1
|
Du K, Zhao Y, Zhang X, Li C, Hao Y, Du X, Yang Y, Qin X, Hu Y, Li Y, Wang Y, Chen Y, Li Y, Wang W, Wang X, Ying S, Zhang L. Staphylococcus aureus lysate induces an IgE response via memory B cells in nasal polyps. J Allergy Clin Immunol 2024; 153:718-731.e11. [PMID: 38056634 DOI: 10.1016/j.jaci.2023.10.033] [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: 09/18/2022] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Locally increased IgE levels plays a pathologic role in chronic rhinosinusitis with nasal polyps (CRSwNP). OBJECTIVE This study aimed to investigate whether Staphylococcus aureus could induce aberrant IgE synthesis in CRSwNP and the potential mechanisms involved. METHODS Total IgE, IL-4, IL-5, and IL-13 concentrations in the supernatants of the cultures stimulated with S aureus lysate were assessed by ELISA. S aureus-induced cellular responses were investigated by single-cell RNA sequencing. Flow cytometry and quantitative reverse transcription PCR were used to analyze B-cell subsets and stimulated cell ε-germline transcript expression, respectively. IgE-positive B-cell and germinal center localization were assessed by immunohistochemistry and immunofluorescence. RESULTS S aureus lysate induced IgE production in the supernatants of nasal polyp (NP) tissues but not in those of healthy nasal mucosa. Moreover, IgE levels increased from days 2 to 4 after stimulation, paralleling the enhanced ε-germline transcript, IL-5, and IL-13 expression. Single-cell RNA sequencing revealed that there were increased IL-5 and IL-13 in group 2 innate lymphoid cells and identified a clonal overlap between unstimulated memory B cells and S aureus-stimulated plasma cells. The enriched IgE within NPs was mainly produced by IgE-negative memory B cells. Cellular evidence indicated that the IgE memory response to S aureus might also exist in the peripheral blood of CRSwNP patients. The S aureus-induced IgE memory response was associated with elevated IgE levels in NPs, asthma, and postoperative CRSwNP recurrence. CONCLUSIONS S aureus induced an IgE response via IgE-negative memory B cells in CRSwNP patients, possibly contributing to CRSwNP development.
Collapse
Affiliation(s)
- Kun Du
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Department of Otolaryngology-Head and Neck Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yan Zhao
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Xin Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chenduo Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yun Hao
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaonan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yiran Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaofeng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Hu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Li
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Yue Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Li
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiangdong Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.
| | - Sun Ying
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China; Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Luo Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China; Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, Orengo JM. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy. Sci Transl Med 2023; 15:eadf9561. [PMID: 38091405 DOI: 10.1126/scitranslmed.adf9561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Anita Boyapati
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - John Janczy
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Pablo Abreu
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Wen-Chi Chen
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Stephen Godin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Harvey Chin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
3
|
Pfützner W, Polakova A, Möbs C. We are memory: B-cell responses in allergy and tolerance. Eur J Immunol 2023; 53:e2048916. [PMID: 37098972 DOI: 10.1002/eji.202048916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/24/2023] [Accepted: 04/24/2023] [Indexed: 04/27/2023]
Abstract
The significance of B-cell memory in sustaining IgE-mediated allergies but also ensuring the development of long-term allergen tolerance has remained enigmatic. However, well-thought murine and human studies have begun to shed more light on this highly disputed subject. The present mini review highlights important aspects, like the involvement of IgG1 memory B cells, the meaning of low- or high-affinity IgE antibody production, the impact of allergen immunotherapy, or the relevance of local memory established by ectopic lymphoid structures. Based on recent findings, future investigations should lead to deeper knowledge and the development of improved therapies treating allergic individuals.
Collapse
Affiliation(s)
- Wolfgang Pfützner
- Clinical & Experimental Allergy, Department of Dermatology and Allergology, Philipps-Universität Marburg, University Hospital Marburg, Marburg, Germany
| | - Alexandra Polakova
- Clinical & Experimental Allergy, Department of Dermatology and Allergology, Philipps-Universität Marburg, University Hospital Marburg, Marburg, Germany
| | - Christian Möbs
- Clinical & Experimental Allergy, Department of Dermatology and Allergology, Philipps-Universität Marburg, University Hospital Marburg, Marburg, Germany
| |
Collapse
|
4
|
Fernandes AMS, da Silva ES, Silveira EF, Belitardo EMMDA, Santiago LF, Silva RC, Dos Santos Alves V, Carneiro DM, Ferreira F, Jacquet A, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. Recombinant T-cell epitope conjugation: A new approach for Dermatophagoides hypoallergen design. Clin Exp Allergy 2023; 53:198-209. [PMID: 36176209 DOI: 10.1111/cea.14238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (AIT) is the only clinical approach that can potentially cure some allergic diseases by inducing immunological tolerance. Dermatophagoides pteronyssinus is considered as the most important source of mite allergens worldwide, with high sensitization rates for the major allergens Der p 1, Der p 2 and Der p 23. The aim of this work is to generate a hypoallergenic hybrid molecule containing T-cell epitopes from these three major allergens. METHODS The hybrid protein termed Der p 2231 containing T-cell epitopes was purified by affinity chromatography. The human IgE reactivity was verified by comparing those with the parental allergens. The hybrid was also characterized immunologically through an in vivo mice model. RESULTS The hybrid rDer p 2231 stimulated in peripheral blood mononuclear cells (PBMCs) isolated from allergic patients with higher levels of IL- 2, IL-10, IL-15 and IFN-γ, as well as lower levels of IL-4, IL-5, IL-13, TNF-α and GM-CSF. The use of hybrid molecules as a therapeutic model in D. pteronyssinus allergic mice led to the reduction of IgE production and lower eosinophilic peroxidase activity in the airways. We found increased levels of IgG antibodies that blocked the IgE binding to the parental allergens in the serum of allergic patients. Furthermore, the stimulation of splenocytes from mice treated with rDer p 2231 induced higher levels of IL-10 and IFN-γ and decreased the secretion of IL-4 and IL-5, when compared with parental allergens and D. pteronyssinus extract. CONCLUSIONS rDer p 2231 has the potential to be used in AIT in patients co-sensitized with D. pteronyssinus major allergens, once it was able to reduce IgE production, inducing allergen-specific blocking antibodies, restoring and balancing Th1/Th2 immune responses, and inducing regulatory T-cells.
