1
|
Chandrakar P, Nelson CS, Podestà MA, Cavazzoni CB, Gempler M, Lee JM, Richardson S, Zhang H, Samarpita S, Ciofani M, Chatila T, Kuchroo VK, Sage PT. Progressively differentiated T FH13 cells are stabilized by JunB to mediate allergen germinal center responses. Nat Immunol 2025:10.1038/s41590-025-02077-y. [PMID: 39891019 DOI: 10.1038/s41590-025-02077-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Allergic diseases are common and affect a large proportion of the population. Interleukin-13 (IL-13)-expressing follicular helper T (TFH13) cells are a newly identified population of TFH cells that have been associated with high-affinity IgE responses. However, the origins, developmental signals, transcriptional programming and precise functions of TFH13 cells are unknown. Here, we examined the developmental signals for TFH13 cells and found a direct and progressive differentiation pathway marked by the production of IL-21. These two pathways differed in kinetics and extrinsic requirements. However, both pathways converged, forming transcriptionally similar TFH13 cells that express the transcription factor JunB as a critical stabilizing factor. Using an intersectional genetics-based TFH13-diphtheria toxin receptor model to perturb these cells, we found that TFH13 cells were essential to drive broad germinal center responses and allergen-specific IgG and IgE. Moreover, we found that IL-21 is a broad positive regulator of allergen germinal center B cells and synergizes with IL-13 produced by TFH13 cells to amplify allergic responses. Thus, TFH13 cells orchestrate multiple features of allergic inflammation.
Collapse
Affiliation(s)
- Pragya Chandrakar
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cody S Nelson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Manuel A Podestà
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Unit of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia B Cavazzoni
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Gempler
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sierra Richardson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hengcheng Zhang
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Snigdha Samarpita
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Ciofani
- Department of Integrative Immunology, Duke University Medical Center, Durham, NC, USA
| | - Talal Chatila
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammatory Diseases, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Peter T Sage
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Dent AL. Regulation of the IgE response by T follicular regulatory cells. Allergol Int 2025; 74:20-24. [PMID: 39232918 DOI: 10.1016/j.alit.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 09/06/2024] Open
Abstract
Allergen-specific IgE is a major mediator of allergic responses and contributes greatly to allergic disease in the human population. Therapies that inhibit the production of IgE would be useful for lessening the burden of allergic disease. A great deal of research has focused on how IgE responses are regulated, and several factors that promote the production of allergic IgE have been characterized. T follicular helper (TFH) cells expressing IL-4 are required for the development of IgE expressing B cells in the germinal center (GC). Ig somatic hypermutation and B cell selection in the GC leads to the development of high affinity allergen-specific IgE that promotes anaphylaxis, a severe form of allergic response. T follicular regulatory (TFR) cells are also found in the GC response and act with TFH cells in the selection of high affinity IgE + B cells. This review examines the current literature on IgE responses and TFR cells. In mouse studies, TFR cells have a suppressive role on IgE responses in allergic airway disease, however TFR cells also play a helper role in the IgE response in food allergy. In human studies, TFR cells correlate with a decreased allergic response but evidence for a direct suppressive role of TFR cells on IgE in vivo is lacking. TFR cells may represent a new target for allergy therapies, but caution must be exercised to promote the suppressor activity of TFR cells and not the helper activity of TFR cells on IgE responses.
Collapse
Affiliation(s)
- Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 W. Walnut St., R2 302 Indianapolis, IN 46202, USA.
| |
Collapse
|
3
|
Cañete PF, Yu D. Follicular T cells and the control of IgE responses. Allergol Int 2025; 74:13-19. [PMID: 39455298 DOI: 10.1016/j.alit.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Atopy is considered the epidemic of the 21st century, and while decades of research have established a direct link between Th2 cells driving pathogenic IgE-mediated allergic disease, only in the past years have T follicular helper (Tfh) cells emerged as pivotal drivers of these responses. In this review, we will examine the molecular mechanisms governing the IgE response, with a particular emphasis on the key cytokines and signaling pathways. We will discuss the exclusion of IgE-producing B cells from germinal centers and explore the recently established role of follicular T cell function and heterogeneity in driving or curtailing these immune responses. Additionally, we will assess the current state of major monoclonal antibodies and allergen immunotherapies designed to counteract Th2-driven inflammation, as well as reflect on the need to investigate how these biologics impact Tfh cell activity, differentiation, and function, as these insights could pave the way for much-needed therapeutic innovation in the treatment of allergic diseases.
Collapse
Affiliation(s)
- Pablo F Cañete
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Di Yu
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
4
|
Koenig JFE. T follicular helper and memory B cells in IgE recall responses. Allergol Int 2025; 74:4-12. [PMID: 39562254 DOI: 10.1016/j.alit.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
IgE antibodies raised against innocuous environmental antigens cause allergic diseases like allergic rhinitis, food allergy, and allergic asthma. While some allergies are often outgrown, others (peanut, shellfish, tree nut) are lifelong in the majority of individuals. Lifelong allergies are the result of persistent production of allergen-specific IgE. However, IgE antibodies and the plasma cells that secrete them tend to be short-lived. Persistent allergen-specific IgE titres are thought to be derived from the continued renewal of IgE plasma cells from memory B cells in response to allergen encounters. The initial generation of allergen-specific IgE is driven by B cell activation by IL-4 producing Tfh cells, but the cellular and molecular mechanisms of the long-term production of IgE are poorly characterized. This review investigates the mechanisms governing IgE production and Tfh activation in the primary and recall responses, towards the objective of identifying molecular targets for therapeutic intervention that durably inactivate the IgE recall response.
Collapse
Affiliation(s)
- Joshua F E Koenig
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada; Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
5
|
Chen JS, Lee D, Gowthaman U. T follicular helper cells in food allergy. Curr Opin Immunol 2024; 91:102461. [PMID: 39276414 DOI: 10.1016/j.coi.2024.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/17/2024]
Abstract
T follicular helper (Tfh) cells help direct the production of antibodies by B cells. In addition to promoting antibody responses to vaccination and infection, Tfh cells have been found to mediate antibody production to food antigens. Work over the past decade has delineated the specific phenotypes of Tfh cells that induce antibodies to food while also clarifying the divergent Tfh cell requirement for different food-specific antibody isotypes. Furthermore, Tfh and antibody responses to food can occur at multiple barrier sites - namely, skin, airway, and gut. Depending on the context of food antigen exposure, the immune response to food at these sites can be protective, as in the case of tolerance or immunotherapy, or pathogenic, as in the case of allergy. This review will highlight recent advances in our understanding of how Tfh cells promote antibodies to food as well as future avenues for continued discovery.
Collapse
Affiliation(s)
- Jennifer S Chen
- Department of Internal Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| | - Donguk Lee
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Uthaman Gowthaman
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
6
|
Fernandes-Braga W, Curotto de Lafaille MA. B cell memory of Immunoglobulin E (IgE) antibody responses in allergy. Curr Opin Immunol 2024; 91:102488. [PMID: 39340881 DOI: 10.1016/j.coi.2024.102488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Immunoglobulin E (IgE)-mediated allergic diseases are driven by high-affinity allergen-specific IgE antibodies. IgE antibodies bind to Fc epsilon receptors on mast cells, prompting their degranulation and initiating inflammatory reactions upon allergen crosslinking. While most IgE-producing plasma cells have short lifespans, and IgE memory B cells are exceedingly rare, studies have indicated that non-IgE-expressing type 2-polarized IgG memory B cells serve as a reservoir of IgE memory in allergies. This review explores the B cell populations underlying IgE-mediated allergies, including the cellular and molecular processes that drive IgE class switching from non-IgE memory B cells. It highlights emerging evidence from human studies identifying type 2 IgG memory B cells as the source of pathogenic IgE in allergic responses.
