1
|
Grijincu M, Buzan MR, Zbîrcea LE, Păunescu V, Panaitescu C. Prenatal Factors in the Development of Allergic Diseases. Int J Mol Sci 2024; 25:6359. [PMID: 38928067 PMCID: PMC11204337 DOI: 10.3390/ijms25126359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Allergic diseases are showing increasing prevalence in Western societies. They are characterized by a heightened reactivity towards otherwise harmless environmental stimuli. Allergic diseases showing a wide range of severity of symptoms have a significant impact on the quality of life of affected individuals. This study aims to highlight the mechanisms that induce these reactions, how they progress, and which prenatal factors influence their development. Most frequently, the reaction is mediated by immunoglobulin E (IgE) produced by B cells, which binds to the surface of mast cells and basophils and triggers an inflammatory response. The antibody response is triggered by a shift in T-cell immune response. The symptoms often start in early childhood with eczema or atopic dermatitis and progress to allergic asthma in adolescence. An important determinant of allergic diseases seems to be parental, especially maternal history of allergy. Around 30% of children of allergic mothers develop allergic sensitization in childhood. Genes involved in the regulation of the epithelial barrier function and the T-cell response were found to affect the predisposition to developing allergic disorders. Cord blood IgE was found to be a promising predictor of allergic disease development. Fetal B cells produce IgE starting at the 20th gestation week. These fetal B cells could be sensitized together with mast cells by maternal IgE and IgE-allergen complexes crossing the placental barrier via the low-affinity IgE receptor. Various factors were found to facilitate these sensitizations, including pesticides, drugs, exposure to cigarette smoke and maternal uncontrolled asthma. Prenatal exposure to microbial infections and maternal IgG appeared to play a role in the regulation of T-cell response, indicating a protective effect against allergy development. Additional preventive factors were dietary intake of vitamin D and omega 3 fatty acids as well as decreased maternal IgE levels. The effect of exposure to food allergens during pregnancy was inconclusive, with studies having found both sensitizing and protective effects. In conclusion, prenatal factors including genetics, epigenetics and fetal environmental factors have an important role in the development of allergic disorders in later life. Children with a genetic predisposition are at risk when exposed to cigarette smoke as well as increased maternal IgE in the prenatal period. Maternal diet during pregnancy and immunization against certain allergens could help in the prevention of allergy in predisposed children.
Collapse
Affiliation(s)
- Manuela Grijincu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Maria-Roxana Buzan
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Lauriana-Eunice Zbîrcea
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Virgil Păunescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| |
Collapse
|
2
|
Ding Z, Mulder J, Robinson MJ. The origins and longevity of IgE responses as indicated by serological and cellular studies in mice and humans. Allergy 2023; 78:3103-3117. [PMID: 37417548 PMCID: PMC10952832 DOI: 10.1111/all.15799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
The existence of long-lived IgE antibody-secreting cells (ASC) is contentious, with the maintenance of sensitization by the continuous differentiation of short-lived IgE+ ASC a possibility. Here, we review the epidemiological profile of IgE production, and give an overview of recent discoveries made on the mechanisms regulating IgE production from mouse models. Together, these data suggest that for most individuals, in most IgE-associated diseases, IgE+ ASC are largely short-lived cells. A subpopulation of IgE+ ASC in humans is likely to survive for tens of months, although due to autonomous IgE B cell receptor (BCR) signaling and antigen-driven IgE+ ASC apoptosis, in general IgE+ ASC probably do not persist for the decades that other ASC are inferred to do. We also report on recently identified memory B cell transcriptional subtypes that are the likely source of IgE in ongoing responses, highlighting the probable importance of IL-4Rα in their regulation. We suggest the field should look at dupilumab and other drugs that prohibit IgE+ ASC production as being effective treatments for IgE-mediated aspects of disease in most individuals.
Collapse
Affiliation(s)
- Zhoujie Ding
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | - Jesse Mulder
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | | |
Collapse
|
3
|
Čelakovská J, Čermáková E, Boudková P, Andrýs C, Krejsek J. Evaluation of Leukocytes, B and T Lymphocytes, and expression of CD200 and CD23 on B lymphocytes in Patients with Atopic Dermatitis on Dupilumab Therapy-Pilot Study. Dermatol Ther (Heidelb) 2023; 13:1171-1192. [PMID: 37097547 PMCID: PMC10149535 DOI: 10.1007/s13555-023-00918-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/16/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND There are a lot of studies that describe the change in quantity of T cells in patients with atopic dermatitis (AD) compared with healthy subjects. Other components of lymphocytes such as B cells are not examined as well as T cells. OBJECTIVE We focus on immunophenotyping of B cells with their subsets (memory, naïve, switched, non-switched) and the expression of CD23 and CD200 markers in patients with AD with and without dupilumab therapy. We also evaluate the count of leukocytes and their subsets, T lymphocytes (CD4+, CD8+), natural killer (NK) cells, and T regulatory cells. METHODS A total of 45 patients suffering from AD were examined: 32 patients without dupilumab treatment (10 men, 22 women, average age 35 years), 13 patients with dupilumab treatment (7 men, 6 women, average age 43.4 years), and 30 subjects as a control group (10 men, 20 women, average age 44.7 years). Immunophenotype was examined by flow cytometry in which monoclonal antibodies with fluorescent molecules were used. We compared the absolute and relative count of leukocytes and their subsets, T lymphocytes (CD4+ , CD8+), NK cells, T regulatory cells, absolute and relative count of B lymphocytes (memory, naïve, non-switched, switched, transient), and expression of CD23 and CD200 activation markers on B cells and on their subsets in patients with AD and control group. For statistical analysis we used nonparametric Kruskal-Wallis one-factor analysis of variance with post hoc by Dunn's test with Bonferroni modification of significance level. RESULTS In patients with AD with and without dupilumab therapy we confirmed the significantly higher count of neutrophils, monocytes, and eosinophils; there was no difference in absolute count of B cells, NK cells and transitional B cells compared with control subjects. We confirmed higher expression of activation marker CD23 on total, memory, naïve, non-switched, and switched B lymphocytes and higher expression of CD200 on total B lymphocytes in both groups of patients with AD compared with controls. In patients without dupilumab therapy we confirmed significantly higher count of relative monocytes, relative eosinophils, and higher expression of CD200 on memory, naïve, and non-switched B lymphocytes compared with controls. In patients with dupilumab therapy we confirmed significantly higher expression of CD200 on switched B lymphocytes, higher count of relative CD4+ T lymphocytes, and lower count of absolute CD8+ T lymphocytes compared with controls. CONCLUSION This pilot study shows higher expression of CD23 on B lymphocytes and on their subsets in patients with AD with and without dupilumab therapy. The higher expression of CD200 on switched B lymphocytes is confirmed only in patients with AD with dupilumab therapy.
Collapse
Affiliation(s)
- Jarmila Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic.
| | - Eva Čermáková
- Department of Medical Biophysics, Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Petra Boudková
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| |
Collapse
|
4
|
Plattner K, Bachmann MF, Vogel M. On the complexity of IgE: The role of structural flexibility and glycosylation for binding its receptors. FRONTIERS IN ALLERGY 2023; 4:1117611. [PMID: 37056355 PMCID: PMC10089267 DOI: 10.3389/falgy.2023.1117611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
It is well established that immunoglobulin E (IgE) plays a crucial role in atopy by binding to two types of Fcε receptors (FcεRI and FcεRII, also known as CD23). The cross-linking of FcεRI-bound IgE on effector cells, such as basophils and mast cells, initiates the allergic response. Conversely, the binding of IgE to CD23 modulates IgE serum levels and antigen presentation. In addition to binding to FcεRs, IgE can also interact with other receptors, such as certain galectins and, in mice, some FcγRs. The binding strength of IgE to its receptors is affected by its valency and glycosylation. While FcεRI shows reduced binding to IgE immune complexes (IgE-ICs), the binding to CD23 is enhanced. There is no evidence that galectins bind IgE-ICs. On the other hand, IgE glycosylation plays a crucial role in the binding to FcεRI and galectins, whereas the binding to CD23 seems to be independent of glycosylation. In this review, we will focus on receptors that bind to IgE and examine how the glycosylation and complexation of IgE impact their binding.