Collapse
Affiliation(s)
- Antônio Márcio Santana Fernandes
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Eduardo Santos da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Elisânia Fontes Silveira
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Leonardo Freire Santiago
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Raphael Chagas Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Vitor Dos Santos Alves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Deise Malta Carneiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Luis Gustavo Carvalho Pacheco
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Neuza Maria Alcantara-Neves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Carina Silva Pinheiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
5
|
Chen Q, Xie M, Liu H, Dent AL. Development of allergen-specific IgE in a food-allergy model requires precisely timed B cell stimulation and is inhibited by Fgl2. Cell Rep 2022; 39:110990. [PMID: 35767958 PMCID: PMC9271337 DOI: 10.1016/j.celrep.2022.110990] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/12/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Immunoglobulin E (IgE) responses are a central feature of allergic disease. Using a well-established food-allergy model in mice, we show that two sensitizations with cognate B cell antigen (Ag) and adjuvant 7 days apart promotes optimal development of IgE+ germinal center (GC) B cells and high-affinity IgE production. Intervals of 3 or 14 days between Ag sensitizations lead to loss of IgE+ GC B cells and an undetectable IgE response. The immunosuppressive factors Fgl2 and CD39 are down-regulated in T follicular helper (TFH) cells under optimal IgE-sensitization conditions. Deletion of Fgl2 in TFH and T follicular regulatory (TFR) cells, but not from TFR cells alone, increase Ag-specific IgE levels and IgE-mediated anaphylactic responses. Overall, we find that Ag-specific IgE responses require precisely timed stimulation of IgE+ GC B cells by Ag. Furthermore, we show that Fgl2 is expressed by TFH cells and represses IgE. This work has implications for the development and treatment of food allergies. Using a mouse food-allergy model, Chen et al. find that allergen-specific IgE responses require precisely timed stimulation of IgE+ germinal center B cells. The authors further show that Fgl2 expressed by T follicular helper cells represses IgE. This work has implications for the development and treatment of food allergy.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Markus Xie
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hong Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
6
|
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: 29] [Impact Index Per Article: 7.3] [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
|
7
|
Study of selective immunoglobulin A deficiency among Egyptian patients with food allergy. Cent Eur J Immunol 2021; 45:184-188. [PMID: 33456329 PMCID: PMC7792446 DOI: 10.5114/ceji.2020.97907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/12/2018] [Indexed: 11/17/2022] Open
Abstract
Introduction IgA deficiency is one of the commonest primary antibody deficiencies. Although many affected individuals could be asymptomatic, selected patients suffer from recurrent mucosal infections, allergies, and autoimmune diseases. Aim of the study To investigate the prevalence of IgA deficiency among Egyptian patients with food allergy. Material and methods We studied 100 patients (62 males, 38 females; mean age, 28.6 years) with multiple food allergies who were recruited on the basis of adequate immunological assessment by history, skin prick test, and confirmed by open challenge test as well as 50 healthy controls. Measurement of levels of IgE and IgA using ELISA technique were performed for all patients and controls. Results Deficiency of IgA was detected in 67% of patients with food allergy. Serum IgA levels were significantly lower among patients with food allergy (67.3 µg/ml; range, 56.7-72.0 µg/ml) as compared to healthy control (78.6 µg/ml; range, 72.8-84 µg/ml). Both IgA and IgE levels were not statistically different between patients with food allergy only and those with combined food and aeroallergen. Among food allergic group, serum IgA levels were inversely correlated with serum IgE levels (r = –0.314, p < 0.001). Conclusions Manifestations of atopy, such as food allergy might be a present feature before diagnosis of primary immune deficiency diseases as IgA deficiency.
Collapse
|
8
|
Hadebe S, Khumalo J, Mangali S, Mthembu N, Ndlovu H, Scibiorek M, Ngomti A, Kirstein F, Brombacher F. Deletion of IL-4Rα signaling on B cells limits hyperresponsiveness depending on antigen load. J Allergy Clin Immunol 2020; 148:99-109.e5. [PMID: 33383090 PMCID: PMC8253118 DOI: 10.1016/j.jaci.2020.12.635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND B cells play an important role in allergies through secretion of IgE. IL-4 receptor α (IL-4Rα) is key in allergic asthma and regulates type 2 cytokine production, IgE secretion, and airway hyperresponsiveness. IL-4 activation of B cells is essential for class switching and contributes to the induction of B effector 2 (Be2) cells. The role of Be2 cells and signaling via IL-4Rα in B cells is not clearly defined. OBJECTIVE We sought to find out whether IL-4Rα-responsive B cells or Be2 function was essential in experimental allergic asthma. METHODS Mice lacking IL-4Rα on B cells (mb1creIL-4Rα-/lox) or littermate controls (IL-4Rα-/lox) and mice lacking IL-4 or IL-4/IL-13 on B cells were sensitized and challenged with high-dose house dust mite (>10 μg) or with low-dose house dust mite (<3 μg). We also adoptively transferred naive IL-4Rα-/lox or IL-4Rα-/- B cells into μMT-/- mice a day before sensitization or a day before challenge. We analyzed lung inflammation, cellular infiltrate, and airway hyperresponsiveness. RESULTS We found that IL-4Rα signaling on B cells was important for optimal TH2 allergic immune responses mainly when the load of antigen is limited. IL-4Rα signaling on B cells was essential for germinal centers and in the effector phase of allergic responses. Be2 cells were essential in airway hyperresponsiveness, but not in other parameters. CONCLUSIONS IL-4Rα signaling on B cells is deleterious in allergic asthma because it is required for optimal TH2 responses, Be2 function, germinal center formation, and T follicular helper cells, especially when the load of the antigen is limiting.
Collapse
Affiliation(s)
- Sabelo Hadebe
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Jermaine Khumalo
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Sandisiwe Mangali
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Nontobeko Mthembu
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Hlumani Ndlovu
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Martyna Scibiorek
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Amkele Ngomti
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Frank Kirstein
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, and South African Medical Research Council Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
9
|
El Ansari YS, Kanagaratham C, Lewis OL, Oettgen HC. IgE and mast cells: The endogenous adjuvant. Adv Immunol 2020; 148:93-153. [PMID: 33190734 DOI: 10.1016/bs.ai.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are most familiar as the effectors of type I hypersensitivity reactions including anaphylaxis. It is becoming clear however that this pair has important immunomodulatory effects on innate and adaptive cells of the immune system. In this purview, they act as endogenous adjuvants to ignite evolving immune responses, promote the transition of allergic disease into chronic illness and disrupt the development of active mechanisms of tolerance to ingested foods. Suppression of IgE-mediated mast cell activation can be exerted by molecules targeting IgE, FcɛRI or signaling kinases including Syk, or by IgG antibodies acting via inhibitory Fcγ receptors. In 2015 we reviewed the evidence for the adjuvant functions of mast cells. This update includes the original text, incorporates some important developments in the field over the past five years and discusses how interventions targeting these pathways might have promise in the development of strategies to treat allergic disease.
