1
|
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: 3.0] [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
|
2
|
Wade-Vallance AK, Yang Z, Libang JB, Robinson MJ, Tarlinton DM, Allen CD. B cell receptor ligation induces IgE plasma cell elimination. J Exp Med 2023; 220:e20220964. [PMID: 36880536 PMCID: PMC9997509 DOI: 10.1084/jem.20220964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/17/2022] [Accepted: 01/31/2023] [Indexed: 03/08/2023] Open
Abstract
The proper regulation of IgE production safeguards against allergic disease, highlighting the importance of mechanisms that restrict IgE plasma cell (PC) survival. IgE PCs have unusually high surface B cell receptor (BCR) expression, yet the functional consequences of ligating this receptor are unknown. Here, we found that BCR ligation induced BCR signaling in IgE PCs followed by their elimination. In cell culture, exposure of IgE PCs to cognate antigen or anti-BCR antibodies induced apoptosis. IgE PC depletion correlated with the affinity, avidity, amount, and duration of antigen exposure and required the BCR signalosome components Syk, BLNK, and PLCγ2. In mice with a PC-specific impairment of BCR signaling, the abundance of IgE PCs was selectively increased. Conversely, BCR ligation by injection of cognate antigen or anti-IgE depleted IgE PCs. These findings establish a mechanism for the elimination of IgE PCs through BCR ligation. This has important implications for allergen tolerance and immunotherapy as well as anti-IgE monoclonal antibody treatments.
Collapse
Affiliation(s)
- Adam K. Wade-Vallance
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy B. Libang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Marcus J. Robinson
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - David M. Tarlinton
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Christopher D.C. Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Allen CDC. Features of B Cell Responses Relevant to Allergic Disease. THE JOURNAL OF IMMUNOLOGY 2022; 208:257-266. [PMID: 35017215 PMCID: PMC10054180 DOI: 10.4049/jimmunol.2100988] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/04/2021] [Indexed: 01/16/2023]
Abstract
This Brief Review delves into B cell responses in the context of allergy. The primary contribution of B cells to allergy is the production of IgE, the Ab isotype that triggers immediate hypersensitivity reactions through the release of mediators from mast cells and basophils. B cells may also have protective roles in allergy, such as through the production of IgG or as regulatory B cells. In this review, I focus on the basic principles of B cell differentiation and discuss features relevant to allergic immune responses. In particular, I discuss: (1) class-switch recombination; (2) plasma cell differentiation; (3) germinal centers and affinity maturation; and (4) memory B cells and recall responses, with an emphasis on IgE, IgG1, and IgG4. I also consider how B cells may contribute to allergic responses independent of Ab production-for example, by serving as APCs.
Collapse
Affiliation(s)
- Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA; and Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
4
|
Udoye CC, Rau CN, Freye SM, Almeida LN, Vera-Cruz S, Othmer K, Korkmaz RÜ, Clauder AK, Lindemann T, Niebuhr M, Ott F, Kalies K, Recke A, Busch H, Fähnrich A, Finkelman FD, Manz RA. B-cell receptor physical properties affect relative IgG1 and IgE responses in mouse egg allergy. Mucosal Immunol 2022; 15:1375-1388. [PMID: 36114245 PMCID: PMC9705252 DOI: 10.1038/s41385-022-00567-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/10/2022] [Accepted: 09/01/2022] [Indexed: 02/04/2023]
Abstract
Mutated and unmutated IgE and IgG play different and partly opposing roles in allergy development, but the mechanisms controlling their relative production are incompletely understood. Here, we analyzed the IgE-response in murine food allergy. Deep sequencing of the complementary-determining region (CDR) repertoires indicated that an ongoing unmutated extrafollicular IgE response coexists with a germinal center response, even after long-lasting allergen challenges. Despite overall IgG1-dominance, a significant proportion of clonotypes contained several-fold more IgE than IgG1. Clonotypes with differential bias to either IgE or IgG1 showed distinct hypermutation and clonal expansion. Hypermutation rates were associated with different physiochemical binding properties of individual B-cell receptors (BCR). Increasing BCR signaling strength inhibited class switching from IgG1 to IgE in vitro, preferentially constraining IgE formation. These data indicate that antigen-binding properties of individual BCRs determine differential IgE hypermutation and IgE versus IgG1 production on the level of single B-cell clones.
