in chronic urticaria using data extracted from PubMed, Google Scholar and clinical trial databases, clinicaltrials.gov and clinicaltrials.eu. For adults, there is good evidence from randomized clinical trials and real-world data that symptomatic treatment with omalizumab is efficacious and safe in chronic spontaneous urticaria (CSU), whereas evidence in chronic inducible urticaria (CINDU) and special populations is limited. Easy-to-use tools to identify non-responders and predict the required duration of treatment have not been established yet. Phase 2 b results of ligelizumab have not only demonstrated efficacy and safety but also superiority to omalizumab. Indeed, there is preliminary evidence that omalizumab non- or partial responders benefit from ligelizumab. Whereas further development of quilizumab was discontinued, other approaches, eg UB-221 or DARPins are under investigation. Anti-IgE treatment with omalizumab represents a landmark in the treatment of chronic urticaria, with and without angioedema, and there is light on the horizon suggesting success may come with various next-generation anti-IgE approaches.
Collapse
Affiliation(s)
- Bettina Wedi
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| | - Stephan Traidl
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Skopnik CM, Al-Qaisi K, Calvert RA, Enghard P, Radbruch A, Sutton BJ, Kubagawa H. Identification of Amino Acid Residues in Human IgM Fc Receptor (FcµR) Critical for IgM Binding. Front Immunol 2021; 11:618327. [PMID: 33584711 PMCID: PMC7873564 DOI: 10.3389/fimmu.2020.618327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
Both non-immune “natural” and antigen-induced “immune” IgM are important for protection against infections and for regulation of immune responses to self-antigens. The roles of its Fc receptor (FcµR) in these IgM effector functions have begun to be explored. In the present study, by taking advantage of the difference in IgM-ligand binding of FcµRs of human (constitutive binding) and mouse (transient binding), we replaced non-conserved amino acid residues of human FcµR with mouse equivalents before establishment of cell lines stably expressing mutant or wild-type (WT) receptors. The resultant eight-different mutant FcµR-bearing cells were compared with WT receptor-bearing cells for cell-surface expression and IgM-binding by flow cytometric assessments using receptor-specific mAbs and IgM paraproteins as ligands. Three sites Asn66, Lys79-Arg83, and Asn109, which are likely in the CDR2, DE loop and CDR3 of the human FcµR Ig-like domain, respectively, were responsible for constitutive IgM binding. Intriguingly, substitution of Glu41 and Met42 in the presumed CDR1 with the corresponding mouse residues Gln and Leu, either single or more prominently in combination, enhanced both the receptor expression and IgM binding. A four-aa stretch of Lys24-Gly27 in the predicted A ß-strand of human FcµR appeared to be essential for maintenance of its proper receptor conformation on plasma membranes because of reduction of both receptor expression and IgM-binding potential when these were mutated. Results from a computational structural modeling analysis were consistent with these mutational data and identified a possible mode of binding of FcµR with IgM involving the loops including Asn66, Arg83 and Asn109 of FcµR interacting principally with the Cµ4 domain including Gln510 and to a lesser extent Cµ3 domain including Glu398, of human IgM. To our knowledge, this is the first experimental report describing the identification of amino acid residues of human FcµR critical for binding to IgM Fc.
Collapse
Affiliation(s)
| | - Khlowd Al-Qaisi
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Rosaleen A Calvert
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité-Universitätmedizin, Berlin, Germany
| | - Andreas Radbruch
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Brian J Sutton
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Hiromi Kubagawa
- Humoral Immune Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
13
|
El Jahrani N, Cretin G, de Brevern AG. CALR-ETdb, the database of calreticulin variants diversity in essential thrombocythemia. Platelets 2021; 33:157-167. [PMID: 33444113 DOI: 10.1080/09537104.2020.1869712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Essential thrombocythemia (ET) is a blood cancer defined by a strong increase of platelet numbers. A quarter of patients suffering from ET show mutations in the last exon of calreticulin (CALR) gene. Two variants named type 1 and type 2 represent 85% of these patients. However, a large number of other variants have been determined. In this study, we have compiled variants taken from COSMIC database and literature leading to 155 different variants. This large number of variants allowed redefining 5 new classes extending the classification of type 1-like and type 2-like to a finer description. These analyses showed that last class, named E, corresponding to more than 10% of CALR variants seemed not attached to ET. Structural properties analyzed showed that CALR variants associated to ET have common features. All the compiled and refined information had been included into a freely dedicated database CALR-ETdb (https://www.dsimb.inserm.fr/CALR-ET).