Collapse
Affiliation(s)
- Kevin Plattner
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Correspondence: Monique Vogel
| |
Collapse
|
5
|
Plattner K, Gharailoo Z, Zinkhan S, Engeroff P, Bachmann MF, Vogel M. IgE glycans promote anti-IgE IgG autoantibodies that facilitate IgE serum clearance via Fc Receptors. Front Immunol 2022; 13:1069100. [PMID: 36544773 PMCID: PMC9761184 DOI: 10.3389/fimmu.2022.1069100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Background Recent studies have shown that IgE glycosylation significantly impacts the ability of IgE to bind to its high-affinity receptor FcεRI and exert effector functions. We have recently demonstrated that immunizing mice with IgE in a complex with an allergen leads to a protective, glycan-dependent anti-IgE response. However, to what extent the glycans on IgE determine the induction of those antibodies and how they facilitate serum clearance is unclear.Therefore, we investigated the role of glycan-specific anti-IgE IgG autoantibodies in regulating serum IgE levels and preventing systemic anaphylaxis by passive immunization. Methods Mice were immunized using glycosylated or deglycosylated IgE-allergen-immune complexes (ICs) to induce anti-IgE IgG antibodies. The anti-IgE IgG antibodies were purified and used for passive immunization. Results Glycosylated IgE-ICs induced a significantly higher anti-IgE IgG response and more IgG-secreting plasma cells than deglycosylated IgE-ICs. Passive immunization of IgE-sensitized mice with purified anti-IgE IgG increased the clearance of IgE and prevented systemic anaphylaxis upon allergen challenge. Anti-IgE IgG purified from the serum of mice immunized with deglycosylated IgE-ICs, led to a significantly reduced elimination and protection, confirming that the IgE glycans themselves are the primary drivers of the protectivity induced by the IgE-immune complexes. Conclusion IgE glycosylation is essential for a robust anti-IgE IgG response and might be an important regulator of serum IgE levels.
Collapse
Affiliation(s)
- Kevin Plattner
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland,Department of Biomedical Research (DBMR), University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
| | - Zahra Gharailoo
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland,Department of Biomedical Research (DBMR), University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
| | - Simon Zinkhan
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland,Department of Biomedical Research (DBMR), University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
| | - Paul Engeroff
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland,Department of Biomedical Research (DBMR), University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland,Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Immunology, University Hospital for Rheumatology and Immunology, Bern, Switzerland,Department of Biomedical Research (DBMR), University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland,*Correspondence: Monique Vogel,
| |
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
|
Caiazzo E, Cerqua I, Turiello R, Riemma MA, De Palma G, Ialenti A, Roviezzo F, Morello S, Cicala C. Lack of Ecto-5′-Nucleotidase Protects Sensitized Mice against Allergen Challenge. Biomolecules 2022; 12:biom12050697. [PMID: 35625624 PMCID: PMC9139122 DOI: 10.3390/biom12050697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
Ecto-5′-nucleotidase (CD73), the ectoenzyme that together with CD39 is responsible for extracellular ATP hydrolysis and adenosine accumulation, regulates immune/inflammatory processes by controlling innate and acquired immunity cell functions. We previously demonstrated that CD73 is required for the assessment of a controlled allergic sensitization, in mice. Here, we evaluated the response to aerosolized allergen of female-sensitized mice lacking CD73 in comparison with their wild type counterpart. Results obtained show, in mice lacking CD73, the absence of airway hyperreactivity in response to an allergen challenge, paralleled by reduced airway CD23+B cells and IL4+T cells pulmonary accumulation together with reduced mast cells accumulation and degranulation. Our findings indicate CD73 as a potential therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Roberta Turiello
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (R.T.); (S.M.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy
| | - Maria Antonietta Riemma
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Giacomo De Palma
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Armando Ialenti
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Fiorentina Roviezzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (R.T.); (S.M.)
| | - Carla Cicala
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.C.); (I.C.); (M.A.R.); (G.D.P.); (A.I.); (F.R.)
- Correspondence:
| |
Collapse
|
8
|
Zhang Y, Liu X, Zhao J, Wang J, Song Q, Zhao C. The phagocytic receptors of β-glucan. Int J Biol Macromol 2022; 205:430-441. [PMID: 35202631 DOI: 10.1016/j.ijbiomac.2022.02.111] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/02/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022]
Abstract
Phagocytosis is a cellular process maintaining tissue balance and plays an essential role in initiating the innate immune response. The process of phagocytosis was triggered by the binding of pathogen-associated molecular patterns (PAMP) with their cell surface receptors on the phagocytes. These receptors not only perform phagocytic functions, but also bridge the gap between extracellular and intracellular communication, leading to signal transduction and the production of inflammatory mediators, which are crucial for clearing the invading pathogens and maintaining cell homeostasis. For the past few years, the application of β-glucan comes down to immunoregulation and anti-tumor territory. As a well-known PAMP, β-glucan is one of the most abundant polysaccharides in nature. By binding to specific receptors on immune cells and activating intracellular signal transduction pathways, it causes phagocytosis and promotes the release of cytokines. Further retrieval and straightening out literature related to β-glucan phagocytic receptors will help better elucidate their immunomodulatory functions. This review attempts to summarize physicochemical properties and specific processes involved in β-glucan induced phagocytosis, its phagocytic receptors, and cascade events triggered by β-glucan at the cellular and molecular levels.
Collapse
Affiliation(s)
- Yazhuo Zhang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China
| | - Xinning Liu
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China
| | - Jun Zhao
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China
| | - Jie Wang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China
| | - Qiaoling Song
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, Shandong 266071, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China.
| |
Collapse
|
9
|
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
|
10
|
Duan S, Arlian BM, Nycholat CM, Wei Y, Tateno H, Smith SA, Macauley MS, Zhu Z, Bochner BS, Paulson JC. Nanoparticles Displaying Allergen and Siglec-8 Ligands Suppress IgE-FcεRI-Mediated Anaphylaxis and Desensitize Mast Cells to Subsequent Antigen Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2290-2300. [PMID: 33911007 PMCID: PMC8113104 DOI: 10.4049/jimmunol.1901212] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/15/2021] [Indexed: 01/01/2023]
Abstract
Siglec-8 is an inhibitory receptor expressed on eosinophils and mast cells. In this study, we took advantage of a novel Siglec-8 transgenic mouse model to assess the impact of modulating IgE-dependent mast cell degranulation and anaphylaxis using a liposomal platform to display an allergen with or without a synthetic glycan ligand for Siglec-8 (Sig8L). The hypothesis is that recruitment of Siglec-8 to the IgE-FcεRI receptor complex will inhibit allergen-induced mast cell degranulation. Codisplay of both allergen and Sig8L on liposomes profoundly suppresses IgE-mediated degranulation of mouse bone marrow-derived mast cells or rat basophilic leukemia cells expressing Siglec-8. In contrast, liposomes displaying only Sig8L have no significant suppression of antigenic liposome-induced degranulation, demonstrating that the inhibitory activity by Siglec-8 occurs only when Ag and Sig8L are on the same particle. In mouse models of anaphylaxis, display of Sig8L on antigenic liposomes completely suppresses IgE-mediated anaphylaxis in transgenic mice with mast cells expressing Siglec-8 but has no protection in mice that do not express Siglec-8. Furthermore, mice protected from anaphylaxis remain desensitized to subsequent allergen challenge because of loss of Ag-specific IgE from the cell surface and accelerated clearance of IgE from the blood. Thus, although expression of human Siglec-8 on murine mast cells does not by itself modulate IgE-FcεRI-mediated cell activation, the enforced recruitment of Siglec-8 to the FcεRI receptor by Sig8L-decorated antigenic liposomes results in inhibition of degranulation and desensitization to subsequent Ag exposure.
Collapse
MESH Headings
- Allergens/administration & dosage
- Anaphylaxis/drug therapy
- Anaphylaxis/genetics
- Anaphylaxis/immunology
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Cell Degranulation/drug effects
- Cell Degranulation/genetics
- Cell Degranulation/immunology
- Cell Line, Tumor
- Desensitization, Immunologic/methods
- Disease Models, Animal
- Drug Delivery Systems/methods
- Humans
- Immunoglobulin E/metabolism
- Lectins/genetics
- Lectins/metabolism
- Ligands
- Liposomes
- Mast Cells/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nanoparticles/chemistry
- Polysaccharides/administration & dosage
- Polysaccharides/metabolism
- Rats
- Receptors, IgE/genetics
- Receptors, IgE/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Shiteng Duan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Yadong Wei
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hiroaki Tateno
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Scott A Smith
- Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Zhou Zhu
- Department of Pediatrics, Brown University Alpert Medical School, Providence, RI
| | - Bruce S Bochner
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
11
|
Engeroff P, Plattner K, Storni F, Thoms F, Frias Boligan K, Muerner L, Eggel A, von Gunten S, Bachmann MF, Vogel M. Glycan-specific IgG anti-IgE autoantibodies are protective against allergic anaphylaxis in a murine model. J Allergy Clin Immunol 2021; 147:1430-1441. [PMID: 33309740 DOI: 10.1016/j.jaci.2020.11.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 09/23/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND IgE causes anaphylaxis in type I hypersensitivity diseases by activating degranulation of effector cells such as mast cells and basophils. The mechanisms that control IgE activity and prevent anaphylaxis under normal conditions are still enigmatic. OBJECTIVE We aimed to unravel how anti-IgE autoantibodies are induced and we aimed to understand their role in regulating serum IgE level and allergic anaphylaxis. METHODS We immunized mice with different forms of IgE and tested anti-IgE autoantibody responses and their specificities. We then analyzed the effect of those antibodies on serum kinetics and their in vitro and in vivo impact on anaphylaxis. Finally, we investigated anti-IgE autoantibodies in human sera. RESULTS Immunization of mice with IgE-immune complexes induced glycan-specific anti-IgE autoantibodies. The anti-IgE autoantibodies prevented effector cell sensitization, reduced total IgE serum levels, protected mice from passive and active IgE sensitization, and resulted in cross-protection against different allergens. Furthermore, glycan-specific anti-IgE autoantibodies were present in sera from subjects with allergy and subjects without allergy. CONCLUSION In conclusion, this study provided the first evidence that in the murine model, the serum level and anaphylactic activity of IgE may be downregulated by glycan-specific IgG anti-IgE autoantibodies.