Collapse
Affiliation(s)
- Christopher C. Udoye
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Christina N. Rau
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Sarah M. Freye
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Larissa N. Almeida
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Sarah Vera-Cruz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Kai Othmer
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Rabia Ü. Korkmaz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ann-Katrin Clauder
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Timo Lindemann
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Markus Niebuhr
- grid.4562.50000 0001 0057 2672Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Fabian Ott
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Kathrin Kalies
- grid.4562.50000 0001 0057 2672Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Andreas Recke
- Department of Dermatology, Allergology and Venereology, University off Lübeck, Lübeck, Germany
| | - Hauke Busch
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Anke Fähnrich
- grid.4562.50000 0001 0057 2672Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Fred D. Finkelman
- grid.239573.90000 0000 9025 8099Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine and the Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Rudolf A. Manz
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Yang Z, Wu CAM, Targ S, Allen CDC. IL-21 is a broad negative regulator of IgE class switch recombination in mouse and human B cells. J Exp Med 2020; 217:133860. [PMID: 32130409 PMCID: PMC7201927 DOI: 10.1084/jem.20190472] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/24/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
IgE antibodies may elicit potent allergic reactions, and their production is tightly controlled. The tendency to generate IgE has been thought to reflect the balance between type 1 and type 2 cytokines, with the latter promoting IgE. Here, we reevaluated this paradigm by a direct cellular analysis, demonstrating that IgE production was not limited to type 2 immune responses yet was generally constrained in vivo. IL-21 was a critical negative regulator of IgE responses, whereas IFN-γ, IL-6, and IL-10 were dispensable. Follicular helper T cells were the primary source of IL-21 that inhibited IgE responses by directly engaging the IL-21 receptor on B cells and triggering STAT3-dependent signaling. We reconciled previous discordant results between mouse and human B cells and revealed that the inhibition of IgE class switch recombination by IL-21 was attenuated by CD40 signaling, whereas IgG1 class switch recombination was potentiated by IL-21 in the context of limited IL-4. These findings establish key features of the extrinsic regulation of IgE production by cytokines.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Chung-An M Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Sasha Targ
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA.,Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
6
|
Asrat S, Kaur N, Liu X, Ben LH, Kajimura D, Murphy AJ, Sleeman MA, Limnander A, Orengo JM. Chronic allergen exposure drives accumulation of long-lived IgE plasma cells in the bone marrow, giving rise to serological memory. Sci Immunol 2020; 5:5/43/eaav8402. [PMID: 31924685 DOI: 10.1126/sciimmunol.aav8402] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 05/21/2019] [Accepted: 12/05/2019] [Indexed: 11/02/2022]
Abstract
Immunoglobulin E (IgE) plays an important role in allergic diseases. Nevertheless, the source of IgE serological memory remains controversial. We reexamined the mechanism of serological memory in allergy using a dual reporter system to track IgE+ plasma cells in mice. Short-term allergen exposure resulted in the generation of IgE+ plasma cells that resided mainly in secondary lymphoid organs and produced IgE that was unable to degranulate mast cells. In contrast, chronic allergen exposure led to the generation of long-lived IgE+ plasma cells that were primarily derived from sequential class switching of IgG1, accumulated in the bone marrow, and produced IgE capable of inducing anaphylaxis. IgE+ plasma cells were found in the bone marrow of human allergic, but not nonallergic donors, and allergen-specific IgE produced by these cells was able to induce mast cell degranulation when transferred to mice. These data demonstrate that long-lived IgE+ bone marrow plasma cells arise during chronic allergen exposure and establish serological memory in both mice and humans.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | - Xia Liu
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | | | - Andrew J Murphy
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA
| | | | - Andre Limnander
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA.
| | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, NY 10591, USA.
| |
Collapse
|
7
|
Pioli PD. Plasma Cells, the Next Generation: Beyond Antibody Secretion. Front Immunol 2019; 10:2768. [PMID: 31824518 PMCID: PMC6883717 DOI: 10.3389/fimmu.2019.02768] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/12/2019] [Indexed: 12/31/2022] Open
Abstract
Plasma cells (PCs) represent the terminal differentiation step of mature B lymphocytes. These cells are most recognizable for their extended lifespan as well as their ability to secrete large amounts of antibodies (Abs) thus positioning this cell type as a key component of humoral immunity. However, it is now appreciated that PCs can have far reaching effects on pathologic as well as non-pathologic processes independent of Ab secretion. This is highlighted by recent studies showing that PCs function as key regulators of processes such as hematopoiesis as well as neuro-inflammation. In part, PCs accomplish this by integrating extrinsic signals from their environment which dictate their downstream functionality. Here we summarize the current understanding of PC biology focusing on their ever-growing functional repertoire independent of Ab production. Furthermore, we discuss potential applications of PC immunotherapy and its implementation for translational benefit.
Collapse
Affiliation(s)
- Peter D Pioli
- Department of Biomedical Sciences, Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
8
|
Gould HJ, Wu YCB. IgE repertoire and immunological memory: compartmental regulation and antibody function. Int Immunol 2019; 30:403-412. [PMID: 30053010 PMCID: PMC6116883 DOI: 10.1093/intimm/dxy048] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/19/2018] [Indexed: 01/05/2023] Open
Abstract
It is now generally recognized that bone marrow is the survival niche for antigen-specific plasma cells with long-term immunological memory. These cells release antibodies into the circulation, needed to prime effector cells in the secondary immune response. These antibodies participate in the surveillance for antigen and afford immune defence against pathogens and toxins previously encountered in the primary immune response. IgE antibodies function together with their effector cells, mast cells, to exert 'immediate hypersensitivity' in mucosal tissues at the front line of immune defence. The constant supply of IgE antibodies from bone marrow plasma cells allows the rapid 'recall response' by mast cells upon re-exposure to antigen even after periods of antigen absence. The speed and sensitivity of the IgE recall response and potency of the effector cell functions are advantageous in the early detection and elimination of pathogens and toxins at the sites of attack. Local antigen provocation also stimulates de novo synthesis of IgE or its precursors of other isotypes that undergo IgE switching in the mucosa. This process, however, introduces a delay before mast cells can be sensitized and resume activity; this is terminated shortly after the antigen is eliminated. Recent results from adaptive immune receptor repertoire sequencing of immunoglobulin genes suggest that the mucosal IgE+ plasmablasts, which have undergone affinity maturation in the course of their evolution in vivo, are a source of long-lived IgE+ plasma cells in the bone marrow that are already fully functional.