Collapse
Affiliation(s)
- Yasmeen S El Ansari
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Owen L Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
10
|
Asrat S, Kaur N, Liu X, Ben LH, Kajimura D, Murphy AJ, Sleeman MA, Limnander A, Orengo JM. Chronic allergen exposure drives accumulation of long-lived IgE plasma cells in the bone marrow, giving rise to serological memory. Sci Immunol 2020; 5:5/43/eaav8402. [PMID: 31924685 DOI: 10.1126/sciimmunol.aav8402] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/21/2019] [Accepted: 12/05/2019] [Indexed: 11/02/2022]
Abstract
Immunoglobulin E (IgE) plays an important role in allergic diseases. Nevertheless, the source of IgE serological memory remains controversial. We reexamined the mechanism of serological memory in allergy using a dual reporter system to track IgE+ plasma cells in mice. Short-term allergen exposure resulted in the generation of IgE+ plasma cells that resided mainly in secondary lymphoid organs and produced IgE that was unable to degranulate mast cells. In contrast, chronic allergen exposure led to the generation of long-lived IgE+ plasma cells that were primarily derived from sequential class switching of IgG1, accumulated in the bone marrow, and produced IgE capable of inducing anaphylaxis. IgE+ plasma cells were found in the bone marrow of human allergic, but not nonallergic donors, and allergen-specific IgE produced by these cells was able to induce mast cell degranulation when transferred to mice. These data demonstrate that long-lived IgE+ bone marrow plasma cells arise during chronic allergen exposure and establish serological memory in both mice and humans.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | - Xia Liu
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | | | - Andrew J Murphy
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | | | - Andre Limnander
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA.
| | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA.
| |
Collapse
|
11
|
Alessandrini F, Musiol S, Schneider E, Blanco-Pérez F, Albrecht M. Mimicking Antigen-Driven Asthma in Rodent Models-How Close Can We Get? Front Immunol 2020; 11:575936. [PMID: 33101301 PMCID: PMC7555606 DOI: 10.3389/fimmu.2020.575936] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Asthma is a heterogeneous disease with increasing prevalence worldwide characterized by chronic airway inflammation, increased mucus secretion and bronchial hyperresponsiveness. The phenotypic heterogeneity among asthmatic patients is accompanied by different endotypes, mainly Type 2 or non-Type 2. To investigate the pathomechanism of this complex disease many animal models have been developed, each trying to mimic specific aspects of the human disease. Rodents have classically been employed in animal models of asthma. The present review provides an overview of currently used Type 2 vs. non-Type 2 rodent asthma models, both acute and chronic. It further assesses the methods used to simulate disease development and exacerbations as well as to quantify allergic airway inflammation, including lung physiologic, cellular and molecular immunologic responses. Furthermore, the employment of genetically modified animals, which provide an in-depth understanding of the role of a variety of molecules, signaling pathways and receptors implicated in the development of this disease as well as humanized models of allergic inflammation, which have been recently developed to overcome differences between the rodent and human immune systems, are discussed. Nevertheless, differences between mice and humans should be carefully considered and limits of extrapolation should be wisely taken into account when translating experimental results into clinical use.
Collapse
Affiliation(s)
- Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Evelyn Schneider
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Frank Blanco-Pérez
- Molecular Allergology/Vice President's Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Melanie Albrecht
- Molecular Allergology/Vice President's Research Group, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
12
|
Looman KIM, Meel ER, Grosserichter‐Wagener C, Vissers FJM, Klingenberg JH, Jong NW, Jongste JC, Pasmans SGMA, Duijts L, Zelm MC, Moll HA. Associations of Th2, Th17, Treg cells, and IgA + memory B cells with atopic disease in children: The Generation R Study. Allergy 2020; 75:178-187. [PMID: 31385614 DOI: 10.1111/all.14010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/10/2019] [Accepted: 06/18/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND New insights into immune cells could contribute to treatment and monitoring of atopic disease. Because nongenetic factors shape the human immune system, we here studied these immune cells in a large cohort with atopic children with adjustment for prenatal and postnatal confounders. METHODS Information on atopic dermatitis, inhalant- and food-allergic sensitization, asthma lung function scores was obtained from 855 10-year-old children within the Generation R cohort. 11-color flow cytometry was performed to determine CD27+ and CD27- IgG+ , IgE+ and IgA+ memory B cells, Th1, Th2, Th17, and Treg-memory cells from venous blood. Associations between any atopic disease, the individual atopic diseases, and immune cell numbers were determined. RESULTS Children with any atopic disease had higher Th2, Treg, Treg-memory, and CD27+ IgA+ memory B-cell numbers compared to children without atopic disease. When studying the individual diseases compared to children without the individual diseases, children with atopic dermatitis, inhalant-, and food-allergic sensitization had higher memory Treg cell numbers 12.3% (95% CI 4.2; 21.0), (11.1% (95% CI 3.0; 19.8), (23.7% (95% CI 7.9; 41.8), respectively. Children with food-allergic sensitization had higher total B and CD27+ IgA+ memory B-cell numbers (15.2% [95% CI 3.2; 28.7], 22.5% [95% CI 3.9; 44.3], respectively). No associations were observed between asthma and B- or T-cell numbers. CONCLUSION Children with any atopic disease and children with inhalant- and food-allergic sensitization or atopic dermatitis had higher circulating memory Treg cells, but not higher IgE+ B-cell numbers. The associations of higher Treg and CD27+ IgA+ B-cell numbers in children with food-allergic sensitization are suggestive of TGF-β-mediated compensation for chronic inflammation.
Collapse
Affiliation(s)
- Kirsten I. M. Looman
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Evelien R. Meel
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | | | - Floor J. M. Vissers
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Janice H. Klingenberg
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Nicolette W. Jong
- Department of Internal Medicine, Division of Allergology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Johan C. Jongste
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | | | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Pediatrics, Division of Neonatology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Menno C. Zelm
- Department of Immunology and Pathology, Central Clinical School Monash University and Alfred Hospital Melbourne Victoria Australia
| | - Henriëtte A. Moll
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| |
Collapse
|
13
|
Chu DK, McCullagh DJ, Waserman S. Anaphylaxis for Internists: Definition, Evaluation, and Management, with a Focus on Commonly Encountered Problems. Med Clin North Am 2020; 104:25-44. [PMID: 31757236 DOI: 10.1016/j.mcna.2019.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Anaphylaxis is an acute systemic allergic reaction that can be life threatening. In adults, the most common causes of anaphylaxis are foods, drugs, and insect stings. This article reviews the definition, classification, evaluation, differential diagnosis, prognosis, complications, and management of anaphylaxis. Tailored for internists, the article focuses on anaphylactic medication allergies. It provides a guide to optimally evaluate and manage patients with antibiotic allergy using a simple, rapid risk stratification technique, graded antibiotic challenge (test dose), and/or allergist-guided drug desensitization. It also reviews other causes of anaphylaxis that internists are likely to encounter, and an approach to their management.