Collapse
Affiliation(s)
- Nora El Jahrani
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Gabriel Cretin
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| | - Alexandre G de Brevern
- Université de Paris, UMR_S 1134, Université De La Réunion, Université Des Antilles, Paris, France.,INSERM, U 1134, DSIMB, Paris, France.,Institut National De La Transfusion Sanguine (INTS), Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
14
|
Guntern P, Eggel A. Past, present, and future of anti-IgE biologics. Allergy 2020; 75:2491-2502. [PMID: 32249957 DOI: 10.1111/all.14308] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/09/2020] [Accepted: 03/28/2020] [Indexed: 12/31/2022]
Abstract
About 20 years after the identification of immunoglobulin E (IgE) and its key role in allergic hypersensitivity reactions against normally harmless substances, scientists have started inventing strategies to block its pathophysiological activity in 1986. The initial concept of specific IgE targeting through the use of anti-IgE antibodies has gained a lot of momentum and within a few years independent research groups have reported successful generation of first murine monoclonal anti-IgE antibodies. Subsequent generation of optimized chimeric and humanized versions of these antibodies has paved the way for the development of therapeutic anti-IgE biologicals as we know them today. With omalizumab, there is currently still only one therapeutic anti-IgE antibody approved for the treatment of allergic conditions. Since its application is limited to the treatment of moderate-to-severe persistent asthma and chronic spontaneous urticaria, major efforts have been undertaken to develop alternative anti-IgE biologicals that could potentially be used in a broader spectrum of allergic diseases. Several new drug candidates have been generated and are currently assessed in pre-clinical studies or clinical trials. In this review, we highlight the molecular properties of past and present anti-IgE biologicals and suggest concepts that might improve treatment efficacy of future drug candidates.
Collapse
Affiliation(s)
- Pascal Guntern
- Graduate School of Cellular and Biomedical Sciences University of Bern Bern Switzerland
- Department of BioMedical Research University of Bern Bern Switzerland
- Department of Rheumatology, Immunology and Allergology University Hospital Bern Bern Switzerland
| | - Alexander Eggel
- Department of BioMedical Research University of Bern Bern Switzerland
- Department of Rheumatology, Immunology and Allergology University Hospital Bern Bern Switzerland
| |
Collapse
|
15
|
Safar HA, El-Hashim AZ, Amoudy H, Mustafa AS. Mycobacterium tuberculosis-Specific Antigen Rv3619c Effectively Alleviates Allergic Asthma in Mice. Front Pharmacol 2020; 11:532199. [PMID: 33101014 PMCID: PMC7546857 DOI: 10.3389/fphar.2020.532199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/31/2020] [Indexed: 01/20/2023] Open
Abstract
Despite significant advances, asthma remains a cause of premature death, and current treatments are suboptimal. Antigen-specific Th2 cells and their cytokines are primary mediators of the pathophysiological changes seen in asthma. Studies in animal models have shown that mycobacteria can suppress the asthma phenotype by alteration of the Th1/Th2 cytokines ratio. In this study, utilizing a Th1 delivery system to modulate the allergic airway inflammation in a Th2-driven model of asthma, we evaluated the efficacy of immunization with Mycobacterium tuberculosis-specific antigen Rv3619c, either alone or in combination with low dose dexamethasone. The rv3619c gene was cloned in an expression plasmid pGES-TH-1, expressed in Escherichia coli, and the recombinant protein Rv3619c was purified to homogeneity using affinity chromatography. Mice were immunized with the recombinant protein emulsified in Freund's Incomplete Adjuvant (IFA) alone and in combination with low dose dexamethasone, and then challenged with ovalbumin (OVA). Airway inflammation was assessed by quantifying airway cytology, histological changes and Th2 cytokine (IL-5) secretion from splenocytes. OVA-specific IgE, IgG and IgG1 from sera was assessed, as well as pERK1/2 expression in the lung tissue. Immunization with recombinant Rv3619c alone inhibited the OVA-induced increase in total cell counts, eosinophil airway cell infiltration in BAL fluid, perivascular and peribronchial inflammation and fibrosis, and goblet cell hyper/metaplasia. In addition, Rv3619c/IFA inhibited the OVA-induced IL-5 in spleen cells, OVA-specific IgE, IgG, and IgG1 levels in sera, and pERK1/2 expression in lung tissue. Immunization with Rv3619c/IFA in combination with low dose dexamethasone resulted in an enhanced effect on some but not all the asthma features. Taken together, this study demonstrates that immunization with Rv3619c/IFA, alone or in combination with dexamethasone, may be an effective treatment strategy for the prevention of asthma.