Collapse
Affiliation(s)
- Hannah J Gould
- Randall Centre in Cell and Molecular Biophysics, King's College London, London, UK.,MRC Asthma UK Center in Allergic Mechanisms of Asthma, London, UK
| | - Yu-Chang Bryan Wu
- Randall Centre in Cell and Molecular Biophysics, King's College London, London, UK.,MRC Asthma UK Center in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|
9
|
AllergoOncology: High innate IgE levels are decisive for the survival of cancer-bearing mice. World Allergy Organ J 2019; 12:100044. [PMID: 31388397 PMCID: PMC6669725 DOI: 10.1016/j.waojou.2019.100044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/22/2019] [Accepted: 06/12/2019] [Indexed: 01/09/2023] Open
Abstract
Background Atopics have a lower risk for malignancies, and IgE targeted to tumors is superior to IgG in fighting cancer. Whether IgE-mediated innate or adaptive immune surveillance can confer protection against tumors remains unclear. Objective We aimed to investigate the effects of active and passive immunotherapy to the tumor-associated antigen HER-2 in three murine models differing in Epsilon-B-cell-receptor expression affecting the levels of expressed IgE. Methods We compared the levels of several serum specific anti-HER-2 antibodies (IgE, IgG1, IgG2a, IgG2b, IgA) and the survival rates in low-IgE ΔM1M2 mice lacking the transmembrane/cytoplasmic domain of Epsilon-B-cell-receptors expressing reduced IgE levels, high-IgE KN1 mice expressing chimeric Epsilon-Gamma1-B-cell receptors with 4-6-fold elevated serum IgE levels, and wild type (WT) BALB/c. Prior engrafting mice with D2F2/E2 mammary tumors overexpressing HER-2, mice were vaccinated with HER-2 or vehicle control PBS using the Th2-adjuvant Al(OH)3 (active immunotherapy), or treated with the murine anti-HER-2 IgG1 antibody 4D5 (passive immunotherapy). Results Overall, among the three strains of mice, HER-2 vaccination induced significantly higher levels of HER-2 specific IgE and IgG1 in high-IgE KN1, while low-IgE ΔM1M2 mice had higher IgG2a levels. HER-2 vaccination and passive immunotherapy prolonged the survival in tumor-grafted WT and low-IgE ΔM1M2 strains compared with treatment controls; active vaccination provided the highest benefit. Notably, untreated high-IgE KN1 mice displayed the longest survival of all strains, which could not be further extended by active or passive immunotherapy. Conclusion Active and passive immunotherapies prolong survival in wild type and low-IgE ΔM1M2 mice engrafted with mammary tumors. High-IgE KN1 mice have an innate survival benefit following tumor challenge.
Collapse
Key Words
- ADCC, Antibody-dependent Cell-mediated Cytotoxicity
- ADCP, Antibody-dependent Cellular Phagocytosis
- AllergoOncology
- BCR, B-Cell Receptor
- Cancer vaccine
- HER-2
- HER-2, Human Epidermal Growth Factor Receptor-2, ErbB-2
- IgA, Immunoglobulin A
- IgE
- IgE, Immunoglobulin E
- IgG, Immunoglobulin G
- Onco-immunology
- TAA, Tumor-Associated Antigen
- WT, wild type
Collapse
|
10
|
Saunders SP, Ma EGM, Aranda CJ, Curotto de Lafaille MA. Non-classical B Cell Memory of Allergic IgE Responses. Front Immunol 2019; 10:715. [PMID: 31105687 PMCID: PMC6498404 DOI: 10.3389/fimmu.2019.00715] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/18/2019] [Indexed: 02/03/2023] Open
Abstract
The long-term effectiveness of antibody responses relies on the development of humoral immune memory. Humoral immunity is maintained by long-lived plasma cells that secrete antigen-specific antibodies, and memory B cells that rapidly respond to antigen re-exposure by generating new plasma cells and memory B cells. Developing effective immunological memory is essential for protection against pathogens, and is the basis of successful vaccinations. IgE responses have evolved for protection against helminth parasites infections and against toxins, but IgE is also a potent mediator of allergic diseases. There has been a dramatic increase in the incidence of allergic diseases in recent decades and this has provided the impetus to study the nature of IgE antibody responses. As will be discussed in depth in this review, the IgE memory response has unique features that distinguish it from classical B cell memory.
Collapse
Affiliation(s)
- Sean P Saunders
- Division of Pulmonary, Critical Care and Sleep Medicine, Laboratory of Allergy and Inflammation, Department of Medicine, New York University, New York, NY, United States
| | - Erica G M Ma
- Division of Pulmonary, Critical Care and Sleep Medicine, Laboratory of Allergy and Inflammation, Department of Medicine, New York University, New York, NY, United States.,Sackler Institute of Graduate Biomedical Sciences, New York University, New York, NY, United States
| | - Carlos J Aranda
- Division of Pulmonary, Critical Care and Sleep Medicine, Laboratory of Allergy and Inflammation, Department of Medicine, New York University, New York, NY, United States
| | - Maria A Curotto de Lafaille
- Division of Pulmonary, Critical Care and Sleep Medicine, Laboratory of Allergy and Inflammation, Department of Medicine, New York University, New York, NY, United States.,Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
11
|
Cyster JG, Allen CDC. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell 2019; 177:524-540. [PMID: 31002794 PMCID: PMC6538279 DOI: 10.1016/j.cell.2019.03.016] [Citation(s) in RCA: 528] [Impact Index Per Article: 105.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/21/2022]
Abstract
B cells and the antibodies they produce have a deeply penetrating influence on human physiology. Here, we review current understanding of how B cell responses are initiated; the different paths to generate short- and long-lived plasma cells, germinal center cells, and memory cells; and how each path impacts antibody diversity, selectivity, and affinity. We discuss how basic research is informing efforts to generate vaccines that induce broadly neutralizing antibodies against viral pathogens, revealing the special features associated with allergen-reactive IgE responses and uncovering the antibody-independent mechanisms by which B cells contribute to health and disease.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Christopher D C Allen
- Cardiovascular Research Institute, Department of Anatomy, and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
12
|
Mohan T, Zhu W, Wang Y, Wang BZ. Applications of chemokines as adjuvants for vaccine immunotherapy. Immunobiology 2017; 223:477-485. [PMID: 29246401 DOI: 10.1016/j.imbio.2017.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023]
Abstract
Vaccinations are expected to aid in building immunity against pathogens. This objective often requires the addition of an adjuvant with certain vaccine formulations containing weakly immunogenic antigens. Adjuvants can improve antigen processing, presentation, and recognition, thereby improving the immunogenicity of a vaccine by simulating and eliciting an immune response. Chemokines are a group of small chemoattractant proteins that are essential regulators of the immune system. They are involved in almost every aspect of tumorigenesis, antitumor immunity, and antimicrobial activity and also play a critical role in regulating innate and adaptive immune responses. More recently, chemokines have been used as vaccine adjuvants due to their ability to modulate lymphocyte development, priming and effector functions, and enhance protective immunity. Chemokines that are produced naturally by the body's own immune system could serve as potentially safer and more reliable adjuvant options versus synthetic adjuvants. This review will primarily focus on chemokines and their immunomodulatory activities against various infectious diseases and cancers.