Collapse
Affiliation(s)
- Weslley Fernandes-Braga
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A Curotto de Lafaille
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
Seidler CA, Liedl KR. CDR L3 Loop Rearrangement Switches Multispecific SPE-7 IgE Antibody From Hapten to Protein Binding. J Mol Recognit 2024; 37:e3107. [PMID: 39375932 DOI: 10.1002/jmr.3107] [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: 04/15/2024] [Revised: 07/11/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024]
Abstract
The monoclonal IgE antibody SPE-7 was originally raised against a 2,4-dinitrophenyl (DNP) target. Through its ability to adopt multiple conformations, the antibody is capable of binding to a diverse range of small haptens and large proteins. The present study examines a dataset of experimentally determined crystal structures of the SPE-7 antibody to gain insight into the mechanisms that contribute to its multispecificity. With the emergence of more and more therapeutic antibodies against a huge repertoire of different targets, our research could be of great interest for future drug development. We are able to discriminate between the different paratope-binding states in the conformational ensembles obtained by enhanced sampling molecular dynamics simulations, and to calculate their transition timescales and state probabilities. Furthermore, we describe the key residues responsible for discriminating between the different binding capacities and identify a tryptophan in a central position of the CDR L3 loop as the residue of greatest interest. The overall dynamics of the paratope appear to be mainly influenced by the CDR L3 and CDR L1 loops.
Collapse
Affiliation(s)
- Clarissa A Seidler
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Klaus R Liedl
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
Aguilar D, Zhu F, Millet A, Millet N, Germano P, Pisegna J, Akbari O, Doherty TA, Swidergall M, Jendzjowsky N. Sensory neurons regulate stimulus-dependent humoral immunity in mouse models of bacterial infection and asthma. Nat Commun 2024; 15:8914. [PMID: 39414787 PMCID: PMC11484968 DOI: 10.1038/s41467-024-53269-3] [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: 02/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
Sensory neurons sense pathogenic infiltration to drive innate immune responses, but their role in humoral immunity is unclear. Here, using mouse models of Streptococcus pneumoniae infection and Alternaria alternata asthma, we show that sensory neurons are required for B cell recruitment and antibody production. In response to S. pneumoniae, sensory neuron depletion increases bacterial burden and reduces B cell numbers, IgG release, and neutrophil stimulation. Meanwhile, during A. alternata-induced airway inflammation, sensory neuron depletion decreases B cell population sizes, IgE levels, and asthmatic characteristics. Mechanistically, during bacterial infection, sensory neurons preferentially release vasoactive intestinal polypeptide (VIP). In response to asthma, sensory neurons release substance P. Administration of VIP into sensory neuron-depleted mice suppresses bacterial burden, while VIPR1 deficiency increases infection. Similarly, exogenous substance P delivery aggravates asthma in sensory neuron-depleted mice, while substance P deficiency ameliorates asthma. Our data, thus demonstrate that sensory neurons release select neuropeptides which target B cells dependent on the immunogen.
Collapse
Affiliation(s)
- Diane Aguilar
- Division of Respiratory and Critical Care Medicine and Physiology, Harbor-UCLA Medical Center, Torrance, CA, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Fengli Zhu
- Division of Respiratory and Critical Care Medicine and Physiology, Harbor-UCLA Medical Center, Torrance, CA, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Antoine Millet
- Division of Respiratory and Critical Care Medicine and Physiology, Harbor-UCLA Medical Center, Torrance, CA, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Nicolas Millet
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Division of Infectious Disease, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Patrizia Germano
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA, USA
- Division of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph Pisegna
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System and Department of Medicine, Los Angeles, CA, USA
- Division of Pulmonary and Critical Care, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Taylor A Doherty
- Division of Allergy and Immunology, Department of Medicine, University of California San Diego, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Marc Swidergall
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Division of Infectious Disease, Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, Los Angeles, CA, USA
| | - Nicholas Jendzjowsky
- Division of Respiratory and Critical Care Medicine and Physiology, Harbor-UCLA Medical Center, Torrance, CA, USA.
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA.
- David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Mashima E, Saito-Sasaki N, Sawada Y. Systemic Implications of Bullous Pemphigoid: Bridging Dermatology and Internal Medicine. Diagnostics (Basel) 2024; 14:2272. [PMID: 39451595 PMCID: PMC11506695 DOI: 10.3390/diagnostics14202272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Bullous pemphigoid is an autoimmune bullous disease that frequently affects a large skin surface area, but it can also present in localized areas. It has been hypothesized that bullous pemphigoid affects the systemic functioning of different organs because inflammatory cells and cytokines circulate throughout numerous organs. Results: Recent clinical and experimental studies have revealed an association between bullous pemphigoid and systemic organ disorders. To avoid the emergence of systemic organ diseases, the significance of systemic treatment in cases of severe bullous pemphigoid should be emphasized. Conclusions: Here, we discuss the specific molecular processes underlying typical systemic organ inflammatory diseases associated with bullous pemphigoids.
Collapse
Affiliation(s)
| | | | - Yu Sawada
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (E.M.); (N.S.-S.)
| |
Collapse
|
10
|
Deguine J, Xavier RJ. B cell tolerance and autoimmunity: Lessons from repertoires. J Exp Med 2024; 221:e20231314. [PMID: 39093312 PMCID: PMC11296956 DOI: 10.1084/jem.20231314] [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: 04/25/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Adaptive immune cell function is regulated by a highly diverse receptor recombined from variable germline-encoded segments that can recognize an almost unlimited array of epitopes. While this diversity enables the recognition of any pathogen, it also poses a risk of self-recognition, leading to autoimmunity. Many layers of regulation are present during both the generation and activation of B cells to prevent this phenomenon, although they are evidently imperfect. In recent years, our ability to analyze immune repertoires at scale has drastically increased, both through advances in sequencing and single-cell analyses. Here, we review the current knowledge on B cell repertoire analyses, focusing on their implication for autoimmunity. These studies demonstrate that a failure of tolerance occurs at multiple independent checkpoints in different autoimmune contexts, particularly during B cell maturation, plasmablast differentiation, and within germinal centers. These failures are marked by distinct repertoire features that may be used to identify disease- or patient-specific therapeutic approaches.
Collapse
Affiliation(s)
- Jacques Deguine
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
| | - Ramnik J Xavier
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School , Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Siniscalco ER, Williams A, Eisenbarth SC. All roads lead to IgA: Mapping the many pathways of IgA induction in the gut. Immunol Rev 2024; 326:66-82. [PMID: 39046160 DOI: 10.1111/imr.13369] [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] [Indexed: 07/25/2024]
Abstract
The increasing prevalence of food allergy and related pathologies in recent years has underscored the need to understand the factors affecting adverse reactions to food. Food allergy is caused when food-specific IgE triggers the release of histamine from mast cells. However, other food-specific antibody isotypes exist as well, including IgG and IgA. IgA is the main antibody isotype in the gut and mediates noninflammatory reactions to toxins, commensal bacteria, and food antigens. It has also been thought to induce tolerance to food, thus antagonizing the role of food-specific IgE. However, this has remained unclear as food-specific IgA generation is poorly understood. Particularly, the location of IgA induction, the role of T cell help, and the fates of food-specific B cells remain elusive. In this review, we outline what is known about food-specific IgA induction and highlight areas requiring further study. We also explore how knowledge of food-specific IgA induction can be informed by and subsequently contribute to our overall knowledge of gut immunity.