Collapse
Affiliation(s)
- Paul Engeroff
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Kevin Plattner
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Federico Storni
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Franziska Thoms
- Department of Dermatology, Zurich University Hospital, Zurich, Switzerland
| | | | - Lukas Muerner
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Alexander Eggel
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Martin F Bachmann
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- University Hospital for Rheumatology, Immunology, and Allergology, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
12
|
SoRelle JA, Chen Z, Wang J, Yue T, Choi JH, Wang K, Zhong X, Hildebrand S, Russell J, Scott L, Xu D, Zhan X, Bu CH, Wang T, Choi M, Tang M, Ludwig S, Zhan X, Li X, Moresco EMY, Beutler B. Dominant atopy risk mutations identified by mouse forward genetic analysis. Allergy 2021; 76:1095-1108. [PMID: 32810290 DOI: 10.1111/all.14564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/01/2020] [Accepted: 06/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Atopy, the overall tendency to become sensitized to an allergen, is heritable but seldom ascribed to mutations within specific genes. Atopic individuals develop abnormally elevated IgE responses to immunization with potential allergens. To gain insight into the genetic causes of atopy, we carried out a forward genetic screen for atopy in mice. METHODS We screened mice carrying homozygous and heterozygous N-ethyl-N-nitrosourea (ENU)-induced germline mutations for aberrant antigen-specific IgE and IgG1 production in response to immunization with the model allergen papain. Candidate genes were validated by independent gene mutation. RESULTS Of 31 candidate genes selected for investigation, the effects of mutations in 23 genes on papain-specific IgE or IgG1 were verified. Among the 20 verified genes influencing the IgE response, eight were necessary for the response, while 12 repressed IgE. Nine genes were not previously implicated in the IgE response. Fifteen genes encoded proteins contributing to IgE class switch recombination or B-cell receptor signaling. The precise roles of the five remaining genes (Flcn, Map1lc3b, Me2, Prkd2, and Scarb2) remain to be determined. Loss-of-function mutations in nine of the 12 genes limiting the IgE response were dominant or semi-dominant for the IgE phenotype but did not cause immunodeficiency in the heterozygous state. Using damaging allele frequencies for the corresponding human genes and in silico simulations (Monte Carlo) of undiscovered atopy mutations, we estimated the percentage of humans with heterozygous atopy risk mutations. CONCLUSIONS Up to 37% of individuals may be heterozygous carriers for at least one dominant atopy risk mutation.
Collapse
Affiliation(s)
- Jeffrey A. SoRelle
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
- Department of Pathology University of Texas Southwestern Medical Center Dallas TX USA
| | - Zhe Chen
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Jianhui Wang
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Tao Yue
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Jin Huk Choi
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
- Department of Immunology University of Texas Southwestern Medical Center Dallas TX USA
| | - Kuan‐wen Wang
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Xue Zhong
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Sara Hildebrand
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Jamie Russell
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Lindsay Scott
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Darui Xu
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Xiaowei Zhan
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Chun Hui Bu
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Tao Wang
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
- Department of Population and Data Sciences Quantitative Biomedical Research Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Mihwa Choi
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Miao Tang
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Sara Ludwig
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Xiaoming Zhan
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Eva Marie Y. Moresco
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense University of Texas Southwestern Medical Center Dallas TX USA
| |
Collapse
|
13
|
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
|
14
|
The mechanistic and functional profile of the therapeutic anti-IgE antibody ligelizumab differs from omalizumab. Nat Commun 2020; 11:165. [PMID: 31913280 PMCID: PMC6949303 DOI: 10.1038/s41467-019-13815-w] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/30/2019] [Indexed: 01/21/2023] Open
Abstract
Targeting of immunoglobulin E (IgE) represents an interesting approach for the treatment of allergic disorders. A high-affinity monoclonal anti-IgE antibody, ligelizumab, has recently been developed to overcome some of the limitations associated with the clinical use of the therapeutic anti-IgE antibody, omalizumab. Here, we determine the molecular binding profile and functional modes-of-action of ligelizumab. We solve the crystal structure of ligelizumab bound to IgE, and report epitope differences between ligelizumab and omalizumab that contribute to their qualitatively distinct IgE-receptor inhibition profiles. While ligelizumab shows superior inhibition of IgE binding to FcεRI, basophil activation, IgE production by B cells and passive systemic anaphylaxis in an in vivo mouse model, ligelizumab is less potent in inhibiting IgE:CD23 interactions than omalizumab. Our data thus provide a structural and mechanistic foundation for understanding the efficient suppression of FcεRI-dependent allergic reactions by ligelizumab in vitro as well as in vivo. Immunoglobulin E (IgE) plays a central role in allergic responses, yet therapeutic targeting of IgE with antibodies such as omalizumab is met with various limitations. Here the authors characterize the molecular properties and crystal structure of a new anti-IgE antibody, ligelizumab, for mechanistic insights related to its enhanced suppression activity.
Collapse
|
15
|
Tracing IgE-Producing Cells in Allergic Patients. Cells 2019; 8:cells8090994. [PMID: 31466324 PMCID: PMC6769703 DOI: 10.3390/cells8090994] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/13/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Immunoglobulin E (IgE) is the key immunoglobulin in the pathogenesis of IgE associated allergic diseases affecting 30% of the world population. Recent data suggest that allergen-specific IgE levels in serum of allergic patients are sustained by two different mechanisms: inducible IgE production through allergen exposure, and continuous IgE production occurring even in the absence of allergen stimulus that maintains IgE levels. This assumption is supported by two observations. First, allergen exposure induces transient increases of systemic IgE production. Second, reduction in IgE levels upon depletion of IgE from the blood of allergic patients using immunoapheresis is only temporary and IgE levels quickly return to pre-treatment levels even in the absence of allergen exposure. Though IgE production has been observed in the peripheral blood and locally in various human tissues (e.g., nose, lung, spleen, bone marrow), the origin and main sites of IgE production in humans remain unknown. Furthermore, IgE-producing cells in humans have yet to be fully characterized. Capturing IgE-producing cells is challenging not only because current staining technologies are inadequate, but also because the cells are rare, they are difficult to discriminate from cells bearing IgE bound to IgE-receptors, and plasma cells express little IgE on their surface. However, due to the central role in mediating both the early and late phases of allergy, free IgE, IgE-bearing effector cells and IgE-producing cells are important therapeutic targets. Here, we discuss current knowledge and unanswered questions regarding IgE production in allergic patients as well as possible therapeutic approaches targeting IgE.
Collapse
|
16
|
Engeroff P, Caviezel F, Mueller D, Thoms F, Bachmann MF, Vogel M. CD23 provides a noninflammatory pathway for IgE-allergen complexes. J Allergy Clin Immunol 2019; 145:301-311.e4. [PMID: 31437490 DOI: 10.1016/j.jaci.2019.07.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/21/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Type I hypersensitivity is mediated by allergen-specific IgE, which sensitizes the high-affinity IgE receptor FcεRI on mast cells and basophils and drives allergic inflammation upon secondary allergen contact. CD23/FcεRII, the low-affinity receptor for IgE, is constitutively expressed on B cells and has been shown to regulate immune responses. Simultaneous binding of IgE to FcεRI and CD23 is blocked by reciprocal allosteric inhibition, suggesting that the 2 receptors exert distinct roles in IgE handling. OBJECTIVE We aimed to study how free IgE versus precomplexed IgE-allergen immune complexes (IgE-ICs) target the 2 IgE receptors FcεRI and CD23, and we investigated the functional implications of the 2 pathways. METHODS We performed binding and activation assays with human cells in vitro and IgE pharmacokinetics and anaphylaxis experiments in vivo. RESULTS We demonstrate that FcεRI preferentially binds free IgE and CD23 preferentially binds IgE-ICs. We further show that those different binding properties directly translate to distinct biological functions: free IgE initiated allergic inflammation through FcεRI on allergic effector cells, while IgE-ICs were noninflammatory because of reduced FcεRI binding and enhanced CD23-dependent serum clearance. CONCLUSION We propose that IgE-ICs are noninflammatory through reduced engagement by FcεRI but increased targeting of the CD23 pathway.