Collapse
Affiliation(s)
- Hussain A Safar
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Ahmed Z El-Hashim
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait City, Kuwait
| | - Hanady Amoudy
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
16
|
Effects of omalizumab on basophils: Potential biomarkers in asthma and chronic spontaneous urticaria. Cell Immunol 2020; 358:104215. [PMID: 33137647 DOI: 10.1016/j.cellimm.2020.104215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/28/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022]
Abstract
Omalizumab is an anti-IgE humanized monoclonal antibody approved for the treatment of severe asthma and chronic spontaneous urticaria. Omalizumab binds free serum IgE and antagonizes its interaction with FcεRI, which is considered the main pharmacodynamic mechanism responsible for the clinical response to the treatment. The reduction of IgE serum concentration down-regulates the cellular expression of FcεRI on basophils. However, the biological events occurring on basophils during the therapy with omalizumab are multiple and complex. Here we review the current evidence regarding the specific biological effects of omalizumab on basophils in patients with asthma and chronic spontaneous urticaria. In addition to the modulation of IgE receptors, omalizumab may affect basophils homeostasis, intra-cellular signaling, cellular responsiveness/activation and cytokine release. These effects may be partially responsible for the clinical success of omalizumab and potentially provide useful biological markers for future assessment of the clinical response to the treatment. However, further investigation is required to better elucidate the role of basophils during the treatment with omalizumab.
Collapse
|
17
|
Lefranc MP, Lefranc G. Immunoglobulins or Antibodies: IMGT ® Bridging Genes, Structures and Functions. Biomedicines 2020; 8:E319. [PMID: 32878258 PMCID: PMC7555362 DOI: 10.3390/biomedicines8090319] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
IMGT®, the international ImMunoGeneTics® information system founded in 1989 by Marie-Paule Lefranc (Université de Montpellier and CNRS), marked the advent of immunoinformatics, a new science at the interface between immunogenetics and bioinformatics. For the first time, the immunoglobulin (IG) or antibody and T cell receptor (TR) genes were officially recognized as 'genes' as well as were conventional genes. This major breakthrough has allowed the entry, in genomic databases, of the IG and TR variable (V), diversity (D) and joining (J) genes and alleles of Homo sapiens and of other jawed vertebrate species, based on the CLASSIFICATION axiom. The second major breakthrough has been the IMGT unique numbering and the IMGT Collier de Perles for the V and constant (C) domains of the IG and TR and other proteins of the IG superfamily (IgSF), based on the NUMEROTATION axiom. IMGT-ONTOLOGY axioms and concepts bridge genes, sequences, structures and functions, between biological and computational spheres in the IMGT® system (Web resources, databases and tools). They provide the IMGT Scientific chart rules to identify, to describe and to analyse the IG complex molecular data, the huge diversity of repertoires, the genetic (alleles, allotypes, CNV) polymorphisms, the IG dual function (paratope/epitope, effector properties), the antibody humanization and engineering.