Collapse
Affiliation(s)
- Derek K Chu
- Department of Medicine, McMaster University, 1200 Main Street West, HSC-3V49, Hamilton, Ontario L8N 3Z5, Canada.
| | - David J McCullagh
- Department of Medicine, McMaster University, 1200 Main Street West, HSC-3V49, Hamilton, Ontario L8N 3Z5, Canada
| | - Susan Waserman
- Department of Medicine, McMaster University, 1200 Main Street West, HSC-3V49, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
14
|
Allergen-specific IgG + memory B cells are temporally linked to IgE memory responses. J Allergy Clin Immunol 2019; 146:180-191. [PMID: 31883847 DOI: 10.1016/j.jaci.2019.11.046] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/22/2019] [Accepted: 11/19/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND IgE is the least abundant immunoglobulin and tightly regulated, and IgE-producing B cells are rare. The cellular origin and evolution of IgE responses are poorly understood. OBJECTIVE The cellular and clonal origin of IgE memory responses following mucosal allergen exposure by sublingual immunotherapy (SLIT) were investigated. METHODS In a randomized double-blind, placebo-controlled, time course SLIT study, PBMCs and nasal biopsy samples were collected from 40 adults with seasonal allergic rhinitis at baseline and at 4, 8, 16, 28, and 52 weeks. RNA was extracted from PBMCs, sorted B cells, and nasal biopsy samples for heavy chain variable gene repertoire sequencing. Moreover, mAbs were derived from single B-cell transcriptomes. RESULTS Combining heavy chain variable gene repertoire sequencing and single-cell transcriptomics yielded direct evidence of a parallel boost of 2 clonally and functionally related B-cell subsets of short-lived IgE+ plasmablasts and IgG+ memory B cells. Mucosal grass pollen allergen exposure by SLIT resulted in highly diverse IgE and IgGE repertoires. These were extensively mutated and appeared relatively stable as per heavy chain isotype, somatic hypermutations, and clonal composition. Single IgGE+ memory B-cell and IgE+ preplasmablast transcriptomes encoded antibodies that were specific for major grass pollen allergens and able to elicit basophil activation at very low allergen concentrations. CONCLUSION For the first time, we have shown that on mucosal allergen exposure, human IgE memory resides in allergen-specific IgG+ memory B cells. These cells rapidly switch isotype, expand into short-lived IgE+ plasmablasts, and serve as a potential target for therapeutic intervention.
Collapse
|
15
|
Preventive effects of "ovalbumin-conjugated celastrol-loaded nanomicelles'' in a mouse model of ovalbumin-induced allergic airway inflammation. Eur J Pharm Sci 2019; 143:105172. [PMID: 31841695 DOI: 10.1016/j.ejps.2019.105172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/15/2019] [Accepted: 12/01/2019] [Indexed: 12/22/2022]
Abstract
Allergies affect a significant proportion of the world's population, and existing vaccination strategies to restrict their adverse pathologies often render side-effects. The aim of this study was to design a new vaccine for allergen-specific immunotherapy (SIT), and to investigate its preventive effects during allergic inflammation. We constructed ovalbumin (OVA)-conjugated celastrol-loaded nanomicelles (OVA-NMs-celastrol), wherein celastrol (a bioactive anti-inflammatory compound) was loaded into carboxyl-functioned polymeric nanomicelles using a thin-film hydration method. OVA was used as a model allergen and conjugated on nanomicelles. The OVA-NMs-celastrol obtained were characterized based on particle size, morphology, drug encapsulation efficiency, and drug loading percentage. Further, the preventive effect of OVA-NMs-celastrol was evaluated in a mouse model of allergic asthma. Our results showed that OVA-NMs-celastrol possessed valuable characteristics such as small particle size (50.72 ± 0.98 nm) and spherical-like shape, with celastrol encapsulation efficiency of 99.89 ± 0.85% and a drug loading percentage of 4.76 ± 0.03%. Further, in vivo results showed that treatment with OVA-NMs-celastrol could decrease OVA specific IgE and histamine levels, Th2 cytokine (IL-4, IL-5) levels, and inflammatory cell infiltration in the lung tissues. Moreover, it could enhance the OVA specific IgG1 and IgG2a levels and decrease the IgE / IgG2a ratio. These results demonstrate the successful construction of OVA-NMs-celastrol as a potential vaccine candidate for use in SIT for allergic inflammation.
Collapse
|
16
|
Messingham KN, Crowe TP, Fairley JA. The Intersection of IgE Autoantibodies and Eosinophilia in the Pathogenesis of Bullous Pemphigoid. Front Immunol 2019; 10:2331. [PMID: 31636640 PMCID: PMC6787172 DOI: 10.3389/fimmu.2019.02331] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune blistering disease characterized by autoantibodies targeting cellular adhesion molecules. While IgE autoantibodies are occasionally reported in other autoimmune blistering diseases, BP is unique in that most BP patients develop an IgE autoantibody response. It is not known why BP patients develop self-reactive IgE and the precise role of IgE in BP pathogenesis is not fully understood. However, clinical evidence suggests an association between elevated IgE antibodies and eosinophilia in BP patients. Since eosinophils are multipotent effector cells, capable cytotoxicity and immune modulation, the putative interaction between IgE and eosinophils is a primary focus in current studies aimed at understanding the key components of disease pathogenesis. In this review, we provide an overview of BP pathogenesis, highlighting clinical and experimental evidence supporting central roles for IgE and eosinophils as independent mediators of disease and via their interaction. Additionally, therapeutics targeting IgE, the Th2 axis, or eosinophils are also discussed.
Collapse
Affiliation(s)
- Kelly N Messingham
- Department of Dermatology, The University of Iowa, Iowa City, IA, United States
| | - Tyler P Crowe
- Department of Dermatology, The University of Iowa, Iowa City, IA, United States
| | - Janet A Fairley
- Department of Dermatology, The University of Iowa, Iowa City, IA, United States.,Iowa City VA Medical Center, Iowa City, IA, United States
| |
Collapse
|
17
|
Li X, Wang N, Zhao Y, Zhang Y, Liu Z. Species Specificity on Interaction between IgE and FcεRI. Curr Pharm Biotechnol 2019; 20:690-695. [PMID: 31258078 DOI: 10.2174/1389201020666190619122325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 11/22/2022]
Abstract
Allergic diseases are one of the most prevalent diseases at present, it is imperative to understanding the pathophysiology and treatment strategies for allergic diseases. In this process, the binding of IgE and FcεRI on effector cells plays a critical role in triggering allergic reactions. However, the species specificity of the interaction between IgE and FcεRI has not been clearly explained. This review described the characteristics and the interaction mechanism in the allergic reaction of IgE and FcεRI and summarized the species specificity between IgE and FcεRI.