Collapse
Affiliation(s)
- Paul Engeroff
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Flurin Caviezel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - David Mueller
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Franziska Thoms
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland
| | - Martin F Bachmann
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Chen X, Cai C, Xu D, Liu Q, Zheng S, Liu L, Li G, Zhang X, Li X, Ma Y, Huang L, Chen J, Shi J, Du X, Xia W, Xiang AP, Peng Y. Human Mesenchymal Stem Cell-Treated Regulatory CD23 +CD43 + B Cells Alleviate Intestinal Inflammation. Am J Cancer Res 2019; 9:4633-4647. [PMID: 31367246 PMCID: PMC6643430 DOI: 10.7150/thno.32260] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/21/2019] [Indexed: 01/06/2023] Open
Abstract
Rationale: Mesenchymal stem cells (MSCs) have been demonstrated to ameliorate inflammatory bowel disease by their actions on multiple immune cells, especially on regulatory B cells (Breg cells). However, the phenotypes and functions of human MSCs (hMSCs)-treated Breg cell subsets are not yet clear. Methods: Purified B cells were cocultured with MSCs and the phenotypes and immunomodulatory functions of the B cells were analyzed by FACS and proliferation assays in vitro. Also, a trinitrobenzenesulfonic acid-induced mouse colitis model was employed to detect the function of MSC-treated Breg cells in vivo. Results: We demonstrated that coculturing with hMSCs significantly enhanced the immunomodulatory activity of B cells by up-regulating IL-10 expression. We then identified that a novel regulatory B cell population characterized by CD23 and CD43 phenotypic markers could be induced by hMSCs. The CD23+CD43+ Breg cells substantially inhibited the inflammatory cytokine secretion and proliferation of T cells through an IL-10-dependent pathway. More significantly, intraperitoneal injection of hMSCs ameliorated the clinical and histopathological severity in the mouse experimental colitis model, accompanied by an increase in the number of CD23+CD43+ Breg cells. The adoptive transfer of CD23+CD43+ B cells effectively alleviated murine colitis, as compared with the CD23-CD43- B cells. Treatment with CD23+CD43+ B cells, and not hMSCs, substantially improved the symptoms of colitis in B cell-depleted mice. Conclusion: the novel CD23+CD43+ Breg cell subset appears to be involved in the immunomodulatory function of hMSCs and sheds new light on elucidating the therapeutic mechanism of hMSCs for the treatment of inflammation-related diseases.
Collapse
|
18
|
Yang Z, Jung JB, Allen CDC. Study of IgE-Producing B Cells Using the Verigem Fluorescent Reporter Mouse. Methods Mol Biol 2019; 1799:247-264. [PMID: 29956157 DOI: 10.1007/978-1-4939-7896-0_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunoglobulin E (IgE) is the least abundant antibody isotype in mammalians, yet it plays a critical role in allergy and asthma. IgE-producing (IgE+) B cells are rare and difficult to detect, which have hindered progress to understand their generation and differentiation. Recently developed new fluorescent IgE reporter mice have enabled better understanding of the biology of IgE+ B cells. We here describe the usage of the Verigem IgE reporter mouse to study IgE+ B cells and plasma cells by flow cytometry and microscopy.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.,Sandler Asthma Basic Research Center, University of California, San Francisco, CA, USA
| | - James B Jung
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.,Sandler Asthma Basic Research Center, University of California, San Francisco, CA, USA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA. .,Sandler Asthma Basic Research Center, University of California, San Francisco, CA, USA. .,Department of Anatomy, University of California, San Francisco, CA, USA.
| |
Collapse
|
19
|
Duan S, Koziol-White CJ, Jester WF, Smith SA, Nycholat CM, Macauley MS, Panettieri RA, Paulson JC. CD33 recruitment inhibits IgE-mediated anaphylaxis and desensitizes mast cells to allergen. J Clin Invest 2019; 129:1387-1401. [PMID: 30645205 DOI: 10.1172/jci125456] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Allergen immunotherapy for patients with allergies begins with weekly escalating doses of allergen under medical supervision to monitor and treat IgE mast cell-mediated anaphylaxis. There is currently no treatment to safely desensitize mast cells to enable robust allergen immunotherapy with therapeutic levels of allergen. Here, we demonstrated that liposomal nanoparticles bearing an allergen and a high-affinity glycan ligand of the inhibitory receptor CD33 profoundly suppressed IgE-mediated activation of mast cells, prevented anaphylaxis in Tg mice with mast cells expressing human CD33, and desensitized mice to subsequent allergen challenge for several days. We showed that high levels of CD33 were consistently expressed on human skin mast cells and that the antigenic liposomes with CD33 ligand prevented IgE-mediated bronchoconstriction in slices of human lung. The results demonstrated the potential of exploiting CD33 to desensitize mast cells to provide a therapeutic window for administering allergen immunotherapy without triggering anaphylaxis.
Collapse
Affiliation(s)
- Shiteng Duan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - William F Jester
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - Scott A Smith
- Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
20
|
Guo Y, Chang Q, Cheng L, Xiong S, Jia X, Lin X, Zhao X. C-Type Lectin Receptor CD23 Is Required for Host Defense against Candida albicans and Aspergillus fumigatus Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:2427-2440. [DOI: 10.4049/jimmunol.1800620] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
|
21
|
Chen JB, Ramadani F, Pang MOY, Beavil RL, Holdom MD, Mitropoulou AN, Beavil AJ, Gould HJ, Chang TW, Sutton BJ, McDonnell JM, Davies AM. Structural basis for selective inhibition of immunoglobulin E-receptor interactions by an anti-IgE antibody. Sci Rep 2018; 8:11548. [PMID: 30069035 PMCID: PMC6070508 DOI: 10.1038/s41598-018-29664-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/16/2018] [Indexed: 11/09/2022] Open
Abstract
Immunoglobulin E (IgE) antibodies play a central role in the allergic response: interaction with FcεRI on mast cells and basophils leads to immediate hypersensitivity reactions upon allergen challenge, while interaction with CD23/FcεRII, expressed on a variety of cells, regulates IgE synthesis among other activities. The receptor-binding IgE-Fc region has recently been found to display remarkable flexibility, from acutely bent to extended conformations, with allosteric communication between the distant FcεRI and CD23 binding sites. We report the structure of an anti-IgE antibody Fab (8D6) bound to IgE-Fc through a mixed protein-carbohydrate epitope, revealing further flexibility and a novel extended conformation with potential relevance to that of membrane-bound IgE in the B cell receptor for antigen. Unlike the earlier, clinically approved anti-IgE antibody omalizumab, 8D6 inhibits binding to FcεRI but not CD23; the structure reveals how this discrimination is achieved through both orthosteric and allosteric mechanisms, supporting therapeutic strategies that retain the benefits of CD23 binding.
Collapse
Affiliation(s)
- Jiun-Bo Chen
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
| | - Faruk Ramadani
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Marie O Y Pang
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Rebecca L Beavil
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma Protein Production Facility, London, United Kingdom
| | - Mary D Holdom
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Alkistis N Mitropoulou
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Andrew J Beavil
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Hannah J Gould
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Tse Wen Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Brian J Sutton
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom.
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| | - James M McDonnell
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom.
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| | - Anna M Davies
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, United Kingdom.
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| |
Collapse
|
22
|
Balbino B, Conde E, Marichal T, Starkl P, Reber LL. Approaches to target IgE antibodies in allergic diseases. Pharmacol Ther 2018; 191:50-64. [PMID: 29909239 DOI: 10.1016/j.pharmthera.2018.05.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
IgE is the antibody isotype found at the lowest concentration in the circulation. However IgE can undeniably play an important role in mediating allergic reactions; best exemplified by the clinical benefits of anti-IgE monoclonal antibody (omalizumab) therapy for some allergic diseases. This review will describe our current understanding of the interactions between IgE and its main receptors FcεRI and CD23 (FcεRII). We will review the known and potential functions of IgE in health and disease: in particular, its detrimental roles in allergic diseases and chronic spontaneous urticaria, and its protective functions in host defense against parasites and venoms. Finally, we will present an overview of the drugs that are in clinical development or have therapeutic potential for IgE-mediated allergic diseases.