Collapse
Affiliation(s)
- Teena Mohan
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
13
|
Laffleur B, Debeaupuis O, Dalloul Z, Cogné M. B Cell Intrinsic Mechanisms Constraining IgE Memory. Front Immunol 2017; 8:1277. [PMID: 29180995 PMCID: PMC5694035 DOI: 10.3389/fimmu.2017.01277] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 09/25/2017] [Indexed: 12/25/2022] Open
Abstract
Memory B cells and long-lived plasma cells are key elements of adaptive humoral immunity. Regardless of the immunoglobulin class produced, these cells can ensure long-lasting protection but also long-lasting immunopathology, thus requiring tight regulation of their generation and survival. Among all antibody classes, this is especially true for IgE, which stands as the most potent, and can trigger dramatic inflammatory reactions even when present in minute amounts. IgE responses and memory crucially protect against parasites and toxic components of venoms, conferring selective advantages and explaining their conservation in all mammalian species despite a parallel broad spectrum of IgE-mediated immunopathology. Long-term memory of sensitization and anaphylactic responses to allergens constitute the dark side of IgE responses, which can trigger multiple acute or chronic pathologic manifestations, some punctuated with life-threatening events. This Janus face of the IgE response and memory, both necessary and potentially dangerous, thus obviously deserves the most elaborated self-control schemes.
Collapse
Affiliation(s)
- Brice Laffleur
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | | | - Zeinab Dalloul
- UMR 7276 Centre National de la Recherche Scientifique: Contrôle de la Réponse Immune B et des Lymphoproliférations, Université de Limoges, Limoges, France
| | - Michel Cogné
- UMR 7276 Centre National de la Recherche Scientifique: Contrôle de la Réponse Immune B et des Lymphoproliférations, Université de Limoges, Limoges, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
14
|
Jiménez-Saiz R, Chu DK, Mandur TS, Walker TD, Gordon ME, Chaudhary R, Koenig J, Saliba S, Galipeau HJ, Utley A, King IL, Lee K, Ettinger R, Waserman S, Kolbeck R, Jordana M. Lifelong memory responses perpetuate humoral T H2 immunity and anaphylaxis in food allergy. J Allergy Clin Immunol 2017; 140:1604-1615.e5. [PMID: 28216433 DOI: 10.1016/j.jaci.2017.01.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/10/2017] [Accepted: 01/26/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND A number of food allergies (eg, fish, shellfish, and nuts) are lifelong, without any disease-transforming therapies, and unclear in their underlying immunology. Clinical manifestations of food allergy are largely mediated by IgE. Although persistent IgE titers have been attributed conventionally to long-lived IgE+ plasma cells (PCs), this has not been directly and comprehensively tested. OBJECTIVE We sought to evaluate mechanisms underlying persistent IgE and allergic responses to food allergens. METHODS We used a model of peanut allergy and anaphylaxis, various knockout mice, adoptive transfer experiments, and in vitro assays to identify mechanisms underlying persistent IgE humoral immunity over almost the entire lifespan of the mouse (18-20 months). RESULTS Contrary to conventional paradigms, our data show that clinically relevant lifelong IgE titers are not sustained by long-lived IgE+ PCs. Instead, lifelong reactivity is conferred by allergen-specific long-lived memory B cells that replenish the IgE+ PC compartment. B-cell reactivation requires allergen re-exposure and IL-4 production by CD4 T cells. We define the half-lives of antigen-specific germinal centers (23.3 days), IgE+ and IgG1+ PCs (60 and 234.4 days, respectively), and clinically relevant cell-bound IgE (67.3 days). CONCLUSIONS These findings can explain lifelong food allergies observed in human subjects as the consequence of allergen exposures that recurrently activate memory B cells and identify these as a therapeutic target with disease-transforming potential.