Collapse
Affiliation(s)
- Xiangsheng Li
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Nannan Wang
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yang Zhao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yanfen Zhang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China.,Offices of Science and Technology, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| |
Collapse
|
18
|
Dias ASO, Santos ICL, Delphim L, Fernandes G, Endlich LR, Cafasso MOSD, Maranhão AL, da Silva SR, Andrade RM, Agrawal A, Linhares UC, Bento CAM. Serum leptin levels correlate negatively with the capacity of vitamin D to modulate the in vitro cytokines production by CD4 + T cells in asthmatic patients. Clin Immunol 2019; 205:93-105. [PMID: 31173888 DOI: 10.1016/j.clim.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/25/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022]
Abstract
Both obesity and low vitamin D levels have been associated with allergic asthma (AA) severity. In the present study, severity of AA was associated with obesity but to the in vitro IgE production. In those patients, higher levels of IL-5, IL-6 and IL-17 were quantified in CD4+ T-cell cultures as compared with patients with mild and moderate AA. In addition, the lowest IL-10 levels were detected in the cell cultures from patients with a worse prognosis. Interestingly, the occurrence of AA elevates the plasma levels of leptin, and this adipokine was positively correlated with the release of IL-5, IL-6 and IL-17, but inversely correlated with IL-10 production, by CD4+ T-cells from patients. In AA-derived CD4+ T-cell cultures, 1,25(OH)2D3 was less efficient at inhibiting IL-5, IL-6 and IL-17 production, and up regulating IL-10 release, as those from healthy subjects. Interestingly, the in vitro immunomodulatory effects of vitamin D were inversely correlated with serum leptin levels. In summary, our findings suggested that obesity, probably due to the overproduction of leptin, negatively impacts AA as it favors imbalance between Th2/Th17 and regulatory phenotypes. The deleterious effects of leptin may also be due to its ability to counter-regulate the immunosuppressive effects of vitamin D.
Collapse
Affiliation(s)
- Aleida S O Dias
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil; Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Brazil
| | - Isabelle C L Santos
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Letícia Delphim
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Gabriel Fernandes
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Larissa R Endlich
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | | | - Ana Lúcia Maranhão
- Pulmonology Service, Federal University of the State of Rio de Janeiro, Brazil
| | | | - Regis M Andrade
- Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Brazil
| | - Anshu Agrawal
- Department of Medicine, University of California, Irvine, CA, USA
| | - Ulisses C Linhares
- Department of Morphological Sciences, Federal University of the State of Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil; Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Ramadani F, Bowen H, Gould HJ, Fear DJ. Transcriptional Analysis of the Human IgE-Expressing Plasma Cell Differentiation Pathway. Front Immunol 2019; 10:402. [PMID: 30915071 PMCID: PMC6421273 DOI: 10.3389/fimmu.2019.00402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/15/2019] [Indexed: 11/13/2022] Open
Abstract
IgE is secreted by plasma cells (PCs) and is central to allergic disease. Using an ex vivo tonsil B cell culture system, which mimics the Th2 responses in vivo, we have recently characterized the development pathway of human IgE-expressing PCs. In this system, as in mice, we reported the predisposition of IgE-expressing B cells to differentiate into PCs. To gain a comprehensive understanding of the molecular events involved in the differentiation of human IgE+ B cells into PCs we have used the Illumina HumanHT-12 v4 Expression BeadChip array to analyse the gene expression profile of ex vivo generated human IgE+ B cells at various stages of their differentiation into PCs. We also compared the transcription profiles of IgE+ and IgG1+ cells to discover isotype-specific patterns. Comparisons of IgE+ and IgG1+ cell transcriptional profiles revealed molecular signatures specific for IgE+ cells, which diverge from their IgG1+ cell counterparts upon differentiation into PCs. At the germinal center (GC) stage of development, unlike in some mouse studies of IgE biology, we observed similar rates of apoptosis and no significant differences in the expression of apoptosis-associated genes between the IgE+ and IgG1+ B cells. We identified a gene interaction network associated with early growth response 1 (EGR1) that, together with the up-regulated IRF4, may account for the predisposition of IgE+ B cells to differentiate into PCs. However, despite their swifter rates of PC differentiation, the transcription profile of IgE+ PCs is more closely related to IgE+ and IgG1+ plasmablasts (PBs) than to IgG1+ PCs, suggesting that the terminal differentiation of IgE+ cells is impeded. We also show that IgE+ PCs have increased levels of apoptosis suggesting that the IgE+ PCs generated in our in vitro tonsil B cell cultures, as in mice, are short-lived. We identified gene regulatory networks as well as cell cycle and apoptosis signatures that may explain the diverging PC differentiation programme of these cells. Overall, our study provides a detailed analysis of the transcriptional pathways underlying the differentiation of human IgE-expressing B cells and points to molecular signatures that regulate IgE+ PC differentiation and function.
Collapse
Affiliation(s)
- Faruk Ramadani
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, United Kingdom.,Asthma UK Centre, Allergic Mechanisms in Asthma, King's College London, London, United Kingdom
| | - Holly Bowen
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, United Kingdom.,Asthma UK Centre, Allergic Mechanisms in Asthma, King's College London, London, United Kingdom
| | - Hannah J Gould
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, United Kingdom.,Asthma UK Centre, Allergic Mechanisms in Asthma, King's College London, London, United Kingdom
| | - David J Fear
- Asthma UK Centre, Allergic Mechanisms in Asthma, King's College London, London, United Kingdom.,Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
20
|
Hoh RA, Boyd SD. Gut Mucosal Antibody Responses and Implications for Food Allergy. Front Immunol 2018; 9:2221. [PMID: 30319658 PMCID: PMC6170638 DOI: 10.3389/fimmu.2018.02221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/07/2018] [Indexed: 12/28/2022] Open
Abstract
The gastrointestinal mucosa is a critical environmental interface where plasma cells and B cells are exposed to orally-ingested antigens such as food allergen proteins. It is unclear how the development of B cells and plasma cells in the gastrointestinal mucosa differs between healthy humans and those with food allergy, and how B cells contribute to, or are affected by, the breakdown of oral tolerance. In particular, the antibody gene repertoires associated with symptomatic allergy have only begun to be characterized in full molecular detail. Here, we review literature concerning B cells and plasma cells in the gastrointestinal system in the context of food allergy, with a focus on human studies.