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- IMGT, The International ImMunoGeneTics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine IGH, Université de Montpellier UM, Centre National de la Recherche Scientifique CNRS, UMR 9002 CNRS-UM, 141 Rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| | - Gérard Lefranc
- IMGT, The International ImMunoGeneTics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine IGH, Université de Montpellier UM, Centre National de la Recherche Scientifique CNRS, UMR 9002 CNRS-UM, 141 Rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| |
Collapse
|
18
|
Jensen RK, Jabs F, Miehe M, Mølgaard B, Pfützner W, Möbs C, Spillner E, Andersen GR. Structure of intact IgE and the mechanism of ligelizumab revealed by electron microscopy. Allergy 2020; 75:1956-1965. [PMID: 32037590 DOI: 10.1111/all.14222] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND IgE is the central antibody isotype in TH2-biased immunity and allergic diseases. The structure of intact IgE and the impact of IgE-targeting molecules on IgE however remain elusive. In order to obtain insights into IgE biology and the clinical impact, we aimed for structure determination of IgE and the complex of IgE with the anti-IgE antibody ligelizumab. METHODS Structures of two distinct intact IgE with specificity for cross-reactive carbohydrate determinants and Der p 2 as well as complexes of ligelizumab-Fab with IgE and IgE Fc were assessed by negative stain electron microscopy and solution scattering. Inhibition of IgE binding and displacement of receptor-bound IgE were assessed using cellular assays, basophil activation testing and ELIFAB assays. RESULTS Our data reveal that the investigated IgE molecules share an overall rigid conformation. In contrast to the IgE Fc fragment, the IgE Fc in intact IgE is significantly less asymmetrically bent. The proximal and the distal Fabs are rigidly tethered to the Fc. Binding of ligelizumab to IgE in a 2:1 stoichiometry induces an extended and twofold symmetrical conformation of IgE, which retains a rigid Fab-Fc architecture. Analyses of effector cell activation revealed that ligelizumab inhibits IgE binding without displacing receptor-bound IgE. Together with an interference of CD23 binding, the data underline a functional activity similar to omalizumab. CONCLUSIONS Our data reveal the first structures of intact IgE suggesting that the IgE Fab is fixed relative to the Fc. Furthermore, we provide a structural rationale for the inhibitory mechanism of ligelizumab.
Collapse
Affiliation(s)
- Rasmus K. Jensen
- Department of Molecular Biology and Genetics Aarhus University Aarhus Denmark
| | - Frederic Jabs
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Michaela Miehe
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Brian Mølgaard
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Wolfgang Pfützner
- Clinical and Experimental Allergology, Department of Dermatology and Allergology Philipps-Universität Marburg Marburg Germany
| | - Christian Möbs
- Clinical and Experimental Allergology, Department of Dermatology and Allergology Philipps-Universität Marburg Marburg Germany
| | - Edzard Spillner
- Immunological Biotechnology Department of Engineering Aarhus University Aarhus Denmark
| | - Gregers R. Andersen
- Department of Molecular Biology and Genetics Aarhus University Aarhus Denmark
| |
Collapse
|
19
|
Benjamin SV, Creeke PI, Henry AJ, McDonnell JM. NMR backbone assignment of the Cε4 domain of immunoglobulin E. BIOMOLECULAR NMR ASSIGNMENTS 2020; 14:151-155. [PMID: 32108310 PMCID: PMC7069932 DOI: 10.1007/s12104-020-09936-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/21/2020] [Indexed: 06/10/2023]
Abstract
Immunoglobulin E (IgE) plays a central role in allergic reactions. IgE is a dynamic molecule that is capable of undergoing large conformational changes. X-ray crystal structures of the Fc region of IgE in complex with various ligands have shown that IgE-Fc can exist in extended and various bent conformations. IgE-Fc consists of three domains: Cε2, Cε3 and Cε4. While the complete NMR backbone assignments of the Cε2 and Cε3 domains have been reported previously, the Cε4 domain has not been assigned. Here, we report the complete backbone assignment of the Cε4 homodimer. Cε4 can be used as a model system to study dynamics and allostery in IgE, as both molecules exist as homodimers and exhibit similar binding properties to a number of ligands.