Collapse
Affiliation(s)
- Rodrigo Jiménez-Saiz
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Derek K Chu
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Talveer S Mandur
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Tina D Walker
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Melissa E Gordon
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Roopali Chaudhary
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joshua Koenig
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Saliba
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Heather J Galipeau
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Adam Utley
- Departments of Immunology and Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Irah L King
- Department of Microbiology & Immunology, McGill University, Montreal, Quebec, Canada
| | - Kelvin Lee
- Departments of Immunology and Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Rachel Ettinger
- Department of Respiratory, Inflammation & Autoimmunity, MedImmune, Gaithersburg, Md
| | - Susan Waserman
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Roland Kolbeck
- Department of Respiratory, Inflammation & Autoimmunity, MedImmune, Gaithersburg, Md
| | - Manel Jordana
- McMaster Immunology Research Centre (MIRC), Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
15
|
Initiation, Persistence and Exacerbation of Food Allergy. BIRKHÄUSER ADVANCES IN INFECTIOUS DISEASES 2017. [DOI: 10.1007/978-3-319-69968-4_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
16
|
Nigro EA, Brini AT, Yenagi VA, Ferreira LM, Achatz-Straussberger G, Ambrosi A, Sanvito F, Soprana E, van Anken E, Achatz G, Siccardi AG, Vangelista L. Cutting Edge: IgE Plays an Active Role in Tumor Immunosurveillance in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 197:2583-8. [DOI: 10.4049/jimmunol.1601026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/11/2016] [Indexed: 11/19/2022]
|
17
|
Autonomous membrane IgE signaling prevents IgE-memory formation. Nat Immunol 2016; 17:1109-17. [PMID: 27428827 DOI: 10.1038/ni.3508] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/02/2016] [Indexed: 12/14/2022]
Abstract
Aberrant production of IgE antibodies can lead to allergic diseases. Normally, IgE(+) B cells rarely differentiate into memory B cells (Bmem) or long-lived plasma cells (LLPCs), as they only transiently participate in the germinal center (GC), but the mechanism behind this remains elusive. We found that membrane IgE (mIgE) autonomously triggered rapid plasma-cell differentiation and apoptosis independently of antigen or cellular context, predominantly through the mutually independent CD19-PI3K-Akt-IRF4 and BLNK-Jnk/p38 pathways, respectively, and we identified the ectodomains of mIgE as being responsible. Accordingly, deregulated GC IgE(+) B cell proliferation and prolonged IgE production with exaggerated anaphylaxis were observed in CD19- and BLNK-deficient mice. Our findings reveal an autonomous mIgE signaling mechanism that normally prevents IgE(+) Bmem and LLPC formation, providing insights into the molecular pathogenesis of allergic diseases.
Collapse
|
18
|
Biology of IgE production: IgE cell differentiation and the memory of IgE responses. Curr Top Microbiol Immunol 2015; 388:1-19. [PMID: 25553792 DOI: 10.1007/978-3-319-13725-4_1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The generation of long-lived plasma cells and memory B cells producing high-affinity antibodies depends on the maturation of B cell responses in germinal centers. These processes are essential for long-lasting antibody-mediated protection against infections. IgE antibodies are important for defense against parasites and toxins and can also mediate anti-tumor immunity. However, high-affinity IgE is also the main culprit responsible for the manifestations of allergic disease, including life-threatening anaphylaxisAnaphylaxis . Thus, generation of high-affinity IgE must be tightly regulated. Recent studies of IgE B cell biology have unveiled two mechanisms that limit high-affinity IgE memory responses: First, B cells that have recently switched to IgE production are programmed to rapidly differentiate into plasma cells,Plasma cells and second, IgE germinal centerGerminal center cells are transient and highly apoptotic. Opposing these processes, we now know that germinal center-derived IgG B cells can switch to IgE production, effectively becoming IgE-producing plasma cells. In this chapter, we will discuss the unique molecular and cellular pathways involved in the generation of IgE antibodies.
Collapse
|
19
|
Gauvreau GM, Harris JM, Boulet LP, Scheerens H, Fitzgerald JM, Putnam WS, Cockcroft DW, Davis BE, Leigh R, Zheng Y, Dahlén B, Wang Y, Maciuca R, Mayers I, Liao XC, Wu LC, Matthews JG, O'Byrne PM. Targeting membrane-expressed IgE B cell receptor with an antibody to the M1 prime epitope reduces IgE production. Sci Transl Med 2015; 6:243ra85. [PMID: 24990880 DOI: 10.1126/scitranslmed.3008961] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Elevated serum levels of both total and allergen-specific immunoglobulin E (IgE) correlate with atopic diseases such as allergic rhinitis and allergic asthma. Neutralization of IgE by anti-IgE antibodies can effectively treat allergic asthma. Preclinical studies indicate that targeting membrane IgE-positive cells with antibodies against M1 prime can inhibit the production of new IgE and significantly reduce the levels of serum IgE. We report results from two trials that investigated the safety, pharmacokinetics, and activity of quilizumab, a humanized monoclonal antibody targeting specifically the M1 prime epitope of membrane IgE, in subjects with allergic rhinitis (NCT01160861) or mild allergic asthma (NCT01196039). In both studies, quilizumab treatment was well tolerated and led to reductions in total and allergen-specific serum IgE that lasted for at least 6 months after the cessation of dosing. In subjects with allergic asthma who were subjected to an allergen challenge, quilizumab treatment blocked the generation of new IgE, reduced allergen-induced early and late asthmatic airway responses by 26 and 36%, respectively, and reduced allergen-induced increases in sputum eosinophils by ~50% compared with placebo. These studies indicate that targeting of membrane IgE-expressing cells with anti-M1 prime antibodies can prevent IgE production in humans.
Collapse
Affiliation(s)
| | | | - Louis-Philippe Boulet
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Québec G1V 4G5, Canada
| | | | - J Mark Fitzgerald
- University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | | | | | - Beth E Davis
- University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Richard Leigh
- University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - Yanan Zheng
- Genentech Inc., South San Francisco, CA 94080, USA
| | - Barbro Dahlén
- Karolinska University Hospital, Stockholm S-141 86, Sweden
| | - Yehong Wang
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Irvin Mayers
- University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | | | - Lawren C Wu
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | | |
Collapse
|
20
|
Self-Restrained B Cells Arise following Membrane IgE Expression. Cell Rep 2015; 10:900-909. [PMID: 25683713 DOI: 10.1016/j.celrep.2015.01.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/21/2014] [Accepted: 01/08/2015] [Indexed: 11/20/2022] Open
Abstract
Among immunoglobulins (Igs), IgE can powerfully contribute to antimicrobial immunity and severe allergy despite its low abundance. IgE protein and gene structure resemble other Ig classes, making it unclear what constrains its production to thousand-fold lower levels. Whether class-switched B cell receptors (BCRs) differentially control B cell fate is debated, and study of the membrane (m)IgE class is hampered by its elusive in vivo expression. Here, we demonstrate a self-controlled mIgE+ B cell stage. Primary or transfected mIgE+ cells relocate the BCRs into spontaneously internalized lipid rafts, lose mobility to chemokines, and change morphology. We suggest that combined proapoptotic mechanisms possibly involving Hax1 prevent mIgE+ memory lymphocyte accumulation. By uncoupling in vivo IgE switching from cytokine and antigen stimuli, we show that these features are independent from B cell stimulation and instead result from mIgE expression per se. Consequently, few cells survive IgE class switching, which might ensure minimal long-term IgE memory upon differentiation into plasma cells.