Collapse
Affiliation(s)
- Ramona A Hoh
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, United States
| |
Collapse
|
21
|
O'Konek JJ, Landers JJ, Janczak KW, Goel RR, Mondrusov AM, Wong PT, Baker JR. Nanoemulsion adjuvant-driven redirection of T H2 immunity inhibits allergic reactions in murine models of peanut allergy. J Allergy Clin Immunol 2018; 141:2121-2131. [PMID: 29655584 DOI: 10.1016/j.jaci.2018.01.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 01/17/2018] [Accepted: 01/27/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immunotherapy for food allergies involves progressive increased exposures to food that result in desensitization to food allergens in some subjects but not tolerance to the food. Therefore new approaches to suppress allergic immunity to food are necessary. Previously, we demonstrated that intranasal immunization with a nanoemulsion (NE) adjuvant induces robust mucosal antibody and TH17-polarized immunity, as well as systemic TH1-biased cellular immunity with suppression of pre-existing TH2-biased immunity. OBJECTIVE We hypothesized that immunization with food in conjunction with the nanoemulsion adjuvant could lead to modulation of allergic reactions in food allergy by altering pre-existing allergic immunity and enhancing mucosal immunity. METHODS Mice were sensitized to peanut with aluminum hydroxide or cholera toxin. The animals were then administered 3 monthly intranasal immunizations with peanut in the nanoemulsion adjuvant or saline. Mice were then challenged with peanut to examine allergen reactivity. RESULTS The NE intranasal immunizations resulted in marked decreases in TH2 cytokine, IgG1, and IgE levels, whereas TH1 and mucosal TH17 immune responses were increased. After allergen challenge, these mice showed significant reductions in allergic hypersensitivity. Additionally, the NE immunizations significantly increased antigen-specific IL-10 production and regulatory T-cell counts, and the protection induced by NE was dependent in part on IL-10. Control animals immunized with intranasal peanut in saline had no modulation of their allergic response. CONCLUSIONS NE adjuvant-mediated induction of mucosal TH17 and systemic TH1-biased immunity can suppress TH2-mediated allergy through multiple mechanisms and protect against anaphylaxis. These results suggest the potential therapeutic utility of this approach in the setting of food allergy.
Collapse
Affiliation(s)
- Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| | - Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | | | - Rishi R Goel
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Anna M Mondrusov
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Pamela T Wong
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
22
|
Shamji MH, Durham SR. Mechanisms of allergen immunotherapy for inhaled allergens and predictive biomarkers. J Allergy Clin Immunol 2017; 140:1485-1498. [PMID: 29221580 DOI: 10.1016/j.jaci.2017.10.010] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022]
Abstract
Allergen immunotherapy is effective in patients with IgE-dependent allergic rhinitis and asthma. When immunotherapy is given continuously for 3 years, there is persistent clinical benefit for several years after its discontinuation. This disease-modifying effect is both antigen-specific and antigen-driven. Clinical improvement is accompanied by decreases in numbers of effector cells in target organs, including mast cells, basophils, eosinophils, and type 2 innate lymphoid cells. Immunotherapy results in the production of blocking IgG/IgG4 antibodies that can inhibit IgE-dependent activation mediated through both high-affinity IgE receptors (FcεRI) on mast cells and basophils and low-affinity IgE receptors (FcεRII) on B cells. Suppression of TH2 immunity can occur as a consequence of either deletion or anergy of antigen-specific T cells; induction of antigen-specific regulatory T cells; or immune deviation in favor of TH1 responses. It is not clear whether the altered long-term memory resides within the T-cell or the B-cell compartment. Recent data highlight the role of IL-10-producing regulatory B cells and "protective" antibodies that likely contribute to long-term tolerance. Understanding mechanisms underlying induction and persistence of tolerance should identify predictive biomarkers of clinical response and discover novel and more effective strategies for immunotherapy.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology; Section of Inflammation, Repair and Development; National Heart and Lung Institute; Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology; Section of Inflammation, Repair and Development; National Heart and Lung Institute; Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| |
Collapse
|
23
|
Hikosaka M, Murata A, Yoshino M, Hayashi SI. Correlation between cell aggregation and antibody production in IgE-producing plasma cells. Biochem Biophys Rep 2017; 10:224-231. [PMID: 28955750 PMCID: PMC5614669 DOI: 10.1016/j.bbrep.2017.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/01/2017] [Accepted: 04/18/2017] [Indexed: 01/18/2023] Open
Abstract
Allergic conditions result in the increase of immunoglobulin (Ig)E-producing plasma cells (IgE-PCs); however, it is unclear how IgE production is qualitatively controlled. In this study, we found that IgE-PCs in spleen of immunized mice formed homotypic cell aggregates. By employing IgE-producing hybridomas (IgE-hybridomas) as a model of IgE-PCs, we showed that these cells formed aggregates in the presence of specific antigens (Ags). The formation of the Ag-induced cell aggregation involved secreted IgE and Fcγ receptor (FcγR)II/FcγRIII, but not FcεRs. Ag-induced cell aggregation plus lipopolysaccharide signaling resulted in an enhancement of IgE production in aggregated IgE-hybridomas. Furthermore, the administration of anti-FcγRII/FcγRIII antagonistic monoclonal antibody to immunized mice tended to reduce the splenic IgE-PC aggregation as well as the serum IgE levels. Taken together, our results suggested that Ag-IgE complexes induced IgE-PCs aggregation via FcγRII/FcγRIII, leading to the enhancement of IgE production. These findings suggest the presence of a novel mechanism for regulation of IgE production.