Collapse
Affiliation(s)
- Emily R Siniscalco
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam Williams
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Allergy and Immunology, The Department Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Stephanie C Eisenbarth
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Allergy and Immunology, The Department Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
12
|
Rahman RS, Wesemann DR. Whence and wherefore IgE? Immunol Rev 2024; 326:48-65. [PMID: 39041740 PMCID: PMC11436312 DOI: 10.1111/imr.13373] [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] [Indexed: 07/24/2024]
Abstract
Despite the near ubiquitous presence of Ig-based antibodies in vertebrates, IgE is unique to mammals. How and why it emerged remains mysterious. IgE expression is greatly constrained compared to other IgH isotypes. While other IgH isotypes are relatively abundant, soluble IgE has a truncated half-life, and IgE plasma cells are mostly short-lived. Despite its rarity, IgE is consequential and can trigger life-threatening anaphylaxis. IgE production reflects a dynamic steady state with IgG memory B cells feeding short-lived IgE production. Emerging evidence suggests that IgE may also potentially be produced in longer-lived plasma cells as well, perhaps as an aberrancy stemming from its evolutionary roots from an antibody isotype that likely functioned more like IgG. As a late derivative of an ancient systemic antibody system, the benefits of IgE in mammals likely stems from the antibody system's adaptive recognition and response capability. However, the tendency for massive, systemic, and long-lived production, common to IgH isotypes like IgG, were likely not a good fit for IgE. The evolutionary derivation of IgE from an antibody system that for millions of years was good at antigen de-sensitization to now functioning as a highly specialized antigen-sensitization function required heavy restrictions on antibody production-insufficiency of which may contribute to allergic disease.
Collapse
Affiliation(s)
- Rifat S. Rahman
- Department of Internal Medicine, Columbia University Irving Medical Center, New York, NY
| | - Duane R. Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
13
|
Aguilar D, Zhu F, Millet A, Millet N, Germano P, Pisegna J, Akbari O, Doherty TA, Swidergall M, Jendzjowsky N. Sensory neurons regulate stimulus-dependent humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574231. [PMID: 38260709 PMCID: PMC10802321 DOI: 10.1101/2024.01.04.574231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Sensory neurons sense pathogenic infiltration, serving to inform immune coordination of host defense. However, sensory neuron-immune interactions have been predominantly shown to drive innate immune responses. Humoral memory, whether protective or destructive, is acquired early in life - as demonstrated by both early exposure to streptococci and allergic disease onset. Our study further defines the role of sensory neuron influence on humoral immunity in the lung. Using a murine model of Streptococcus pneumonia pre-exposure and infection and a model of allergic asthma, we show that sensory neurons are required for B-cell and plasma cell recruitment and antibody production. In response to S. pneumoniae, sensory neuron depletion resulted in a larger bacterial burden, reduced B-cell populations, IgG release and neutrophil stimulation. Conversely, sensory neuron depletion reduced B-cell populations, IgE and asthmatic characteristics during allergen-induced airway inflammation. The sensory neuron neuropeptide released within each model differed. With bacterial infection, vasoactive intestinal polypeptide (VIP) was preferentially released, whereas substance P was released in response to asthma. Administration of VIP into sensory neuron-depleted mice suppressed bacterial burden and increased IgG levels, while VIP1R deficiency increased susceptibility to bacterial infection. Sensory neuron-depleted mice treated with substance P increased IgE and asthma, while substance P genetic ablation resulted in blunted IgE, similar to sensory neuron-depleted asthmatic mice. These data demonstrate that the immunogen differentially stimulates sensory neurons to release specific neuropeptides which specifically target B-cells. Targeting sensory neurons may provide an alternate treatment pathway for diseases involved with insufficient and/or aggravated humoral immunity.
Collapse
|
14
|
Hilton LK, Collinge B, Ben-Neriah S, Alduaij W, Shaalan H, Weng AP, Cruz M, Slack GW, Farinha P, Miyata-Takata T, Boyle M, Meissner B, Cook JR, Ondrejka SL, Ott G, Rosenwald A, Campo E, Amador C, Greiner TC, Raess PW, Song JY, Inghirami G, Jaffe ES, Weisenburger DD, Chan WC, Beiske K, Fu K, Delabie J, Pittaluga S, Iqbal J, Wright G, Sehn LH, Savage KJ, Mungall AJ, Feldman AL, Staudt LM, Steidl C, Rimsza LM, Morin RD, Scott DW. Motive and opportunity: MYC rearrangements in high-grade B-cell lymphoma with MYC and BCL2 rearrangements (an LLMPP study). Blood 2024; 144:525-540. [PMID: 38701426 PMCID: PMC11307266 DOI: 10.1182/blood.2024024251] [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: 02/09/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
ABSTRACT Rearrangements that place the oncogenes MYC, BCL2, or BCL6 adjacent to superenhancers are common in mature B-cell lymphomas. Lymphomas with diffuse large B-cell lymphoma (DLBCL) or high-grade morphology with both MYC and BCL2 rearrangements are classified as high-grade B-cell lymphoma with MYC and BCL2 rearrangements ("double hit"; HGBCL-DH-BCL2) and are associated with aggressive disease and poor outcomes. Although it is established that MYC rearrangements involving immunoglobulin (IG) loci are associated with inferior outcomes relative to those involving other non-IG superenhancers, the frequency of and mechanisms driving IG vs non-IG MYC rearrangements have not been elucidated. Here, we used custom targeted capture and/or whole-genome sequencing to characterize oncogene rearrangements across 883 mature B-cell lymphomas including Burkitt lymphoma, follicular lymphoma, DLBCL, and HGBCL-DH-BCL2 tumors. We demonstrate that, although BCL2 rearrangement topology is consistent across entities, HGBCL-DH-BCL2 have distinct MYC rearrangement architecture relative to tumors with single MYC rearrangements or with both MYC and BCL6 rearrangements (HGBCL-DH-BCL6), including both a higher frequency of non-IG rearrangements and different architecture of MYC::IGH rearrangements. The distinct MYC rearrangement patterns in HGBCL-DH-BCL2 occur on the background of high levels of somatic hypermutation across MYC partner loci in HGBCL-DH-BCL2, creating more opportunity to form these rearrangements. Furthermore, because 1 IGH allele is already disrupted by the existing BCL2 rearrangement, the MYC rearrangement architecture in HGBCL-DH-BCL2 likely reflects selective pressure to preserve both BCL2 and B-cell receptor expression. These data provide new mechanistic explanations for the distinct patterns of MYC rearrangements observed across different lymphoma entities.