Collapse
|
21
|
Laffleur B, Denis-Lagache N, Péron S, Sirac C, Moreau J, Cogné M. AID-induced remodeling of immunoglobulin genes and B cell fate. Oncotarget 2015; 5:1118-31. [PMID: 24851241 PMCID: PMC4012742 DOI: 10.18632/oncotarget.1546] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Survival and phenotype of normal and malignant B lymphocytes are critically dependent on constitutive signals by the B cell receptor (BCR) for antigen. In addition, either antigen ligation of the BCR or various mitogenic stimuli result in B cell activation and induction of activation-induced deaminase (AID). AID activity can in turn mediate somatic hypermutation (SHM) of immunoglobulin (Ig) V regions and also deeply remodel the Ig heavy chain locus through class switch recombination (CSR) or locus suicide recombination (LSR). In addition to changes linked to affinity for antigen, modifying the class/isotype (i.e. the structure and function) of the BCR or suddenly deleting BCR expression also modulates the fate of antigen-experienced B cells.
Collapse
|
22
|
Gould HJ, Ramadani F. IgE responses in mouse and man and the persistence of IgE memory. Trends Immunol 2014; 36:40-8. [PMID: 25499855 DOI: 10.1016/j.it.2014.11.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 12/29/2022]
Abstract
Rapid and robust recall or 'memory' responses are an essential feature of adaptive immunity. They constitute a defense against reinfection by pathogens, yet arguably do more harm than good in allergic disease. Immunoglobulin (Ig)E antibodies mediate the allergic reaction characterized by immediate hypersensitivity, a manifestation of IgE memory. The origin of IgE memory remains obscure, mainly due to the low proportion of IgE-expressing B cells in the total B cell population. The recent development of ultrasensitive methods for tracking these cells in vivo has overcome this obstacle, and their use has revealed unexpected pathways to IgE memory in the mouse. Here, we review these findings and consider their bearing on our understanding of IgE memory and allergic disease in man.
Collapse
Affiliation(s)
- Hannah J Gould
- Divisions of Cell and Molecular Biophysics and Asthma, Allergy and Lung Biology, King's College London, London, SE1 1UL, UK.
| | - Faruk Ramadani
- Divisions of Cell and Molecular Biophysics and Asthma, Allergy and Lung Biology, King's College London, London, SE1 1UL, UK
| |
Collapse
|
23
|
Brachs S, Turqueti-Neves A, Stein M, Reimer D, Brachvogel B, Bösl M, Winkler T, Voehringer D, Jäck HM, Mielenz D. Swiprosin-1/EFhd2 limits germinal center responses and humoral type 2 immunity. Eur J Immunol 2014; 44:3206-19. [PMID: 25092375 DOI: 10.1002/eji.201444479] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 06/27/2014] [Accepted: 07/31/2014] [Indexed: 01/19/2023]
Abstract
Activated B cells are selected for in germinal centers by regulation of their apoptosis. The Ca2+ -binding cytoskeletal adaptor protein Swiprosin-1/EFhd2 (EFhd2) can promote apoptosis in activated B cells. We therefore hypothesized that EFhd2 might limit humoral immunity by repressing both the germinal center reaction and the expected enhancement of immune responses in the absence of EFhd2. Here, we established EFhd2(-/-) mice on a C57BL/6 background, which revealed normal B- and T-cell development, basal Ab levels, and T-cell independent type 1, and T-cell independent type 2 responses. However, T cell-dependent immunization with sheep red blood cells and infection with the helminth Nippostrongylus brasiliensis (N.b) increased production of antibodies of multiple isotypes, as well as germinal center formation in EFhd2(-/-) mice. In addition, serum IgE levels and numbers of IgE+ plasma cells were strongly increased in EFhd2(-/-) mice, both after primary as well as after secondary N.b infection. Finally, mixed bone marrow chimeras unraveled an EFhd2-dependent B cell-intrinsic contribution to increased IgE plasma cell numbers in N.b-infected mice. Hence, we established a role for EFhd2 as a negative regulator of germinal center-dependent humoral type 2 immunity, with implications for the generation of IgE.