Collapse
Affiliation(s)
- Stefi V Benjamin
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Paul I Creeke
- UCB Pharma, 216 Bath Road, Slough, Berkshire, SL1 3WE, UK
| | | | - James M McDonnell
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
20
|
Mitropoulou AN, Ceska T, Heads JT, Beavil AJ, Henry AJ, McDonnell JM, Sutton BJ, Davies AM. Engineering the Fab fragment of the anti-IgE omalizumab to prevent Fab crystallization and permit IgE-Fc complex crystallization. Acta Crystallogr F Struct Biol Commun 2020; 76:116-129. [PMID: 32133997 PMCID: PMC7057348 DOI: 10.1107/s2053230x20001466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/03/2020] [Indexed: 12/01/2022] Open
Abstract
Immunoglobulin E (IgE) plays a central role in the allergic response, in which cross-linking of allergen by FcεRI-bound IgE triggers mast cell and basophil degranulation and the release of inflammatory mediators. The high-affinity interaction between IgE and FcεRI is a long-standing target for therapeutic intervention in allergic disease. Omalizumab is a clinically approved anti-IgE monoclonal antibody that binds to free IgE, also with high affinity, preventing its interaction with FcεRI. All attempts to crystallize the pre-formed complex between the omalizumab Fab and the Fc region of IgE (IgE-Fc), to understand the structural basis for its mechanism of action, surprisingly failed. Instead, the Fab alone selectively crystallized in different crystal forms, but their structures revealed intermolecular Fab/Fab interactions that were clearly strong enough to disrupt the Fab/IgE-Fc complexes. Some of these interactions were common to other Fab crystal structures. Mutations were therefore designed to disrupt two recurring packing interactions observed in the omalizumab Fab crystal structures without interfering with the ability of the omalizumab Fab to recognize IgE-Fc; this led to the successful crystallization and subsequent structure determination of the Fab/IgE-Fc complex. The mutagenesis strategy adopted to achieve this result is applicable to other intractable Fab/antigen complexes or systems in which Fabs are used as crystallization chaperones.
Collapse
Affiliation(s)
- Alkistis N. Mitropoulou
- Randall Centre for Cell and Molecular Biophysics, King’s College London, New Hunt’s House, London SE1 1UL, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Tom Ceska
- UCB Celltech, 208 Bath Road, Slough SL1 3WE, UK
| | | | - Andrew J. Beavil
- Randall Centre for Cell and Molecular Biophysics, King’s College London, New Hunt’s House, London SE1 1UL, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics, King’s College London, New Hunt’s House, London SE1 1UL, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Brian J. Sutton
- Randall Centre for Cell and Molecular Biophysics, King’s College London, New Hunt’s House, London SE1 1UL, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Anna M. Davies
- Randall Centre for Cell and Molecular Biophysics, King’s College London, New Hunt’s House, London SE1 1UL, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|
21
|
Feng Y, Wang Y, Zhang S, Haneef K, Liu W. Structural and immunogenomic insights into B-cell receptor activation. J Genet Genomics 2020; 47:27-35. [PMID: 32111437 DOI: 10.1016/j.jgg.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023]
Abstract
B cells express B-cell receptors (BCRs) which recognize antigen to trigger signaling cascades for B-cell activation and subsequent antibody production. BCR activation has a crucial influence on B-cell fate. How BCR is activated upon encountering antigen remains to be solved, although tremendous progresses have been achieved in the past few years. Here, we summarize the models that have been proposed to explain BCR activation, including the cross-linking model, the conformation-induced oligomerization model, the dissociation activation model, and the conformational change model. Especially, we elucidate the partially resolved structures of antibodies and/or BCRs by far and discusse how these current structural and further immunogenomic messages and more importantly the future studies may shed light on the explanation of BCR activation and the relevant diseases in the case of dysregulation.