Collapse
Affiliation(s)
- Laura K. Hilton
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Brett Collinge
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | | | - Waleed Alduaij
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | - Haya Shaalan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Andrew P. Weng
- Terry Fox Laboratory, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Manuela Cruz
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Graham W. Slack
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Pedro Farinha
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | | | - Merrill Boyle
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | | | - James R. Cook
- Department of Clinical Pathology, Cleveland Clinic, Cleveland, OH
| | | | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | | | - Elias Campo
- Hematopathology Section, Hospital Clinic of Barcelona, Institut d’Investigaciones Biomediques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Catalina Amador
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Timothy C. Greiner
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Philipp W. Raess
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | - Joo Y. Song
- Department of Pathology, City of Hope, Duarte, CA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Elaine S. Jaffe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Dennis D. Weisenburger
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Wing C. Chan
- Department of Pathology, City of Hope, Duarte, CA
| | - Klaus Beiske
- Department of Pathology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kai Fu
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Jan Delabie
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Javeed Iqbal
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - George Wright
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laurie H. Sehn
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Kerry J. Savage
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Andrew J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew L. Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Lisa M. Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ
| | - Ryan D. Morin
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Bueno-Díaz C, Zuurveld M, Ayechu-Muruzabal V, Korsten SGPJ, Martín-Pedraza L, Parrón-Ballesteros J, Redegeld F, Garssen J, Villalba M, Willemsen LEM. Mustard seed major allergen Sin a1 activates intestinal epithelial cells and also dendritic cells that drive type 2 immune responses. Food Funct 2024; 15:6488-6501. [PMID: 38804660 DOI: 10.1039/d4fo01980f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mustard seeds belong to the food category of mandatory labelling due to the severe reactions they can trigger in allergic patients. However, the mechanisms underlying allergic sensitization to mustard seeds are poorly understood. The aim of this work is to study type 2 immune activation induced by the mustard seed major allergen Sin a1 via the intestinal mucosa, employing an in vitro model mimicking allergen exposure via the intestinal epithelial cells (IECs). Sin a1 was isolated from the total protein extract and exposed to IEC, monocyte derived dendritic cells (DCs) or IEC/DC co-cultures. A system of consecutive co-cultures was employed to study the generic capacity of Sin a1 to induce type 2 activation leading to sensitization: IEC/DC, DC/T-cell, T/B-cell and stem cell derived mast cells (MCs) derived from healthy donors. Immune profiles were determined by ELISA and flow cytometry. Sin a1 activated IEC and induced type-2 cytokine secretion in IEC/DC co-culture or DC alone (IL-15, IL-25 and TSLP), and primed DC induced type 2 T-cell skewing. IgG secretion in the T-cell/B-cell phase was enhanced in the presence of Sin a1 in the first stages of the co-culture. Anti-IgE did not induce degranulation but promoted IL-13 and IL-4 release by MC primed with the supernatant from B-cells co-cultured with Sin a1-IEC/DC or -DC primed T-cells. Sin a1 enhanced the release of type-2 inflammatory mediators by epithelial and dendritic cells; the latter instructed generic type-2 responses in T-cells that resulted in B-cell activation, and finally MC activation upon anti-IgE exposure. This indicates that via activation of IEC and/or DC, mustard seed allergen Sin a1 is capable of driving type 2 immunity which may lead to allergic sensitization.
Collapse
Affiliation(s)
- Cristina Bueno-Díaz
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| | - Marit Zuurveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| | - Verónica Ayechu-Muruzabal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| | - Sandra G P J Korsten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
- Tiofarma B.V., Oud-Beijerland, The Netherlands
| | | | - Jorge Parrón-Ballesteros
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Frank Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
- Danone Nutricia Research B.V., Utrecht, The Netherlands
| | - Mayte Villalba
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Linette E M Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
16
|
Tangye SG, Mackie J, Pathmanandavel K, Ma CS. The trajectory of human B-cell function, immune deficiency, and allergy revealed by inborn errors of immunity. Immunol Rev 2024; 322:212-232. [PMID: 37983844 DOI: 10.1111/imr.13288] [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] [Indexed: 11/22/2023]
Abstract
The essential role of B cells is to produce protective immunoglobulins (Ig) that recognize, neutralize, and clear invading pathogens. This results from the integration of signals provided by pathogens or vaccines and the stimulatory microenvironment within sites of immune activation, such as secondary lymphoid tissues, that drive mature B cells to differentiate into memory B cells and antibody (Ab)-secreting plasma cells. In this context, B cells undergo several molecular events including Ig class switching and somatic hypermutation that results in the production of high-affinity Ag-specific Abs of different classes, enabling effective pathogen neutralization and long-lived humoral immunity. However, perturbations to these key signaling pathways underpin immune dyscrasias including immune deficiency and autoimmunity or allergy. Inborn errors of immunity that disrupt critical immune pathways have identified non-redundant requirements for eliciting and maintaining humoral immune memory but concomitantly prevent immune dysregulation. Here, we will discuss our studies on human B cells, and how our investigation of cytokine signaling in B cells have identified fundamental requirements for memory B-cell formation, Ab production as well as regulating Ig class switching in the context of protective versus allergic immune responses.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Karrnan Pathmanandavel
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Ota M, Hoehn KB, Fernandes-Braga W, Ota T, Aranda CJ, Friedman S, Miranda-Waldetario MG, Redes J, Suprun M, Grishina G, Sampson HA, Malbari A, Kleinstein SH, Sicherer SH, de Lafaille MAC. CD23 +IgG1 + memory B cells are poised to switch to pathogenic IgE production in food allergy. Sci Transl Med 2024; 16:eadi0673. [PMID: 38324641 PMCID: PMC11008013 DOI: 10.1126/scitranslmed.adi0673] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/15/2023] [Indexed: 02/09/2024]
Abstract
Food allergy is caused by allergen-specific immunoglobulin E (IgE) antibodies, but little is known about the B cell memory of persistent IgE responses. Here, we describe, in human pediatric peanut allergy, a population of CD23+IgG1+ memory B cells arising in type 2 immune responses that contain high-affinity peanut-specific clones and generate IgE-producing cells upon activation. The frequency of CD23+IgG1+ memory B cells correlated with circulating concentrations of IgE in children with peanut allergy. A corresponding population of "type 2-marked" IgG1+ memory B cells was identified in single-cell RNA sequencing experiments. These cells differentially expressed interleukin-4 (IL-4)- and IL-13-regulated genes, such as FCER2/CD23+, IL4R, and germline IGHE, and carried highly mutated B cell receptors (BCRs). In children with high concentrations of serum peanut-specific IgE, high-affinity B cells that bind the main peanut allergen Ara h 2 mapped to the population of "type 2-marked" IgG1+ memory B cells and included clones with convergent BCRs across different individuals. Our findings indicate that CD23+IgG1+ memory B cells transcribing germline IGHE are a unique memory population containing precursors of high-affinity pathogenic IgE-producing cells that are likely to be involved in the long-term persistence of peanut allergy.
Collapse
Affiliation(s)
- Miyo Ota
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| | - Kenneth B. Hoehn
- Department of Pathology, Yale School of Medicine; New Haven, CT 06520, USA
| | - Weslley Fernandes-Braga
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| | - Takayuki Ota
- Department of Dermatology, Janssen Research & Development LLC; San Diego, CA 92121, USA
| | - Carlos J. Aranda
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| | - Sara Friedman
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| | - Mariana G.C. Miranda-Waldetario
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| | - Jamie Redes
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
- Graduate School of Biomedical Sciences, ISMMS; New York, NY 10029, USA
| | - Maria Suprun
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
| | - Galina Grishina
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
| | - Hugh A. Sampson
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
| | - Alefiyah Malbari
- Kravis Children’s Hospital, Department of Pediatrics, ISMMS; New York, NY 10029, USA
| | - Steven H. Kleinstein
- Department of Pathology, Yale School of Medicine; New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine; New Haven, CT 06520, USA
- Program in Computational Biology & Bioinformatics, Yale University; New Haven, CT 06511, USA
| | - Scott H. Sicherer
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
| | - Maria A. Curotto de Lafaille
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai (ISMMS); New York, NY 10029, USA
- Precision Immunology Institute (PrIISM), and Department of Immunology and Immunotherapy, ISMMS; New York, NY. 10029, USA
| |
Collapse
|
18
|
Huang R, Hu L, Jiang F. Study of cytokine-induced immunity in bullous pemphigoid: recent developments. Ann Med 2023; 55:2280991. [PMID: 38109924 PMCID: PMC10732206 DOI: 10.1080/07853890.2023.2280991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/04/2023] [Indexed: 12/20/2023] Open
Abstract
Bullous pemphigoid (BP) is an organ-specific disease. Its pathogenesis has not been clearly studied yet; However, studies in recent years have shown that its pathogenesis is related to T helper cells. The pathogenesis of BP is mainly related to Th2 and Th17-related cytokines. IL-4, IL-5 and IL-13 cause eosinophil recruitment, promote antibody production, trigger pruritus and promote blister formation and other symptoms. IL-17 and IL-23 promote the production of matrix metalloproteinase-9 (MMP-9) by related cells, which causes dermo-epidermal junction (DEJ) separation to form bullae and blisters, and can persist in BP inflammation. The serum concentrations of IL-17 and IL-23 are related to the prognosis of BP. In this paper, we focus on the role of related cytokines in the pathogenesis of bullous pemphigoid and the relationship between the related cytokine populations secreted by three major T helper cells-helper T lymphocytes 1 (Th1), Th2, and Th17. A better understanding of the biological and immunological functions of cytokines associated with BP patients will provide opportunities for therapeutic targets in BP.