Collapse
Affiliation(s)
- Bianca Balbino
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Eva Conde
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France; Neovacs SA, Paris, France
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000, Liege, Belgium; Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Philipp Starkl
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Austria; Department of Medicine I, Research Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France.
| |
Collapse
|
23
|
Leung NYH, Wai CYY, Shu SA, Chang CC, Chu KH, Leung PSC. Low-Dose Allergen-Specific Immunotherapy Induces Tolerance in a Murine Model of Shrimp Allergy. Int Arch Allergy Immunol 2017; 174:86-96. [PMID: 29065408 DOI: 10.1159/000479694] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The efficacy and safety of allergen-specific immunotherapy (AIT) are highly dose-dependent. METHODS We investigated the dosage effects of AIT and the underlying mechanisms in a murine model of shrimp hypersensitivity. BALB/c mice were sensitized with recombinant shrimp allergen rMet e 1 and challenged orally with a high dose of rMet e 1 to elicit an allergic response. These sensitized mice were then treated with a low (0.01 mg), medium (0.05 mg), or high dosage (0.1 mg) of rMet e 1 intraperitoneally before receiving a second oral challenge. The allergic responses and immunological changes in the gut were compared between animals receiving different dosages. RESULTS We found that all sensitized mice that received rMet e 1 immunotherapy were desensitized, regardless of the dosage, and protected at the second oral challenge. Nevertheless, the mice in the high-dosage group experienced severe systemic reactions during the treatment phase. In contrast, regulatory T (Treg) cell-associated genes were upregulated only in the low- and medium-dosage groups, and Foxp3+ cells were more abundant in the gut lymphoid tissues than in the high-dosage group. CONCLUSIONS Our results demonstrate that low-dosage immunotherapy favors the induction of local Foxp3+ Treg cells and the upregulation of regulatory cytokines. The safety advantages and long-term efficacy of low-dosage immunotherapy should be taken into consideration when developing immunotherapy dose schedules.
Collapse
Affiliation(s)
- Nicki Yat Hin Leung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | | | | | | | | | | |
Collapse
|
24
|
Lawrence MG, Woodfolk JA, Schuyler AJ, Stillman LC, Chapman MD, Platts-Mills TAE. Half-life of IgE in serum and skin: Consequences for anti-IgE therapy in patients with allergic disease. J Allergy Clin Immunol 2017; 139:422-428.e4. [PMID: 27496596 PMCID: PMC5405770 DOI: 10.1016/j.jaci.2016.04.056] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 04/13/2016] [Accepted: 04/29/2016] [Indexed: 01/08/2023]
Abstract
We present results from clinical studies on plasma infusion done in the late 1970s in patients with hypogammaglobulinemia in which we documented the short half-life of both total and allergen-specific IgE in serum. The development of specific allergic sensitization in the skin of those patients followed by the gradual decrease in sensitization over 50 days was also documented. The data are included here along with a discussion of the existing literature about the half-life of IgE in both the circulation and skin. This rostrum reinterprets the earlier clinical studies in light of new insights and mechanisms that could explain the rapid removal of IgE from the circulation. These mechanisms have clinical implications that relate to the increasing use of anti-IgE mAbs for the treatment of allergic disease.
Collapse
Affiliation(s)
- Monica G Lawrence
- University of Virginia Asthma and Allergic Diseases Center, Charlottesville, Va
| | - Judith A Woodfolk
- University of Virginia Asthma and Allergic Diseases Center, Charlottesville, Va
| | | | | | | | | |
Collapse
|
25
|
Tsiantoulas D, Bot I, Ozsvar-Kozma M, Göderle L, Perkmann T, Hartvigsen K, Conrad DH, Kuiper J, Mallat Z, Binder CJ. Increased Plasma IgE Accelerate Atherosclerosis in Secreted IgM Deficiency. Circ Res 2016; 120:78-84. [PMID: 27903567 DOI: 10.1161/circresaha.116.309606] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/23/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022]
Abstract
RATIONALE Deficiency of secreted IgM (sIgM-/-) accelerates atherosclerosis in Ldlr-/-mice. Several atheroprotective effects of increased levels of IgM antibodies have been suggested, including preventing inflammation induced by oxidized low-density lipoprotein and promoting apoptotic cell clearance. However, the mechanisms by which the lack of sIgM promotes lesion formation remain unknown. OBJECTIVE To identify the mechanisms by which sIgM deficiency accelerates atherosclerosis in mice. METHODS AND RESULTS We here show that both sIgM-/- and Ldlr-/-sIgM-/- mice develop increased plasma IgE titers because of impaired generation of B cells expressing the low-affinity IgE receptor CD23, which mediates the clearance of IgE antibodies. We further report that Ldlr-/-sIgM-/- mice exhibit increased numbers of activated mast cells and neutrophils in the perivascular area of atherosclerotic plaques. Treatment with an anti-IgE-neutralizing antibody fully reversed vascular inflammation and accelerated atherosclerotic lesion formation in cholesterol-fed Ldlr-/-sIgM-/- mice. CONCLUSIONS Thus, our data identify a previously unsuspected mechanism by which sIgM deficiency aggravates atherosclerosis.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Ilze Bot
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Maria Ozsvar-Kozma
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Laura Göderle
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Thomas Perkmann
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Karsten Hartvigsen
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Daniel H Conrad
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Johan Kuiper
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Ziad Mallat
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.)
| | - Christoph J Binder
- From the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (D.T., M.O.-K., L.G., K.H., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., M.O.-K., L.G., T.P., K.H., C.J.B.); Division of Biopharmaceutics, LACDR Leiden University, The Netherlands (I.B., J.K.); Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond (D.H.C.); and Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (Z.M.).
| |
Collapse
|
26
|
Kelly BT, Grayson MH. Immunoglobulin E, what is it good for? Ann Allergy Asthma Immunol 2016; 116:183-7. [PMID: 26945494 DOI: 10.1016/j.anai.2015.10.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 12/18/2022]
|
27
|
Yamaki K, Yoshino S. Establishment of a Mouse Anti-ovalbumin IgE Monoclonal Antibody That Induces FcɛRII (CD23)-dependent Activation Without FcɛRI-Dependent Activation. Monoclon Antib Immunodiagn Immunother 2015; 34:423-31. [PMID: 26683182 DOI: 10.1089/mab.2015.0032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IgE mainly activates cells via two receptors, FcɛRI and FcɛRII. Blocking antibodies against and animals genetically targeted for these receptors have been successfully used to distinguish between these two activating pathways. In the present study, we investigated whether our newly established anti-ovalbumin (OVA) monoclonal IgE OE-2 induced FcɛRII-dependent activation, but not FcɛRI-dependent activation in vivo and in vitro, in contrast to the previously established anti-OVA IgE OE-1, which stimulated FcɛRI and FcɛRII. The FcɛRI-mediated degranulation of RBL2H3 cells and passive systemic anaphylaxis in mice were induced by OE-1 but not OE-2. On the other hand, the production of nitric oxide by rat peritoneal macrophages and the primary antibody response in mice against co-injected OVA, which were mediated through FcɛRII, were induced and enhanced by OE-1 and OE-2. Differences in the epitopes recognized by OE-1 and OE-2 may partially explain why OE-1, but not OE-2, triggered FcɛRI-dependent activation. OE-1 bridged FcɛRI through effective aggregation with OVA, whereas OE-2 crosslinked the receptor strongly and only moderately upon the addition of an anti-kappa antibody and polymerized OVA, namely, an OVA-conjugated resin, respectively, resulting in degranulation. Our results offer a novel approach for determining the relative importance of FcɛRI and FcɛRII in various IgE-dependent responses by using OE-1 and OE-2.
Collapse
Affiliation(s)
- Kouya Yamaki
- Department of Pharmacology, Kobe Pharmaceutical University , Kobe, Hyogo, Japan
| | - Shin Yoshino
- Department of Pharmacology, Kobe Pharmaceutical University , Kobe, Hyogo, Japan
| |
Collapse
|
28
|
Abstract
Immunoglobulin E (IgE) antibodies play a crucial role in host defense against parasite infections. However, inappropriate IgE responses are also involved in the pathogenesis of allergic diseases. The generation of IgE antibodies is a tightly controlled process regulated by multiple transcription factors, cytokines, and immune cells including γδ T cells. Accumulating evidence demonstrates that γδ T cells play a critical role in regulating IgE responses; however, both IgE-enhancing and IgE-suppressive effects are suggested for these cells in different experimental systems. In this review, we examine the available evidence and discuss the role of γδ T cells in IgE regulation both in the context of antigen-induced immune responses and in the state of partial immunodeficiency.