Collapse
Affiliation(s)
- Yangyang Feng
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Shaocun Zhang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Kabeer Haneef
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
22
|
Sex difference in IgE sensitization associated with alcohol consumption in the general population. Sci Rep 2019; 9:12131. [PMID: 31431645 PMCID: PMC6702201 DOI: 10.1038/s41598-019-48305-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/30/2019] [Indexed: 01/09/2023] Open
Abstract
The association of alcohol consumption and immunoglobulin E (IgE) sensitization is debated. Few population-based studies have investigated whether such associations differ by sex. We explored the association of alcohol consumption with IgE sensitization in the general population, stratified by sex. We analyzed data for 1,723 adults from the 2010 Korean National Health and Nutrition Examination Survey. We divided subjects into three groups according to their self-reported alcohol consumption or serum level of gamma-glutamyltransferase (GGT), an objective marker of alcohol consumption. After adjustments, the odds ratios (ORs) of male high-risk drinkers were 2.09 (95% confidence interval [CI], 1.34–3.28) for total IgE and 1.71 (95% CI, 1.03–2.83) for Dermatophagoides farinae (DF)-specific IgE compared with male low-risk drinkers. In females, the dog-specific IgE level was associated with high-risk drinking (OR, 11.74; 95% CI, 2.04–67.24). The ORs of males in the high-serum-GGT group were 2.73 (95% CI, 1.72–4.33) for total IgE and 2.17 (95% CI, 1.35–3.47) for DF-specific IgE compared with those in the low-serum-GGT group. This study suggests a possible link between alcohol consumption and IgE sensitization, moreover, the risk of IgE sensitization was significantly higher in male high-risk drinkers. Therefore, clinicians should consider the risk of IgE sensitization possibly afflicting male high-risk drinkers.
Collapse
|
23
|
Sutton BJ, Davies AM, Bax HJ, Karagiannis SN. IgE Antibodies: From Structure to Function and Clinical Translation. Antibodies (Basel) 2019; 8:E19. [PMID: 31544825 PMCID: PMC6640697 DOI: 10.3390/antib8010019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 12/15/2022] Open
Abstract
Immunoglobulin E (IgE) antibodies are well known for their role in mediating allergic reactions, and their powerful effector functions activated through binding to Fc receptors FcεRI and FcεRII/CD23. Structural studies of IgE-Fc alone, and when bound to these receptors, surprisingly revealed not only an acutely bent Fc conformation, but also subtle allosteric communication between the two distant receptor-binding sites. The ability of IgE-Fc to undergo more extreme conformational changes emerged from structures of complexes with anti-IgE antibodies, including omalizumab, in clinical use for allergic disease; flexibility is clearly critical for IgE function, but may also be exploited by allosteric interference to inhibit IgE activity for therapeutic benefit. In contrast, the power of IgE may be harnessed to target cancer. Efforts to improve the effector functions of therapeutic antibodies for cancer have almost exclusively focussed on IgG1 and IgG4 subclasses, but IgE offers an extremely high affinity for FcεRI receptors on immune effector cells known to infiltrate solid tumours. Furthermore, while tumour-resident inhibitory Fc receptors can modulate the effector functions of IgG antibodies, no inhibitory IgE Fc receptors are known to exist. The development of tumour antigen-specific IgE antibodies may therefore provide an improved immune functional profile and enhanced anti-cancer efficacy. We describe proof-of-concept studies of IgE immunotherapies against solid tumours, including a range of in vitro and in vivo evaluations of efficacy and mechanisms of action, as well as ex vivo and in vivo safety studies. The first anti-cancer IgE antibody, MOv18, the clinical translation of which we discuss herein, has now reached clinical testing, offering great potential to direct this novel therapeutic modality against many other tumour-specific antigens. This review highlights how our understanding of IgE structure and function underpins these exciting clinical developments.
Collapse
Affiliation(s)
- Brian J Sutton
- King's College London, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK.
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.
| | - Anna M Davies
- King's College London, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK.
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.
| | - Heather J Bax
- King's College London, St John's Institute of Dermatology, London SE1 9RT, UK.
| | | |
Collapse
|