Collapse
Affiliation(s)
- Ruiting Huang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lingyu Hu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fuqiong Jiang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
19
|
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, Orengo JM. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy. Sci Transl Med 2023; 15:eadf9561. [PMID: 38091405 DOI: 10.1126/scitranslmed.adf9561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Anita Boyapati
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - John Janczy
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Pablo Abreu
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Wen-Chi Chen
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Stephen Godin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Harvey Chin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
20
|
Udoye CC, Ehlers M, Manz RA. The B Cell Response and Formation of Allergenic and Anti-Allergenic Antibodies in Food Allergy. BIOLOGY 2023; 12:1501. [PMID: 38132327 PMCID: PMC10740584 DOI: 10.3390/biology12121501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Food allergies are a growing public health concern worldwide, especially in children and young adults. Allergen-specific IgE plays a central role in the pathogenesis of food allergies, but their titers poorly correlate with allergy development. Host immune systems yield allergen-specific immunoglobulin (Ig)A, IgE and IgG subclasses with low or high affinities and differential Fc N-glycosylation patterns that can affect the allergic reaction to food in multiple ways. High-affinity IgE is required to induce strong mast cell activation eventually leading to allergic anaphylaxis, while low-affinity IgE can even inhibit the development of clinically relevant allergic symptoms. IgA and IgG antibodies can inhibit IgE-mediated mast cell activation through various mechanisms, thereby protecting IgE-positive individuals from allergy development. The production of IgE and IgG with differential allergenic potential seems to be affected by the signaling strength of individual B cell receptors, and by cytokines from T cells. This review provides an overview of the diversity of the B cell response and the diverse roles of antibodies in food allergy.
Collapse
Affiliation(s)
- Christopher C. Udoye
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, 23538 Lübeck, Germany
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
21
|
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
|
22
|
Kawakami Y, Takazawa I, Fajt ML, Kasakura K, Lin J, Ferrer J, Kantor DB, Phipatanakul W, Heymann PW, Benedict CA, Kawakami Y, Kawakami T. Histamine-releasing factor in severe asthma and rhinovirus-associated asthma exacerbation. J Allergy Clin Immunol 2023; 152:633-640.e4. [PMID: 37301412 PMCID: PMC10917146 DOI: 10.1016/j.jaci.2023.04.021] [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: 11/01/2022] [Revised: 04/07/2023] [Accepted: 04/28/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Histamine-releasing factor (HRF) is implicated in allergic diseases. We previously showed its pathogenic role in murine models of asthma. OBJECTIVE We aim to present data analysis from 3 separate human samples (sera samples from asthmatic patients, nasal washings from rhinovirus [RV]-infected individuals, and sera samples from patients with RV-induced asthma exacerbation) and 1 mouse sample to investigate correlates of HRF function in asthma and virus-induced asthma exacerbations. METHODS Total IgE and HRF-reactive IgE/IgG as well as HRF in sera from patients with mild/moderate asthma or severe asthma (SA) and healthy controls (HCs) were quantified by ELISA. HRF secretion in culture media from RV-infected adenovirus-12 SV40 hybrid virus transformed human bronchial epithelial cells and in nasal washings from experimentally RV-infected subjects was analyzed by Western blotting. HRF-reactive IgE/IgG levels in longitudinal serum samples from patients with asthma exacerbations were also quantified. RESULTS HRF-reactive IgE and total IgE levels were higher in patients with SA than in HCs, whereas HRF-reactive IgG (and IgG1) level was lower in asthmatic patients versus HCs. In comparison with HRF-reactive IgElow asthmatic patients, HRF-reactive IgEhigh asthmatic patients had a tendency to release more tryptase and prostaglandin D2 on anti-IgE stimulation of bronchoalveolar lavage cells. RV infection induced HRF secretion from adenovirus-12 SV40 hybrid virus transformed bronchial epithelial cells, and intranasal RV infection of human subjects induced increased HRF secretion in nasal washes. Asthmatic patients had higher levels of HRF-reactive IgE at the time of asthma exacerbations associated with RV infection, compared with those after the resolution. This phenomenon was not seen in asthma exacerbations without viral infections. CONCLUSIONS HRF-reactive IgE is higher in patients with SA. RV infection induces HRF secretion from respiratory epithelial cells both in vitro and in vivo. These results suggest the role of HRF in asthma severity and RV-induced asthma exacerbation.
Collapse
Affiliation(s)
- Yu Kawakami
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - Ikuo Takazawa
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - Merritt L Fajt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Kazumi Kasakura
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - Joseph Lin
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - Julienne Ferrer
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - David B Kantor
- Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Children's Hospital, Boston, Mass
| | | | - Peter W Heymann
- Asthma and Allergic Diseases Center, University of Virginia, Charlottsville, Va; Division of Pediatric Respiratory Medicine, University of Virginia, Charlottsville, Va
| | - Chris A Benedict
- Benedict Laboratory, Center for Autoimmunity and Inflammation and Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, Calif
| | - Yuko Kawakami
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif
| | - Toshiaki Kawakami
- Laboratory of Allergic Diseases, La Jolla Institute for Immunology, La Jolla, Calif.
| |
Collapse
|
23
|
Chakma CR, Good-Jacobson KL. Requirements of IL-4 during the Generation of B Cell Memory. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1853-1860. [PMID: 37276051 DOI: 10.4049/jimmunol.2200922] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/02/2023] [Indexed: 06/07/2023]
Abstract
IL-4 has long been established as a key regulator of Th cells and for promoting effective B cell survival and isotype class switching. Yet, despite having been extensively studied, the specific role of IL-4 in generating humoral memory in vivo is unclear. In this review, we explore the recent studies that unravel the cellular sources and spatiotemporal production of IL-4, the relationship between IL-4 and IL-21 during germinal center responses and the formation of Ab-secreting cells, and the current understanding of whether IL-4 promotes or suppresses memory B cell generation in vitro and in vivo.