Collapse
Affiliation(s)
- Mari Hikosaka
- Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-Cho, Yonago, Tottori 683-8503, Japan
| | - Akihiko Murata
- Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-Cho, Yonago, Tottori 683-8503, Japan
| | - Miya Yoshino
- Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-Cho, Yonago, Tottori 683-8503, Japan
| | - Shin-Ichi Hayashi
- Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-Cho, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
24
|
Hamizan AW, Rimmer J, Alvarado R, Sewell WA, Kalish L, Sacks R, Harvey RJ. Positive allergen reaction in allergic and nonallergic rhinitis: a systematic review. Int Forum Allergy Rhinol 2017; 7:868-877. [PMID: 28727909 DOI: 10.1002/alr.21988] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/02/2017] [Accepted: 06/18/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The diagnosis of allergic rhinitis (AR) is based on cutaneous and serological assessment to determine immunoglobulin E (IgE)-mediated disease. However, discrepancies between these tests and nasal provocation exist. Patients diagnosed as non-allergic rhinitis (NAR) but with positive nasal allergen provocation test (NAPT) may represent a local allergic condition or entopy, still suitable to allergy interventions. The objective of this study was to determine the frequency of nasal reactivity toward allergens among AR and NAR patients, and to describe the diagnostic characteristics of NAPT methodologies. METHODS EMBASE (1947-) and Medline (1946-) were searched until December 8, 2015. A search strategy was used to identify studies on AR or NAR patients subjected to diagnostic local nasal provocation. All studies providing original NAPT data among the AR or NAR population were included. Meta-analysis of proportion data was presented as a weighted probability % (95% confidence interval [CI]). RESULTS The search yielded 4504 studies and 46 were included. The probability of nasal allergen reactivity for the AR population was 86.3% (95% CI, 84.4 to 88.1) and in NAR was 24.7% (95% CI, 22.3 to 27.2). Reactivity was high with pollen for both AR 97.1% (95% CI, 94.2 to 99.2) and NAR 47.5% (95% CI, 34.8 to 60.4), and lowest with dust for both AR 79.1% (95% CI, 76.4 to 81.6) and NAR 12.2% (95% CI, 9.9 to 14.7). NAPT yielded high positivity when defined by subjective end-points: AR 91.0% (95% CI, 86.6 to 94.8) and NAR 30.2% (95% CI, 22.9 to 37.9); and lower with objective end-points: AR 80.8% (95% CI, 76.8 to 84.5) and NAR 14.1% (95% CI, 11.2 to 17.2). CONCLUSION Local allergen reactivity is demonstrated in 26.5% of patients previously considered non-allergic. Similarly, AR, when defined by skin-prick test (SPT) or serum specific IgE (sIgE), may lead to 13.7% of patients with inaccurate allergen sensitization or non-allergic etiologies.
Collapse
Affiliation(s)
- Aneeza W Hamizan
- Rhinology and Skull Base Research Group, St Vincent's Centre for Applied Medical Research, University of New South Wales, Sydney, Australia.,Department of Otolaryngology and Head and Neck Surgery, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Janet Rimmer
- St Vincent's Clinic, St Vincent's Hospital, Sydney, Australia.,The Woolcock Institute, Sydney University, Sydney, Australia
| | - Raquel Alvarado
- Rhinology and Skull Base Research Group, St Vincent's Centre for Applied Medical Research, University of New South Wales, Sydney, Australia
| | - William A Sewell
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia.,Garvan Institute, Sydney, Australia
| | - Larry Kalish
- Sydney Medical School, University of Sydney, Sydney, Australia.,Department of Otolaryngology-Head and Neck Surgery, Concord General Hospital, University of Sydney, Sydney, Australia
| | - Raymond Sacks
- Department of Otolaryngology-Head and Neck Surgery, Concord General Hospital, University of Sydney, Sydney, Australia.,Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Richard J Harvey
- Rhinology and Skull Base Research Group, St Vincent's Centre for Applied Medical Research, University of New South Wales, Sydney, Australia.,Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
25
|
Kowalewicz-Kulbat M, Locht C. BCG and protection against inflammatory and auto-immune diseases. Expert Rev Vaccines 2017; 16:1-10. [PMID: 28532186 DOI: 10.1080/14760584.2017.1333906] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Bacillus Calmette-Guérin (BCG) is the only available vaccine against tuberculosis. Although its protective efficacy against pulmonary tuberculosis is still under debate, it provides protection against other mycobacterial diseases. BCG is also an effective therapy against superficial bladder cancer and potentially decreases overall childhood mortality. Areas covered: The purpose of this paper is to provide a state-of-the-art summary of the beneficial effects of BCG in inflammatory and auto-immune diseases. As a strong inducer of Th1 type immunity, BCG has been reported to protect against atopic conditions, such as allergic asthma, a Th2-driven disorder. Its protective effect has been well documented in mice, but still awaits definitive evidence in humans. Similarly, murine studies have shown a protective effect of BCG against auto-immune diseases, such as multiple sclerosis and insulin-dependent diabetes, but studies in humans have come to conflicting conclusions. Expert commentary: Studies in mice have shown a beneficial effect of the BCG vaccine against allergic asthma, multiple sclerosis and diabetes. However, the understanding of its mechanism is still fragmentary and requires further in depth research. Some observational or intervention studies in humans have also suggested a beneficial effect, but definitive evidence for this requires confirmation in carefully conducted prospective studies.
Collapse
Affiliation(s)
- Magdalena Kowalewicz-Kulbat
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland
| | - Camille Locht
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland.,b Center for Infection and Immunity of Lille , Institut Pasteur de Lille , Lille , France.,c Center for Infection and Immunity of Lille , Inserm U1019 , Lille , France.,d Center for Infection and Immunity of Lille , CNRS UMR 8204 , Lille , France.,e Center for Infection and Immunity of Lille , Université Lille Nord de France , Lille , France
| |
Collapse
|
26
|
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]
|
27
|
Rondón C, Bogas G, Barrionuevo E, Blanca M, Torres MJ, Campo P. Nonallergic rhinitis and lower airway disease. Allergy 2017; 72:24-34. [PMID: 27439024 DOI: 10.1111/all.12988] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2016] [Indexed: 12/17/2022]
Abstract
In the past years, several investigators have demonstrated the existence of local nasal responses in some patients with typical allergic rhinitis symptoms but without atopy and have defined a new phenotype called local allergic rhinitis (LAR) or 'entopy'. In a percentage of LAR subjects, the upper airway disease is also associated with lower airway symptoms. After the description of this phenotype, the differential diagnosis between LAR and nonallergic rhinitis (NAR) has become a challenge for the clinician. To correctly identify LAR patients is of high importance for treatment and management of these patients, and for an appropriate inclusion of patients in clinical trials and genetics studies. The treatment of LAR patients, in contrast with NAR, is oriented to allergen avoidance and specific treatment. Allergen immunotherapy, the aetiological treatment for allergic respiratory diseases, has demonstrated to be an effective and safe treatment in LAR, increasing immunological tolerance, and reducing the clinical symptoms and the use of medication. In this article, the important and novel aspects of LAR in terms of mechanisms, diagnosis and treatment will be discussed. Also, the involvement of the lower airway and the potential role of IgE in the bronchial disease will be also reviewed.