Collapse
Affiliation(s)
- Sebastian Brachs
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus Fiebiger Center, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Nuremberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wu LC, Scheerens H. Targeting IgE production in mice and humans. Curr Opin Immunol 2014; 31:8-15. [PMID: 25156315 DOI: 10.1016/j.coi.2014.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 11/26/2022]
Abstract
Immunoglobulin E (IgE) is pathogenic in allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, and food allergy. Recent studies using genetically modified IgE reporter mice indicate that the majority of serum IgE in mice is produced by short-lived IgE plasma cells, with minor contributions from long-lived IgE plasma cells, and implicate IgG1 and IgE memory B cells as potential sources of IgE memory. Clinical studies using antibodies against IL-13 or the IL-4 and IL-13 receptor subunit IL-4Rα, as well as an antibody against the M1 prime domain of human membrane IgE, indicate that, similar to mice, a proportion of IgE in humans is derived from ongoing IgE immune responses and short-lived plasma cells. Targeting IgE production may lead to new therapies for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Lawren C Wu
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Heleen Scheerens
- Department of Pharmacodynamic Biomarkers, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
25
|
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: 194] [Impact Index Per Article: 19.4] [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
|
26
|
Yang Z, Robinson MJ, Allen CDC. Regulatory constraints in the generation and differentiation of IgE-expressing B cells. Curr Opin Immunol 2014; 28:64-70. [PMID: 24632082 DOI: 10.1016/j.coi.2014.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/01/2014] [Accepted: 02/03/2014] [Indexed: 11/25/2022]
Abstract
B cells expressing antibodies of the immunoglobulin E (IgE) isotype are rare, yet are heavily implicated in the pathogenesis of allergies and asthma. This review discusses recent methodological advances that permit sensitive probing of IgE-expressing (IgE(+)) B cells in vivo and have accordingly clarified the basic behavior and fate of IgE(+) B cells during immune responses in mouse models. IgE antibody secreting plasma cells can arise from extrafollicular foci, germinal centers, and memory B cells. However, compared to B cells expressing other isotypes, IgE(+) B cells are susceptible to multiple additional regulatory constraints that restrict the size of the IgE(+) B cell pool at each stage, coordinately limiting the overall magnitude, affinity, and duration of the IgE antibody response.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marcus J Robinson
- Cardiovascular Research Institute and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher D C Allen
- Cardiovascular Research Institute and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy and Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
27
|
He JS, Meyer-Hermann M, Xiangying D, Zuan LY, Jones LA, Ramakrishna L, de Vries VC, Dolpady J, Aina H, Joseph S, Narayanan S, Subramaniam S, Puthia M, Wong G, Xiong H, Poidinger M, Urban JF, Lafaille JJ, Curotto de Lafaille MA. The distinctive germinal center phase of IgE+ B lymphocytes limits their contribution to the classical memory response. ACTA ACUST UNITED AC 2013; 210:2755-71. [PMID: 24218137 PMCID: PMC3832920 DOI: 10.1084/jem.20131539] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Direct class switching to IgE generates IgE+ GC cells that are highly apoptotic and do not contribute to the memory compartment, while sequential switching through an IgG+ intermediate results in the generation of long-lived IgE+ plasma cells. The mechanisms involved in the maintenance of memory IgE responses are poorly understood, and the role played by germinal center (GC) IgE+ cells in memory responses is particularly unclear. IgE+ B cell differentiation is characterized by a transient GC phase, a bias toward the plasma cell (PC) fate, and dependence on sequential switching for the production of high-affinity IgE. We show here that IgE+ GC B cells are unfit to undergo the conventional GC differentiation program due to impaired B cell receptor function and increased apoptosis. IgE+ GC cells fail to populate the GC light zone and are unable to contribute to the memory and long-lived PC compartments. Furthermore, we demonstrate that direct and sequential switching are linked to distinct B cell differentiation fates: direct switching generates IgE+ GC cells, whereas sequential switching gives rise to IgE+ PCs. We propose a comprehensive model for the generation and memory of IgE responses.
Collapse
Affiliation(s)
- Jin-Shu He
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Komegae EN, Grund LZ, Lopes-Ferreira M, Lima C. The longevity of Th2 humoral response induced by proteases natterins requires the participation of long-lasting innate-like B cells and plasma cells in spleen. PLoS One 2013; 8:e67135. [PMID: 23840604 PMCID: PMC3696013 DOI: 10.1371/journal.pone.0067135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 05/16/2013] [Indexed: 11/22/2022] Open
Abstract
The generation of long-lived antibody-secreting cells (ASC) and memory B cells are critical events for an effective vaccine and the choice of adjuvant can influence these processes. Various cellular and molecular mechanism involved in the protease action that determine Th2 responses have been identified. However, direct or indirect actions in the regulation of the induction, survival and longevity of ASC in differential compartments remain largely unknown. We investigated whether the proteolytic activity of proteins are determinant for the modulation of the memory immune response in mice, promoting the differentiation of memory B cells to terminally differentiated end stage cells. Here, we show that the proteolytic activity of Natterins, from the venom of Thalassophryne nattereri Brazilian fish, besides inducing a Th2 response with plasmatic titers of high-affinity antigen-specific IgE over extended periods is sufficient for the generation of signals that contribute to the formation of a survival niche in the spleen, essential for the longevity of the main subtype of ASC with B220neg phenotype.
Collapse
Affiliation(s)
- Evilin Naname Komegae
- Immunoregulation Unit, Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Lidiane Zito Grund
- Immunoregulation Unit, Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Monica Lopes-Ferreira
- Immunoregulation Unit, Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Carla Lima
- Immunoregulation Unit, Special Laboratory of Applied Toxinology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
29
|
Winter O, Dame C, Jundt F, Hiepe F. Pathogenic long-lived plasma cells and their survival niches in autoimmunity, malignancy, and allergy. THE JOURNAL OF IMMUNOLOGY 2013; 189:5105-11. [PMID: 23169863 DOI: 10.4049/jimmunol.1202317] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Long-lived plasma cells survive in a protected microenvironment for years or even a lifetime and provide humoral memory by establishing persistent Ab titers. Long-lived autoreactive, malignant, and allergen-specific plasma cells are likewise protected in their survival niche and are refractory to immunosuppression, B cell depletion, and irradiation. Their elimination remains an essential therapeutic challenge. Recent data indicate that long-lived plasma cells reside in a multicomponent plasma cell niche with a stable mesenchymal and a dynamic hematopoietic component, both providing essential soluble and membrane-bound survival factors. Alternative niches with different hematopoietic cell components compensate fluctuations of single cell types but may also harbor distinct plasma cell subsets. In this Brief Review, we discuss conventional therapies in autoimmunity and multiple myeloma in comparison with novel drugs that target plasma cells and their niches. In the future, such strategies may enable the specific depletion of pathogenic plasma cells while leaving the protective humoral memory intact.