Collapse
Affiliation(s)
- Clarissa R Chakma
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Strobl MR, Demir H, Sánchez Acosta G, Drescher A, Kitzmüller C, Möbs C, Pfützner W, Bohle B. The role of IgG 1 and IgG 4 as dominant IgE-blocking antibodies shifts during allergen immunotherapy. J Allergy Clin Immunol 2023; 151:1371-1378.e5. [PMID: 36657603 DOI: 10.1016/j.jaci.2023.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND The induction of allergen-specific IgE-blocking antibodies is a hallmark of allergen immunotherapy (AIT). The inhibitory bioactivity has largely been attributed to IgG4; however, our recent studies indicated the dominance of IgG1 early in AIT. OBJECTIVES Here, the IgE-blocking activity and avidity of allergen-specific IgG1 and IgG4 antibodies were monitored throughout 3 years of treatment. METHODS Serum samples from 24 patients were collected before and regularly during AIT with birch pollen. Bet v 1-specific IgG1 and IgG4 levels were determined by ELISA and ImmunoCAP, respectively. Unmodified and IgG1- or IgG4-depleted samples were compared for their inhibition of Bet v 1-induced basophil activation. The stability of Bet v 1-antibody complexes was compared by ELISA and by surface plasmon resonance. RESULTS Bet v 1-specific IgG1 and IgG4 levels peaked at 12 and 24 months of AIT, respectively. Serological IgE-blocking peaked at 6 months and remained high thereafter. In the first year of therapy, depletion of IgG1 clearly diminished the inhibition of basophil activation while the absence of IgG4 hardly reduced IgE-blocking. Then, IgG4 became the main inhibitory isotype in most individuals. Both isotypes displayed high avidity to Bet v 1 ab initio of AIT, which did not increase during treatment. Bet v 1-IgG1 complexes were enduringly more stable than Bet v 1-IgG4 complexes were. CONCLUSIONS In spite of the constant avidity of AIT-induced allergen-specific IgG1 and IgG4 antibodies, their dominance in IgE-blocking shifted in the course of treatment. The blocking activity of allergen-specific IgG1 should not be underestimated, particularly early in AIT.
Collapse
Affiliation(s)
- Maria R Strobl
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hilal Demir
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gabriela Sánchez Acosta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Anja Drescher
- Cytiva Europe GmbH, Freiburg, Department of Dermatology and Allergology, Philipps-Universität Marburg, Freiburg and Marburg, Germany
| | - Claudia Kitzmüller
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Möbs
- Clinical and Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Wolfgang Pfützner
- Clinical and Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Barbara Bohle
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
Rivera CE, Zhou Y, Chupp DP, Yan H, Fisher AD, Simon R, Zan H, Xu Z, Casali P. Intrinsic B cell TLR-BCR linked coengagement induces class-switched, hypermutated, neutralizing antibody responses in absence of T cells. SCIENCE ADVANCES 2023; 9:eade8928. [PMID: 37115935 PMCID: PMC10146914 DOI: 10.1126/sciadv.ade8928] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Maturation of antibody responses entails somatic hypermutation (SHM), class-switch DNA recombination (CSR), plasma cell differentiation, and generation of memory B cells, and it is thought to require T cell help. We showed that B cell Toll-like receptor 4 (TLR4)-B cell receptor (BCR) (receptor for antigen) coengagement by 4-hydroxy-3-nitrophenyl acetyl (NP)-lipopolysaccharide (LPS) (Escherichia coli lipid A polysaccharide O-antigen) or TLR5-BCR coengagement by Salmonella flagellin induces mature antibody responses to NP and flagellin in Tcrβ-/-Tcrδ-/- and NSG/B mice. TLR-BCR coengagement required linkage of TLR and BCR ligands, "linked coengagement." This induced B cell CSR/SHM, germinal center-like differentiation, clonal expansion, intraconal diversification, plasma cell differentiation, and an anamnestic antibody response. In Tcrβ-/-Tcrδ-/- mice, linked coengagement of TLR4-BCR by LPS or TLR5-BCR by flagellin induced protective antibodies against E. coli or Salmonella Typhimurium. Our findings unveiled a critical role of B cell TLRs in inducing neutralizing antibody responses, including those to microbial pathogens, without T cell help.
Collapse
Affiliation(s)
- Carlos E. Rivera
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Yulai Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Daniel P. Chupp
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Hui Yan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Amanda D. Fisher
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Raphael Simon
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hong Zan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Zhenming Xu
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Paolo Casali
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
- Department of Medicine, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
26
|
Berin MC. Targeting type 2 immunity and the future of food allergy treatment. J Exp Med 2023; 220:213917. [PMID: 36880703 PMCID: PMC9997511 DOI: 10.1084/jem.20221104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 03/08/2023] Open
Abstract
IgE-mediated food allergy affects 6-8% of the population in the United States. Type 2 immune responses are central to the pathogenesis of food allergy, but type 2 CD4+ T cell responses have been found to be heterogeneous in food allergy suggesting a division of labor between Tfh13 and peTH2 cells in promotion of IgE class switching, modulation of intestinal barrier function, and regulation of mast cell expansion. Oral immunotherapy for the treatment of food allergy incompletely targets subsets of type 2 immunity in a transient manner, but new therapeutics targeting different levels of type 2 immunity are in current or planned trials for food allergy. These new treatments and the basis for their use are the focus of this review.
Collapse
Affiliation(s)
- M Cecilia Berin
- Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| |
Collapse
|
27
|
Chen Q, Liu H, Luling N, Reinke J, Dent AL. Evidence that High-Affinity IgE Can Develop in the Germinal Center in the Absence of an IgG1-Switched Intermediate. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:905-915. [PMID: 36779803 PMCID: PMC10038918 DOI: 10.4049/jimmunol.2200521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/12/2023] [Indexed: 02/14/2023]
Abstract
High-affinity allergen-specific IgE is essential for the severe allergic anaphylaxis response. High-affinity Abs are formed by successive rounds of selection of Ag-specific B cells in the germinal center (GC); however, several studies have shown that IgE+ GC B cells are impaired in their ability to undergo selection in the GC. A pathway, known as the "indirect switching pathway" for IgE, has been described whereby Ag-specific B cells initially switch to the IgG1 isotype and undergo affinity selection in the GC, with a secondary switch to the IgE isotype after affinity selection. In previous work, using a food allergy model in mice, we investigated how high-affinity IgE develops in the GC, but we did not test the indirect switching model. In this study, we analyzed the importance of the indirect switching pathway by constructing IgG1-cre Bcl6-fl/fl mice. In these mice, once B cells switch to IgG1, they delete Bcl6 and thus cannot enter or persist in the GC. When we tested IgG1-cre Bcl6-fl/fl mice with our food allergy model, we found that, as expected, IgG1 Abs had decreased affinity, but unexpectedly, the affinity of IgE for allergen was unchanged. IgG1-cre Bcl6-fl/fl mice underwent anaphylaxis in response to allergen, consistent with the formation of high-affinity IgE. Thus, in a food allergy response, high-affinity IgE can be efficiently formed in the absence of indirect switching to IgG1, either by direct selection of IgE+ GC B cells or indirect selection of IgM+ GC B cells that later switch to IgE.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Hong Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Noelle Luling
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Julia Reinke
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
28
|
Adam S, Dekumyoy P, Nacapunchai D, Ketboonlue T, Charunwatthana P, Dhitavat J, Koompapong K, Chonsawat P, Watthanakulpanich D. Assessment of an Immuno-Diagnostic Method for Hookworm-Related Cutaneous Larva Migrans Using Crude Extracts of Ancylostoma caninum. Trop Med Infect Dis 2023; 8:tropicalmed8040209. [PMID: 37104335 PMCID: PMC10142037 DOI: 10.3390/tropicalmed8040209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
People can become infected with cutaneous larva migrans (CLM) through skin penetration by the infective zoonotic larvae of hookworms. Few studies have investigated CLM’s immunodiagnosis, and the existing studies were limited to crude somatic or excretory/secretory antigens (Ags) from adult worms. Here, we aimed to develop an indirect enzyme-linked immunosorbent assay (ELISA) to differentiate and diagnose hwCLM by detecting immunoglobulin (Ig)E, IgG, and IgG subclasses 1–4 (IgG1–4) against the somatic Ag of adult Ancylostoma caninum checkerboard titrations of adult A. caninum worm extract. Pooled serum controls were immunocharacterized using an indirect ELISA. The IgG1–4 and IgE results were unsatisfactory; however, the use of total IgG achieved results comparable to those of immunoblotting. Thus, we continued to analyze the IgG-ELISA using serum samples from patients with hwCLM and heterologous infections as well as from healthy controls. The sensitivity and excellent specificity of the total IgG-ELISA were 93.75% and 98.37%, respectively, and its positive and negative predictive values were 75% and 99.67%, respectively. Antibodies from five cases of angiostrongyliasis, gnathostomiasis, and dirofilariasis cross-reacted with the somatic Ag of adult A. caninum. This new assay can adequately serodiagnose hwCLM when combined with clinical features and/or histological examination.