Collapse
|
29
|
Evans H, Killoran KE, Mitre BK, Morris CP, Kim SY, Mitre E. Ten Weeks of Infection with a Tissue-Invasive Helminth Protects against Local Immune Complex-Mediated Inflammation, but Not Cutaneous Type I Hypersensitivity, in Previously Sensitized Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:2973-84. [PMID: 26324775 DOI: 10.4049/jimmunol.1500081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/29/2015] [Indexed: 11/19/2022]
Abstract
In this study, we evaluated the effect chronic helminth infection has on allergic disease in mice previously sensitized to OVA. Ten weeks of infection with Litomosoides sigmodontis reduced immunological markers of type I hypersensitivity, including OVA-specific IgE, basophil activation, and mast cell degranulation. Despite these reductions, there was no protection against immediate clinical hypersensitivity following intradermal OVA challenge. However, late-phase ear swelling, due to type III hypersensitivity, was significantly reduced in chronically infected animals. Levels of total IgG2a, OVA-specific IgG2a, and OVA-specific IgG1 were reduced in the setting of infection. These reductions were most likely due to increased Ab catabolism as ELISPOT assays demonstrated that infected animals do not have suppressed Ab production. Ear histology 24 h after challenge showed infected animals have reduced cellular infiltration in the ear, with significant decreases in numbers of neutrophils and macrophages. Consistent with this, infected animals had less neutrophil-specific chemokines CXCL-1 and CXCL-2 in the ear following challenge. Additionally, in vitro stimulation with immune complexes resulted in significantly less CXCL-1 and CXCL-2 production by eosinophils from chronically infected mice. Expression of FcγRI was also significantly reduced on eosinophils from infected animals. These data indicate that chronic filarial infection suppresses eosinophilic responses to Ab-mediated activation and has the potential to be used as a therapeutic for pre-existing hypersensitivity diseases.
Collapse
Affiliation(s)
- Holly Evans
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
| | | | - Blima K Mitre
- Department of Pathology, University of Pittsburgh Medical Center Passavant Hospital, Pittsburgh, PA 15213; and
| | - C Paul Morris
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - So-Young Kim
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814;
| |
Collapse
|
30
|
Cheng LE, Sullivan BM, Retana LE, Allen CDC, Liang HE, Locksley RM. IgE-activated basophils regulate eosinophil tissue entry by modulating endothelial function. ACTA ACUST UNITED AC 2015; 212:513-24. [PMID: 25779634 PMCID: PMC4387286 DOI: 10.1084/jem.20141671] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/11/2015] [Indexed: 01/22/2023]
Abstract
Basophils orchestrate eosinophil recruitment during IgE-dependent dermatitis by interacting with inflamed endothelium and producing IL-4. IL-4 in turn induces endothelial VCAM-1 expression, which is required for subsequent eosinophil accumulation. Vertebrate immunity has evolved a modular architecture in response to perturbations. Allergic inflammation represents such a module, with signature features of antigen-specific IgE and tissue eosinophilia, although the cellular and molecular circuitry coupling these responses remains unclear. Here, we use genetic and imaging approaches in models of IgE-dependent eosinophilic dermatitis to demonstrate a requisite role for basophils. After antigenic inflammation, basophils initiate transmigration like other granulocytes but, upon activation via their high-affinity IgE receptor, alter their migratory kinetics to persist at the endothelium. Prolonged basophil–endothelial interactions, in part dependent on activation of focal adhesion kinases, promote delivery of basophil-derived IL-4 to the endothelium and subsequent induction of endothelial vascular cell adhesion molecule-1 (VCAM-1), which is required for eosinophil accumulation. Thus, basophils are gatekeepers that link adaptive immunity with innate effector programs by altering access to tissue sites by activation-induced interactions with the endothelium.
Collapse
Affiliation(s)
- Laurence E Cheng
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Brandon M Sullivan
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Lizett E Retana
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Christopher D C Allen
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Hong-Erh Liang
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Richard M Locksley
- Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143 Department of Pediatrics, Department of Medicine, Department of Anatomy, Sandler Asthma Basic Research Center, Cardiovascular Research Institute, Department of Microbiology and Immunology, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
31
|
Roviezzo F, Sorrentino R, Bertolino A, De Gruttola L, Terlizzi M, Pinto A, Napolitano M, Castello G, D'Agostino B, Ianaro A, Sorrentino R, Cirino G. S1P-induced airway smooth muscle hyperresponsiveness and lung inflammation in vivo: molecular and cellular mechanisms. Br J Pharmacol 2015; 172:1882-93. [PMID: 25439580 DOI: 10.1111/bph.13033] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 11/24/2014] [Accepted: 11/27/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine-1-phosphate (S1P) has been shown to be involved in the asthmatic disease as well in preclinical mouse experimental models of this disease. The aim of this study was to understand the mechanism(s) underlying S1P effects on the lung. EXPERIMENTAL APPROACH BALB/c, mast cell-deficient and Nude mice were injected with S1P (s.c.) on days 0 and 7. Functional, molecular and cellular studies were performed. KEY RESULTS S1P administration to BALB/c mice increased airway smooth muscle reactivity, mucus production, PGD2 , IgE, IL-4 and IL-13 release. These features were associated to a higher recruitment of mast cells to the lung. Mast cell-deficient Kit (W) (-sh/) (W) (-sh) mice injected with S1P did not display airway smooth muscle hyper-reactivity. However, lung inflammation and IgE production were still present. Treatment in vivo with the anti-CD23 antibody B3B4, which blocks IgE production, inhibited both S1P-induced airway smooth muscle reactivity in vitro and lung inflammation. S1P administration to Nude mice did not elicit airway smooth muscle hyper-reactivity and lung inflammation. Naïve (untreated) mice subjected to the adoptive transfer of CD4+ T-cells harvested from S1P-treated mice presented all the features elicited by S1P in the lung. CONCLUSIONS AND IMPLICATIONS S1P triggers a cascade of events that sequentially involves T-cells, IgE and mast cells reproducing several asthma-like features. This model may represent a useful tool for defining the role of S1P in the mechanism of action of currently-used drugs as well as in the development of new therapeutic approaches for asthma-like diseases.
Collapse
Affiliation(s)
- F Roviezzo
- Dipartimento di Farmacia, Università di Napoli Federico II, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Evans H, Mitre E. Worms as therapeutic agents for allergy and asthma: understanding why benefits in animal studies have not translated into clinical success. J Allergy Clin Immunol 2014; 135:343-53. [PMID: 25174866 DOI: 10.1016/j.jaci.2014.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 06/04/2014] [Accepted: 07/08/2014] [Indexed: 12/26/2022]
Abstract
Helminth infections are associated with decreased rates of autoimmunity and allergy, and several clinical studies have demonstrated that intentional infection with helminths can reduce symptoms of autoimmune diseases. In contrast, though numerous animal studies have demonstrated that helminth infections ameliorate allergic diseases, clinical trials in humans have not shown benefit. In this article, we review in detail the 2 human studies that have prospectively tested whether helminth infections protect against allergy. We next review the research designs and results obtained from animal studies, and compare these to the human trials. We then postulate possible reasons for the lack of efficacy observed in clinical trials to date and discuss potential future areas of research in this field.
Collapse
Affiliation(s)
- Holly Evans
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Md.
| |
Collapse
|
34
|
Wu LC, Zarrin AA. The production and regulation of IgE by the immune system. Nat Rev Immunol 2014; 14:247-59. [PMID: 24625841 DOI: 10.1038/nri3632] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IgE not only provides protective immunity against helminth parasites but can also mediate the type I hypersensitivity reactions that contribute to the pathogenesis of allergic diseases such as asthma, allergic rhinitis and atopic dermatitis. Despite the importance of IgE in immune biology and allergic pathogenesis, the cells and the pathways that produce and regulate IgE are poorly understood. In this Review, we summarize recent advances in our understanding of the production and the regulation of IgE in vivo, as revealed by studies in mice, and we discuss how these findings compare to what is known about human IgE biology.