Collapse
Affiliation(s)
- Oliver Winter
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, 10117 Berlin, Germany.
| | | | | | | |
Collapse
|
30
|
|
31
|
Yang Z, Sullivan BM, Allen CDC. Fluorescent in vivo detection reveals that IgE(+) B cells are restrained by an intrinsic cell fate predisposition. Immunity 2012; 36:857-72. [PMID: 22406270 DOI: 10.1016/j.immuni.2012.02.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/30/2012] [Accepted: 02/09/2012] [Indexed: 10/28/2022]
Abstract
IgE antibodies may be protective in parasite immunity, but their aberrant production can lead to allergic disease and life-threatening anaphylaxis. Despite the importance of IgE regulation, few studies have directly examined the B cells that express IgE, because these cells are rare and difficult to detect. Here, we describe fluorescent IgE reporter mice and validate a flow cytometry procedure to allow sensitive and specific identification of IgE-expressing B cells in vivo. Similar to IgG1(+) cells, IgE(+) B cells differentiated into germinal center (GC) B cells and plasma cells (PCs) during primary immune responses to a T cell-dependent hapten-protein conjugate and the helminth Nippostrongylus brasiliensis. However, the participation of IgE(+) B cells in GCs was transient. IgE(+) B cells had an atypical propensity to upregulate the transcription factor Blimp-1 and undergo PC differentiation. Most IgE(+) PCs were short lived and showed reduced affinity maturation, revealing intrinsic mechanisms that restrict the IgE antibody response.
Collapse
Affiliation(s)
- Zhiyong Yang
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
32
|
Long-lived autoreactive plasma cells drive persistent autoimmune inflammation. Nat Rev Rheumatol 2011; 7:170-8. [DOI: 10.1038/nrrheum.2011.1] [Citation(s) in RCA: 253] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Rogosch T, Kerzel S, Sikula L, Gentil K, Liebetruth M, Schlingmann KP, Maier RF, Zemlin M. Plasma Cells and Nonplasma B Cells Express Differing IgE Repertoires in Allergic Sensitization. THE JOURNAL OF IMMUNOLOGY 2010; 184:4947-54. [DOI: 10.4049/jimmunol.0900859] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Chen JB, Wu PC, Hung AFH, Chu CY, Tsai TF, Yu HM, Chang HY, Chang TW. Unique epitopes on C epsilon mX in IgE-B cell receptors are potentially applicable for targeting IgE-committed B cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:1748-56. [PMID: 20083663 DOI: 10.4049/jimmunol.0902437] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Membrane-bound IgE (mIgE) is part of the IgE-BCR and is essential for generating isotype-specific IgE responses. On mIgE(+) B cells, the membrane-bound epsilon-chain (mepsilon) exists predominantly in the long isoform, mepsilon(L), containing an extra 52 aa CepsilonmX domain between CH4 and the C-terminal membrane-anchoring segment; the short isoform of mepsilon, mepsilon(S), exists in minor proportions. CepsilonmX thus provides an attractive site for immunologic targeting of mIgE(+) B cells. In this study, we show that nine newly prepared CepsilonmX-specific mAbs, as well as the previously reported a20, bound to mIgE.Fc(L)-expressing CHO cells, while only 4B12 and 26H2 bound to mIgE.Fc(L)-expressing B cell line Ramos cells. The mAb 4B12 bound to the N-terminal part, 26H2 the middle part, and all others the C-terminal part of CepsilonmX. Expression of Igalpha and Igbeta on the mIgE.Fc(L)-CHO cells reduces the binding of a20 to CepsilonmX as compared with that of 4B12 and 26H2. The chimeric mAbs c4B12 and c26H2, when cross-linked by secondary antibodies, lysed mIgE.Fc(L)-Ramos cells by apoptosis through a BCR-dependent caspase pathway. Using PBMCs as the source of effector cells, c4B12 and c26H2 demonstrated Ab-dependent cellular cytotoxicity toward mIgE.Fc(L)-Ramos cells in a dose-dependent fashion. In cultures of PBMCs from atopic dermatitis patients, c4B12 and c26H2 inhibited the synthesis of IgE driven by anti-CD40 and IL-4. These results suggest that 4B12 and 26H2 and an immunogen using the peptide segments recognized by these mAbs are potentially useful for targeting mIgE(+) B cells to control IgE production.
Collapse
Affiliation(s)
- Jiun-Bo Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Achatz-Straussberger G, Zaborsky N, Königsberger S, Feichtner S, Lenz S, Peckl-Schmid D, Lamers M, Achatz G. Limited humoral immunoglobulin E memory influences serum immunoglobulin E levels in blood. Clin Exp Allergy 2009; 39:1307-13. [PMID: 19489847 DOI: 10.1111/j.1365-2222.2009.03278.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The switch of B cells expressing membrane-bound Igs, which serve as antigen receptors, to antibody-secreting plasmablasts and finally to non-dividing, long-lived plasma cells (PCs) lacking an antigen receptor, marks the terminal differentiation of a B cell. Antibody-secreting PCs represent the key cell type for the maintenance of a proactive humoral immunological memory. Although some populations of long-lived PCs persist in the spleen, most of them return to their 'place of birth' and travel to the bone marrow or invade inflamed tissues, where they survive up to several months in survival niches as resident, immobile cells. Existing data strongly support the notion that isotype-specific receptor signalling influences the migration behaviour of plasmablasts to the bone marrow. The recent observation in the murine system that the immigration of plasmablasts and the final differentiation to long-lived PCs in the bone marrow is dependent on the expressed B-cell isotype and the related expression of chemokine receptors leads to the conclusion that during a T-helper type 2 (Th2)-mediated immune response in wild type mice, IgE plasmablasts do not have the same chance to contribute to long-lived PC memory as IgG1 plasmablasts. The overall limited humoral IgE memory additionally restricts the quantity of IgE Igs in the serum.
Collapse
|