Collapse
Affiliation(s)
- Sitthithana Adam
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Paron Dekumyoy
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Duangporn Nacapunchai
- College of Allied Health Sciences, Suan Sunandha Rajabhat University, Bangkok 10300, Thailand
| | - Thawatchai Ketboonlue
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Prakaykaew Charunwatthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Jittima Dhitavat
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Khuanchai Koompapong
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Putza Chonsawat
- Hospital for Tropical Diseases, Diagnostic Laboratory Unit, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Dorn Watthanakulpanich
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
- Correspondence: ; Tel.: +66-662-643-5600
| |
Collapse
|
29
|
Aranda CJ, Gonzalez-Kozlova E, Saunders SP, Fernandes-Braga W, Ota M, Narayanan S, He JS, Del Duca E, Swaroop B, Gnjatic S, Shattner G, Reibman J, Soter NA, Guttman-Yassky E, Curotto de Lafaille MA. IgG memory B cells expressing IL4R and FCER2 are associated with atopic diseases. Allergy 2023; 78:752-766. [PMID: 36445014 PMCID: PMC9991991 DOI: 10.1111/all.15601] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/25/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Atopic diseases are characterized by IgE antibody responses that are dependent on cognate CD4 T cell help and T cell-produced IL-4 and IL-13. Current models of IgE cell differentiation point to the role of IgG memory B cells as precursors of pathogenic IgE plasma cells. The goal of this work was to identify intrinsic features of memory B cells that are associated with IgE production in atopic diseases. METHODS Peripheral blood B lymphocytes were collected from individuals with physician diagnosed asthma or atopic dermatitis (AD) and from non-atopic individuals. These samples were analyzed by spectral flow cytometry, single cell RNA sequencing (scRNAseq), and in vitro activation assays. RESULTS We identified a novel population of IgG memory B cells characterized by the expression of IL-4/IL-13 regulated genes FCER2/CD23, IL4R, IL13RA1, and IGHE, denoting a history of differentiation during type 2 immune responses. CD23+ IL4R+ IgG+ memory B cells had increased occurrence in individuals with atopic disease. Importantly, the frequency of CD23+ IL4R+ IgG+ memory B cells correlated with levels of circulating IgE. Consistently, in vitro stimulated B cells from atopic individuals generated more IgE+ cells than B cells from non-atopic subjects. CONCLUSIONS These findings suggest that CD23+ IL4R+ IgG+ memory B cells transcribing IGHE are potential precursors of IgE plasma cells and are linked to pathogenic IgE production.
Collapse
Affiliation(s)
- Carlos J Aranda
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | | | - Sean P Saunders
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Weslley Fernandes-Braga
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | - Miyo Ota
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| | - Sriram Narayanan
- Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Jin-Shu He
- Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Ester Del Duca
- Department of Dermatology, ISMMS, New York, New York, USA
| | - Bose Swaroop
- Department of Dermatology, ISMMS, New York, New York, USA
| | - Sacha Gnjatic
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
- Tisch Cancer Institute, ISMMS, New York, New York, USA
| | - Gail Shattner
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Joan Reibman
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | | | | | - Maria A Curotto de Lafaille
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
- Precision Immunology Institute (PrIISM), ISMMS, New York, New York, USA
| |
Collapse
|
30
|
Chen Z, Cui Y, Yao Y, Liu B, Yunis J, Gao X, Wang N, Cañete PF, Tuong ZK, Sun H, Wang H, Yang S, Wang R, Leong YA, Simon Davis D, Qin J, Liang K, Deng J, Wang CK, Huang YH, Roco JA, Nettelfield S, Zhu H, Xu H, Yu Z, Craik D, Liu Z, Qi H, Parish C, Yu D. Heparan sulfate regulates IL-21 bioavailability and signal strength that control germinal center B cell selection and differentiation. Sci Immunol 2023; 8:eadd1728. [PMID: 36800411 DOI: 10.1126/sciimmunol.add1728] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
In antibody responses, mutated germinal center B (BGC) cells are positively selected for reentry or differentiation. As the products from GCs, memory B cells and antibody-secreting cells (ASCs) support high-affinity and long-lasting immunity. Positive selection of BGC cells is controlled by signals received through the B cell receptor (BCR) and follicular helper T (TFH) cell-derived signals, in particular costimulation through CD40. Here, we demonstrate that the TFH cell effector cytokine interleukin-21 (IL-21) joins BCR and CD40 in supporting BGC selection and reveal that strong IL-21 signaling prioritizes ASC differentiation in vivo. BGC cells, compared with non-BGC cells, show significantly reduced IL-21 binding and attenuated signaling, which is mediated by low cellular heparan sulfate (HS) sulfation. Mechanistically, N-deacetylase and N-sulfotransferase 1 (Ndst1)-mediated N-sulfation of HS in B cells promotes IL-21 binding and signal strength. Ndst1 is down-regulated in BGC cells and up-regulated in ASC precursors, suggesting selective desensitization to IL-21 in BGC cells. Thus, specialized biochemical regulation of IL-21 bioavailability and signal strength sets a balance between the stringency and efficiency of GC selection.
Collapse
Affiliation(s)
- Zhian Chen
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Yanfang Cui
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, Central China Normal University, Wuhan, China
| | - Yin Yao
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Department of Otolaryngology-Head and Neck Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing, China
| | - Joseph Yunis
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Xin Gao
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Naiqi Wang
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Pablo F Cañete
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.,Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Hongjian Sun
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Hao Wang
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Siling Yang
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Runli Wang
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Yew Ann Leong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - David Simon Davis
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jiahuan Qin
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaili Liang
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Deng
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Conan K Wang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD, Australia
| | - Yen-Hua Huang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Jonathan A Roco
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Sam Nettelfield
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Huaming Zhu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Zhijia Yu
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - David Craik
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD, Australia
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing, China
| | - Christopher Parish
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
31
|
Simonin EM, Babasyan S, Tarsillo J, Wagner B. IgE+ plasmablasts predict the onset of clinical allergy. Front Immunol 2023; 14:1104609. [PMID: 36817463 PMCID: PMC9932261 DOI: 10.3389/fimmu.2023.1104609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction IgE+ plasmablasts develop following allergen exposure and B cell activation. They secrete IgE and therefore are directly linked to maintain the mechanisms of IgE-mediated allergies. Here, we show that the presence of IgE+ plasmablasts in peripheral blood not only coincides with clinical allergy, but also predicts the upcoming development of clinical disease. Methods Using an equine model of naturally occurring allergy, we compared the timing of allergen exposure, arrival of IgE+ plasmablasts in peripheral blood, and onset of clinical disease. Results We found that IgE+ plasmablasts predict the development of clinical allergy by at least 3 weeks and can be measured directly by flow cytometry or by IgE secretion following in vitro culture. We also compared the IgE secretion by IgE+ plasmablasts with total plasma IgE concentrations and found that while IgE secretion consistently correlates with clinical allergy, total plasma IgE does not. Discussion Together, we describe IgE+ plasmablasts as a reliable and sensitive predictive biomarker of allergic disease development.