Collapse
Affiliation(s)
- C. Rondón
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| | - G. Bogas
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| | - E. Barrionuevo
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| | - M. Blanca
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| | - M. J. Torres
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| | - P. Campo
- Allergy Unit; IBIMA-Regional University Hospital of Málaga; UMA; Malaga Spain
| |
Collapse
|
28
|
Ramadani F, Bowen H, Upton N, Hobson PS, Chan YC, Chen JB, Chang TW, McDonnell JM, Sutton BJ, Fear DJ, Gould HJ. Ontogeny of human IgE-expressing B cells and plasma cells. Allergy 2017; 72:66-76. [PMID: 27061189 PMCID: PMC5107308 DOI: 10.1111/all.12911] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2016] [Indexed: 12/31/2022]
Abstract
Background IgE‐expressing (IgE+) plasma cells (PCs) provide a continuous source of allergen‐specific IgE that is central to allergic responses. The extreme sparsity of IgE+ cells in vivo has confined their study almost entirely to mouse models. Objective To characterize the development pathway of human IgE+PCs and to determine the ontogeny of human IgE+PCs. Methods To generate human IgE+ cells, we cultured tonsil B cells with IL‐4 and anti‐CD40. Using FACS and RT‐PCR, we examined the phenotype of generated IgE+ cells, the capacity of tonsil B‐cell subsets to generate IgE+PCs and the class switching pathways involved. Results We have identified three phenotypic stages of IgE+PC development pathway, namely (i) IgE+germinal centre (GC)‐like B cells, (ii) IgE+PC‐like ‘plasmablasts’ and (iii) IgE+PCs. The same phenotypic stages were also observed for IgG1+ cells. Total tonsil B cells give rise to IgE+PCs by direct and sequential switching, whereas the isolated GC B‐cell fraction, the main source of IgE+PCs, generates IgE+PCs by sequential switching. PC differentiation of IgE+ cells is accompanied by the down‐regulation of surface expression of the short form of membrane IgE (mIgES), which is homologous to mouse mIgE, and the up‐regulation of the long form of mIgE (mIgEL), which is associated with an enhanced B‐cell survival and expressed in humans, but not in mice. Conclusion Generation of IgE+PCs from tonsil GC B cells occurs mainly via sequential switching from IgG. The mIgEL/mIgES ratio may be implicated in survival of IgE+ B cells during PC differentiation and allergic disease.
Collapse
Affiliation(s)
- F. Ramadani
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - H. Bowen
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - N. Upton
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - P. S. Hobson
- Division of Asthma; Allergy and Lung Biology; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - Y.-C. Chan
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - J.-B. Chen
- Genomics Research Center; Academia Sinica; Taipei Taiwan
| | - T. W. Chang
- Genomics Research Center; Academia Sinica; Taipei Taiwan
| | - J. M. McDonnell
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - B. J. Sutton
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - D. J. Fear
- Division of Asthma; Allergy and Lung Biology; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| | - H. J. Gould
- Randall Division of Cell and Molecular Biohphysics; King's College; London UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms in Asthma; London UK
| |
Collapse
|
29
|
Wu YL, Stubbington MJT, Daly M, Teichmann SA, Rada C. Intrinsic transcriptional heterogeneity in B cells controls early class switching to IgE. J Exp Med 2016; 214:183-196. [PMID: 27994069 PMCID: PMC5206502 DOI: 10.1084/jem.20161056] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/27/2016] [Accepted: 11/13/2016] [Indexed: 12/11/2022] Open
Abstract
Combining novel mouse reporters and single-cell transcriptomic analyses, Wu et al. uncover differential activation thresholds for the transcripts that direct antibody class switching to IgE versus IgG1 in response to IL-4 and explain how cell-intrinsic transcriptional heterogeneity governs CSR. Noncoding transcripts originating upstream of the immunoglobulin constant region (I transcripts) are required to direct activation-induced deaminase to initiate class switching in B cells. Differential regulation of Iε and Iγ1 transcription in response to interleukin 4 (IL-4), hence class switching to IgE and IgG1, is not fully understood. In this study, we combine novel mouse reporters and single-cell RNA sequencing to reveal the heterogeneity in IL-4–induced I transcription. We identify an early population of cells expressing Iε but not Iγ1 and demonstrate that early Iε transcription leads to switching to IgE and occurs at lower activation levels than Iγ1. Our results reveal how probabilistic transcription with a lower activation threshold for Iε directs the early choice of IgE versus IgG1, a key physiological response against parasitic infestations and a mediator of allergy and asthma.
Collapse
Affiliation(s)
- Yee Ling Wu
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | | | - Maria Daly
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Sarah A Teichmann
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, England, UK
| | - Cristina Rada
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| |
Collapse
|
30
|
Seifert M, Küppers R. Human memory B cells. Leukemia 2016; 30:2283-2292. [DOI: 10.1038/leu.2016.226] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022]
|
31
|
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
|
32
|
Diagnostic Utility of Total IgE in Foods, Inhalant, and Multiple Allergies in Saudi Arabia. J Immunol Res 2016; 2016:1058632. [PMID: 27314052 PMCID: PMC4897674 DOI: 10.1155/2016/1058632] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/04/2016] [Accepted: 04/28/2016] [Indexed: 11/17/2022] Open
Abstract
Objective. To assess the diagnostic significance of total IgE in foods, inhalant, and multiple allergies. Methods. Retrospective review of the laboratory records of patients who presented with clinical suspicion of food or inhalant allergy between January 2013 and December 2014. Total IgE level was defined as positive for a value >195 kU/L; and diagnosis was confirmed by the detection of specific IgE (golden standard) for at least one food or inhalant allergen and at least two allergens in multiple allergies. Results. A total of 1893 (male ratio = 0.68, mean age = 39.0 ± 19.2 years) patients were included. Total IgE had comparable sensitivity (55.8% versus 59.6%) and specificity (83.9% versus 84.4%) in food versus inhalant allergy, respectively, but a superior PPV in inhalant allergy (79.1% versus 54.4%). ROC curve analysis showed a better diagnostic value in inhalant allergies (AUC = 0.817 (95% CI = 0.796-0.837) versus 0.770 (95% CI = 0.707-0.833)). In multiple allergies, total IgE had a relatively good sensitivity (78.6%), while negative IgE testing (<195 kU/L) predicted the absence of multiple allergies with 91.5% certitude. Conclusion. Total IgE assay is not efficient as a diagnostic test for foods, inhalant, or multiple allergies. The best strategy should refer to specific IgE testing guided by a comprehensive atopic history.
Collapse
|
33
|
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
|
34
|
Smit JJ, Noti M, O’Mahony L. The use of animal models to discover immunological mechanisms underpinning sensitization to food allergens. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.ddmod.2016.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
35
|
|