Collapse
Affiliation(s)
- Lawren C Wu
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Ali A Zarrin
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| |
Collapse
|
35
|
Lexmond WS, Pardo M, Rooney K, Goettel JA, Snapper SB, Yen EH, Dehlink E, Nurko S, Fiebiger E. Elevated levels of leukotriene C4 synthase mRNA distinguish a subpopulation of eosinophilic oesophagitis patients. Clin Exp Allergy 2014; 43:902-13. [PMID: 23889244 DOI: 10.1111/cea.12146] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/23/2013] [Accepted: 05/03/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cysteinyl leukotrienes contribute to Th2-type inflammatory immune responses. Their levels in oesophageal tissue, however, do not distinguish patients with eosinophilic oesophagitis (EoE) from controls. OBJECTIVE We asked whether mRNA levels of leukotriene C4 synthase (LTC4 S), a key regulator of leukotriene production, could serve as a marker for EoE. METHODS Digital mRNA expression profiling (nCounter(®) Technology) was performed on proximal and distal oesophageal biopsies of 30 paediatric EoE patients and 40 non-EoE controls. Expression data were confirmed with RT-qPCR. LTC4 S mRNA levels were quantified in whole blood samples. Leukotriene E4 was measured in urine. RESULTS LTC4 S mRNA levels were elevated in proximal (2.6-fold, P < 0.001) and distal (2.9-fold, P < 0.001) oesophageal biopsies from EoE patients. Importantly, increased LTC4 S mRNA transcripts identified a subpopulation of EoE patients (28%). This patient subgroup had higher serum IgE levels (669 U/mL vs. 106 U/mL, P = 0.01), higher mRNA transcript numbers of thymic stromal lymphopoietin (TSLP) (1.6-fold, P = 0.009) and CD4 (1.4-fold, P = 0.04) but lower IL-23 mRNA levels (0.5-fold, P = 0.04). In contrast, elevated levels of IL-23 mRNA were found in oesophageal biopsies of patients with reflux oesophagitis. LTC4 S mRNA transcripts in whole blood and urinary excretion of leukotriene E4 were similar in EoE patient subgroups and non-EoE patients. CONCLUSION & CLINICAL RELEVANCE Elevated oesophageal expression of LTC4 S mRNA is found in a subgroup of EoE patients, concomitant with higher serum IgE levels and an oesophageal transcriptome indicative of a more-pronounced allergic phenotype. Together with TSLP and IL-23 mRNA levels, oesophageal LTC4 S mRNA may facilitate diagnosis of an EoE subpopulation for personalized therapy.
Collapse
Affiliation(s)
- W S Lexmond
- Division of Gastroenterology and Nutrition, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Greer AM, Wu N, Putnam AL, Woodruff PG, Wolters P, Kinet JP, Shin JS. Serum IgE clearance is facilitated by human FcεRI internalization. J Clin Invest 2014; 124:1187-98. [PMID: 24569373 DOI: 10.1172/jci68964] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 12/12/2013] [Indexed: 11/17/2022] Open
Abstract
The high-affinity IgE receptor FcεRI is constitutively expressed in mast cells and basophils and is required for transmitting stimulatory signals upon engagement of IgE-bound allergens. FcεRI is also constitutively expressed in dendritic cells (DCs) and monocytes in humans; however, the specific functions of the FcεRI expressed by these cells are not completely understood. Here, we found that FcεRI expressed by human blood DC antigen 1-positive (BDCA1+) DCs and monocytes, but not basophils, traffics to endolysosomal compartments under steady-state conditions. Furthermore, IgE bound to FcεRI on BDCA1+ DCs was rapidly endocytosed, transported to the lysosomes, and degraded in vitro. IgE injected into mice expressing human FcεRIα (FCER1A-Tg mice) was endocytosed by conventional DCs and monocytes, and endocytosis was associated with rapid clearance of circulating IgE from these mice. Importantly, this rapid IgE clearance was dependent on monocytes or DCs but not basophils. These findings strongly suggest that constitutive internalization of human FcεRI by DCs and monocytes distinctively contributes to serum IgE clearance.
Collapse
|
37
|
Nigro E, Siccardi A, Vangelista L. Role and Redirection of IgE against Cancer. Antibodies (Basel) 2013; 2:371-391. [DOI: 10.3390/antib2020371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
IgE is a highly elusive antibody class, yet a tremendously powerful elicitor of immune reactions. Despite huge efforts spent on the characterization and understanding of the IgE system many questions remain either unanswered or only marginally addressed. One above all relates to the role of IgE. A common doubt is based on whether IgE mode of action should only be relegated to anti-parasite immunity and allergic manifestations. In search for a hidden role of IgE, reports from several laboratories are described herein in which a natural IgE link to cancer or the experimental redirection of IgE against cancer have been investigated. Epidemiological and investigational studies are trying to elucidate a possible direct intervention of endogenous IgE against cancer, raising thus far no definitive evidence. Conversely, experimental approaches implementing several strategies and engineered IgE formats built up a series of convincing results indicating that cancer might be tackled by the effector functions of this immunoglobulin class. Because of its peculiar immune features, IgE may present a superior anti-tumor performance as compared to IgG. However, extreme care should be taken on how IgE-based anti-tumor approaches should be devised. Overall, IgE appears as a promising resource, likely destined to enrich the anti-cancer arsenal.
Collapse
Affiliation(s)
- Elisa Nigro
- Molecular Immunology Group, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| | - Antonio Siccardi
- Molecular Immunology Group, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| | - Luca Vangelista
- Protein Engineering and Therapeutics Group, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
38
|
Lübben W, Turqueti-Neves A, Okhrimenko A, Stöberl C, Schmidt V, Pfeffer K, Dehnert S, Wünsche S, Storsberg S, Paul S, Bauer S, Riethmüller G, Voehringer D, Yu P. IgE knock-in mice suggest a role for high levels of IgE in basophil-mediated active systemic anaphylaxis. Eur J Immunol 2013; 43:1231-42. [PMID: 23423996 DOI: 10.1002/eji.201242675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 01/16/2013] [Accepted: 02/14/2013] [Indexed: 01/25/2023]
Abstract
Immunoglobulin E (IgE) production is tightly regulated at the cellular and genetic levels and is believed to be central to allergy development. At least two cellular pathways exist that lead to systemic anaphylaxis reactions in vivo: IgE-sensitized mast cells and IgG1-sensitized basophils. Passive anaphylaxis, by application of allergen and allergen-specific antibodies in mice, indicates a differential contribution of immunoglobulin isotypes to anaphylaxis. However, analysis of a dynamic immunization-mediated antibody response in anaphylaxis is difficult. Here, we generated IgE knock-in mice (IgE(ki) ), which express the IgE heavy chain instead of IgG1, in order to analyze the contribution of IgG1 and IgE to active anaphylaxis in vivo. IgE(ki) mice display increased IgE production both in vitro and in vivo. The sensitization of IgE(ki) mice by immunization followed by antigen challenge leads to increased anaphylaxis. Homozygous IgE(ki) mice, which lack IgG1 due to the knock-in strategy, are most susceptible to active systemic anaphylaxis. The depletion of basophils demonstrates their importance in IgE-mediated anaphylaxis. Therefore, we propose that an enhanced, antigen-specific, polyclonal IgE response, as is the case in allergic patients, is probably the most efficient way to sensitize basophils to contribute to systemic anaphylaxis in vivo.
Collapse
Affiliation(s)
- Wolger Lübben
- Institute for Immunology, Philipps-Universität Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Potaczek DP, Kabesch M. Current concepts of IgE regulation and impact of genetic determinants. Clin Exp Allergy 2013; 42:852-71. [PMID: 22909159 DOI: 10.1111/j.1365-2222.2011.03953.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunoglobulin E (IgE) mediated immune responses seem to be directed against parasites and neoplasms, but are best known for their involvement in allergies. The IgE network is tightly controlled at different levels as outlined in this review. Genetic determinants were suspected to influence IgE regulation and IgE levels considerably for many years. Linkage and candidate gene studies suggested a number of loci and genes to correlate with total serum IgE levels, and recently genome-wide association studies (GWAS) provided the power to identify genetic determinants for total serum IgE levels: 1q23 (FCER1A), 5q31 (RAD50, IL13, IL4), 12q13 (STAT6), 6p21.3 (HLA-DRB1) and 16p12 (IL4R, IL21R). In this review, we analyse the potential role of these GWAS hits in the IgE network and suggest mechanisms of how genes and genetic variants in these loci may influence IgE regulation.
Collapse
Affiliation(s)
- D P Potaczek
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
40
|
Cheng LE, Hartmann K, Roers A, Krummel MF, Locksley RM. Perivascular mast cells dynamically probe cutaneous blood vessels to capture immunoglobulin E. Immunity 2013; 38:166-75. [PMID: 23290520 DOI: 10.1016/j.immuni.2012.09.022] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 09/26/2012] [Indexed: 12/27/2022]
Abstract
Mast cells are tissue-resident immune cells that play a central role in allergic disease. These contributions are largely dependent on the acquisition of antigen-specific immunoglobulin E (IgE). Despite this requirement, studies of mast cell and IgE interactions have overlooked the mechanism by which mast cells acquire IgE from the blood. To address this gap, we developed reporter IgE molecules and employed imaging techniques to study mast cell function in situ. Our data demonstrate that skin mast cells exhibit selective uptake of IgE based on perivascular positioning. Furthermore, perivascular mast cells acquire IgE by extending cell processes across the vessel wall to capture luminal IgE. These data demonstrate how tissue mast cells acquire IgE and reveal a strategy by which extravascular cells monitor blood contents to capture molecules central to cellular function.
Collapse
Affiliation(s)
- Laurence E Cheng
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
41
|
Vaali K, Lappalainen J, Lin AH, Mäyränpää MI, Kovanen PT, Berstad A, Eklund KK. Imatinib mesylate alleviates diarrhea in a mouse model of intestinal allergy. Neurogastroenterol Motil 2012; 24:e325-35. [PMID: 22709239 DOI: 10.1111/j.1365-2982.2012.01941.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND When sensitized epicutaneously and challenged orally with ovalbumin, Balb/c mice develop allergen-induced diarrhea. As mast cells play important roles in diarrhea, we studied whether allergic diarrhea could be alleviated with imatinib mesylate. METHODS Balb/c mice were sensitized and challenged with ovalbumin and treated orally with imatinib. Cytokine mRNA expressions were determined with quantitative RT-PCR and numbers of small intestinal mast cells determined by staining for chloroacetate esterase and mucosal mast cell protease-1. Immunofluorescence staining was used to assess the intestinal CCL1 expression. KEY RESULTS Ovalbumin-sensitized and challenged Balb/c mice developed diarrhea, which was associated with increased number of mast cells and expression of interleukin (IL)-4 and -13, and chemokines CCL1 and CCL17 in the small intestine. Treatment with imatinib reduced the incidence of diarrhea, inhibited the development of mastocytosis and jejunal mRNA expression of IL-13, CCL1, CCL17 and CCL22. Mast cell-deficient W/W(-V) mice, and surprisingly, also their mast cell-competent control (+/+) littermates failed to develop diarrhea as a response to ovalbumin. This strain-dependent difference was associated with the inability of +/+ and W/W(-V) mice to increase the number of intestinal mast cells and expression of IL-4, IL-13, CCL1 and CCL17 after ovalbumin challenge. CONCLUSIONS & INFERENCES Development of allergic diarrhea is associated with the ability of mice to develop intestinal mastocytosis. Imatinib inhibited the development of intestinal mastocytosis, reduced the incidence of diarrhea, and reduced the expression of IL-13, CCL1, and CCL17. Targeting intestinal mast cells could be a feasible approach to treat allergic diarrhea.
Collapse
Affiliation(s)
- K Vaali
- Institute of Medicine, University of Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
42
|
Cooper AM, Hobson PS, Jutton MR, Kao MW, Drung B, Schmidt B, Fear DJ, Beavil AJ, McDonnell JM, Sutton BJ, Gould HJ. Soluble CD23 controls IgE synthesis and homeostasis in human B cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:3199-207. [PMID: 22393152 DOI: 10.4049/jimmunol.1102689] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CD23, the low-affinity receptor for IgE, exists in membrane and soluble forms. Soluble CD23 (sCD23) fragments are released from membrane (m)CD23 by the endogenous metalloprotease a disintegrin and metalloprotease 10. When purified tonsil B cells are incubated with IL-4 and anti-CD40 to induce class switching to IgE in vitro, mCD23 is upregulated, and sCD23 accumulates in the medium prior to IgE synthesis. We have uncoupled the effects of mCD23 cleavage and accumulation of sCD23 on IgE synthesis in this system. We show that small interfering RNA inhibition of CD23 synthesis or inhibition of mCD23 cleavage by an a disintegrin and metalloprotease 10 inhibitor, GI254023X, suppresses IL-4 and anti-CD40-stimulated IgE synthesis. Addition of a recombinant trimeric sCD23 enhances IgE synthesis in this system. This occurs even when endogenous mCD23 is protected from cleavage by GI254023X, indicating that IgE synthesis is positively controlled by sCD23. We show that recombinant trimeric sCD23 binds to cells coexpressing mIgE and mCD21 and caps these proteins on the B cell membrane. Upregulation of IgE by sCD23 occurs after class-switch recombination, and its effects are isotype-specific. These results suggest that mIgE and mCD21 cooperate in the sCD23-mediated positive regulation of IgE synthesis on cells committed to IgE synthesis. Feedback regulation may occur when the concentration of secreted IgE becomes great enough to allow binding to mCD23, thus preventing further release of sCD23. We interpret these results with the aid of a model for the upregulation of IgE by sCD23.
Collapse
Affiliation(s)
- Alison M Cooper
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, London SE1 9RT, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Franko J, Jackson LG, Hubbs A, Kashon M, Meade BJ, Anderson SE. Evaluation of furfuryl alcohol sensitization potential following dermal and pulmonary exposure: enhancement of airway responsiveness. Toxicol Sci 2012; 125:105-15. [PMID: 22003193 PMCID: PMC4699323 DOI: 10.1093/toxsci/kfr271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Furfuryl alcohol is considered by the U.S. Environmental Protection Agency to be a high volume production chemical, with over 1 million pounds produced annually. Due to its high production volume and its numerous industrial and consumer uses, there is considerable potential for work-related exposure, as well as exposure to the general population, through pulmonary, oral, and dermal routes of exposure. Human exposure data report a high incidence of asthma in foundry mold workers exposed to furan resins, suggesting potential immunologic effects. Although furfuryl alcohol was nominated and evaluated for its carcinogenic potential by the National Toxicology Program, studies evaluating its immunotoxicity are lacking. The studies presented here evaluated the immunotoxic potential of furfuryl alcohol following exposure by the dermal and pulmonary routes using a murine model. When tested in a combined irritancy local lymph node assay, furfuryl alcohol was identified to be an irritant and mild sensitizer (EC3 = 25.6%). Pulmonary exposure to 2% furfuryl alcohol resulted in enhanced airway hyperreactivity, eosinophilic infiltration into the lungs, and enhanced cytokine production (IL-4, IL-5, and interferon-γ) by ex vivo stimulated lung-associated draining lymphoid cells. Airway hyperreactivity and eosinophilic lung infiltration were augmented by prior dermal exposure to furfuryl alcohol. These results suggest that furfuryl alcohol may play a role in the development of allergic airway disease and encourage the need for additional investigation.
Collapse
Affiliation(s)
- Jennifer Franko
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| | - Laurel G. Jackson
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| | - Ann Hubbs
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| | - Michael Kashon
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| | - B. J. Meade
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| | - Stacey E. Anderson
- National Institute for Occupational Safety and Health, Health Effects Laboratory Division, Morgantown, West Virginia 26505
| |
Collapse
|
44
|
Abstract
IgE antibodies are involved in allergic reactions. High affinity IgE antibodies can cause anaphylaxis when cross-linked by minute amounts of antigen. The issue of how the IgE response is initiated and maintained is addressed in this review. A model has been proposed by which IgE(+) cells expressing antibodies that bind with high affinity to their antigens are generated through an IgG1 intermediate, which goes through affinity maturation in germinal centers (GC) before undergoing sequential switching to IgE. Mice deficient in IgG1 produce IgE at almost normal levels, but the IgE antibodies produced in IgG1-deficient mice lack the antigen-binding strength and the somatic mutations associated with affinity maturation. A GFP reporter strain, which expresses a modified IgE molecule, was recently developed and was utilized to challenge the sequential switching model. Several molecules that are highly expressed in GC can antagonize class switching to IgE in GC antagonize partially class switching to IgE; in addition, GC IgE(+) cells are gradually lost from GC as the immune response progresses, as shown with another recently developed, Venus-expressing IgE reporter mouse strain. In contrast, as a population, IgG1 cells thrive in the GC environment. Membrane IgE-expressing plasmablasts and plasma cells (PC) were recognized as a major component of the IgE response in secondary lymphoid organs. The swift development of IgE cells toward the PC fate, together with the affinity maturation of the IgE response via an IgG intermediate, represent the most salient features of the IgE immune responses, which make them distinct from IgG responses.
Collapse
Affiliation(s)
- Huizhong Xiong
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, USA
| | | | | |
Collapse
|
45
|
Burton OT, Oettgen HC. Beyond immediate hypersensitivity: evolving roles for IgE antibodies in immune homeostasis and allergic diseases. Immunol Rev 2011; 242:128-43. [PMID: 21682742 DOI: 10.1111/j.1600-065x.2011.01024.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Immunoglobulin E (IgE) antibodies have long been recognized as the antigen-specific triggers of allergic reactions. This review briefly introduces the established functions of IgE in immediate hypersensitivity and then focuses on emerging evidence from our own investigations as well as those of others that IgE plays important roles in protective immunity against parasites and exerts regulatory influences in the expression of its own receptors, FcεRI and CD23, as well as controlling mast cell homeostasis. We provide an overview of the multifaceted ways in which IgE antibodies contribute to the pathology of food allergy and speculate regarding potential mechanisms of action of IgE blockade.
Collapse
Affiliation(s)
- Oliver T Burton
- Division of Immunology, Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|