Collapse
Affiliation(s)
| | | | - Justine Tarsillo
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | | |
Collapse
|
32
|
Ota M, Hoehn KB, Ota T, Aranda CJ, Friedman S, Braga WF, Malbari A, Kleinstein SH, Sicherer SH, Curotto de Lafaille MA. The memory of pathogenic IgE is contained within CD23 + IgG1 + memory B cells poised to switch to IgE in food allergy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525506. [PMID: 36747707 PMCID: PMC9900782 DOI: 10.1101/2023.01.25.525506] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Food allergy is caused by allergen-specific IgE antibodies but little is known about the B cell memory of persistent IgE responses. Here we describe in human pediatric peanut allergy CD23 + IgG1 + memory B cells arising in type 2 responses that contain peanut specific clones and generate IgE cells on activation. These 'type2-marked' IgG1 + memory B cells differentially express IL-4/IL-13 regulated genes FCER2 / CD23, IL4R , and germline IGHE and carry highly mutated B cell receptors (BCRs). Further, high affinity memory B cells specific for the main peanut allergen Ara h 2 mapped to the population of 'type2-marked' IgG1 + memory B cells and included convergent BCRs across different individuals. Our findings indicate that CD23 + IgG1 + memory B cells transcribing germline IGHE are a unique memory population containing precursors of pathogenic IgE. One-Sentence Summary We describe a unique population of IgG + memory B cells poised to switch to IgE that contains high affinity allergen-specific clones in peanut allergy.
Collapse
|
33
|
Olewicz-Gawlik A, Kowala-Piaskowska A. Self-reactive IgE and anti-IgE therapy in autoimmune diseases. Front Pharmacol 2023; 14:1112917. [PMID: 36755957 PMCID: PMC9899859 DOI: 10.3389/fphar.2023.1112917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Growing evidence indicates the pathogenic role of autoreactive IgE in autoimmune diseases. Incidence of autoimmune and allergic diseases in the industrialized countries is consistently icreasing, thus leading to concerted efforts to comprehend the regulation of IgE-mediated mechanisms. The first reports of a presence of IgE autoantibodies in patients with autoimmune diseases have been published a long time ago, and it is now recognized that self-reactive IgE can mediate inflammatory response in bullous pemhigoid, systemic lupus erythematosus, chronic urticaria, and atopic dermatitis. The advances in understanding the pathomechanisms of these disorders brought to a successful use of anti-IgE strategies in their management. The present review discusses the current state of knowledge on the IgE-mediated autoimmunity and anti-IgE treatment, and pave the way for further exploration of the subject.
Collapse
Affiliation(s)
- Anna Olewicz-Gawlik
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland,Department of Infectious Diseases, Hepatology and Acquired Immunodeficiencies, Poznan University of Medical Sciences, Poznan, Poland,*Correspondence: Anna Olewicz-Gawlik,
| | - Arleta Kowala-Piaskowska
- Department of Infectious Diseases, Hepatology and Acquired Immunodeficiencies, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
34
|
LaHood NA, Min J, Keswani T, Richardson CM, Amoako K, Zhou J, Marini-Rapoport O, Bernard H, Hazebrouck S, Shreffler WG, Love JC, Pomes A, Pedersen LC, Mueller GA, Patil SU. Immunotherapy-induced neutralizing antibodies disrupt allergen binding and sustain allergen tolerance in peanut allergy. J Clin Invest 2023; 133:e164501. [PMID: 36647835 PMCID: PMC9843057 DOI: 10.1172/jci164501] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/15/2022] [Indexed: 01/18/2023] Open
Abstract
In IgE-mediated food allergies, exposure to the allergen activates systemic allergic responses. Oral immunotherapy (OIT) treats food allergies through incremental increases in oral allergen exposure. However, OIT only induces sustained clinical tolerance and decreased basophil sensitivity in a subset of individuals despite increases in circulating allergen-specific IgG in all treated individuals. Therefore, we examined the allergen-specific antibodies from 2 OIT cohorts of patients with sustained and transient responses. Here, we compared antibodies from individuals with sustained or transient responses and discovered specific tolerance-associated conformational epitopes of the immunodominant allergen Ara h 2 recognized by neutralizing antibodies. First, we identified what we believe to be previously unknown conformational, intrahelical epitopes using x-ray crystallography with recombinant antibodies. We then identified epitopes only recognized in sustained tolerance. Finally, antibodies recognizing tolerance-associated epitopes effectively neutralized allergen to suppress IgE-mediated effector cell activation. Our results demonstrate the molecular basis of antibody-mediated protection in IgE-mediated food allergy, by defining how these antibodies disrupt IgE-allergen interactions to prevent allergic reactions. Our approach to studying the structural and functional basis for neutralizing antibodies demonstrates the clinical relevance of specific antibody clones in antibody-mediated tolerance. We anticipate that our findings will form the foundation for treatments of peanut allergy using neutralizing antibodies and hypoallergens.
Collapse
Affiliation(s)
- Nicole A. LaHood
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jungki Min
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Tarun Keswani
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Kwasi Amoako
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jingjia Zhou
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Hervé Bernard
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Stéphane Hazebrouck
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Wayne G. Shreffler
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - J. Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Lars C. Pedersen
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Geoffrey A. Mueller
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Sarita U. Patil
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Allergenic food protein consumption is associated with systemic IgG antibody responses in non-allergic individuals. Immunity 2022; 55:2454-2469.e6. [PMID: 36473469 DOI: 10.1016/j.immuni.2022.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/01/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Although food-directed immunoglobulin E (IgE) has been studied in the context of allergies, the prevalence and magnitude of IgG responses against dietary antigens are incompletely characterized in the general population. Here, we measured IgG binding against food and environmental antigens obtained from allergen databases and the immune epitope database (IEDB), represented in a phage displayed library of 58,233 peptides. By profiling blood samples of a large cohort representing the average adult Israeli population (n = 1,003), we showed that many food antigens elicited systemic IgG in up to 50% of individuals. Dietary intake of specific food protein correlated with antibody binding, suggesting that diet can shape the IgG epitope repertoire. Our work documents abundant systemic IgG responses against food antigens and provides a reference map of the exact immunogenic epitopes on a population scale, laying the foundation to unravel the role of food- and environmental antigen-directed antibody binding in disease contexts.
Collapse
|
36
|
Cai S, Chen Y, Hu Z, Zhou T, Huang Y, Lin S, Gao R, Zhong J, Dong L. The landscape of T and B lymphocytes interaction and synergistic effects of Th1 and Th2 type response in the involved tissue of IgG4-RD revealed by single cell transcriptome analysis. J Autoimmun 2022; 133:102944. [PMID: 36401985 DOI: 10.1016/j.jaut.2022.102944] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/23/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To investigate the landscape of T-B cell interaction, immune receptor profiles and effects of different types of immune responses in the involved tissues of IgG4-RD. METHODS Single cell RNA sequencing, bulk sample RNA sequencing, immune receptor repertoire analysis (both BCR and TCR), multi-color flow cytometry, and in-vitro assays with model cells (e.g. EBV-immortalized B cells from IgG4-RD patient) and histologic methods were applied to investigate the immunopathological features of IgG4-RD from multiple aspects. RESULTS Ectopic germinal center formation was observed in IgG4-RD patients at advanced disease stage, and a large part of B cells in involved tissue were germinal center B cell-like. Germinal center reaction in IgG4-RD led to the irregularities of both TCR and BCR clones in the involved tissues, and limited clonal overlaps among different samples. Enhanced Th1- and Th2-type responses were observed in involved tissues of IgG4-RD and patients with both increased Th1- and Th2-type response related cell subsets possessed more severe inflammatory indices. Analyses to the origin of IGHG4 transcripts in IgG4-RD indicated that IgG4 could be switched from IgM directly, or from other IgG subclasses. In vitro assays with EBV-immortalized B cells, fibroblasts and epithelial cells revealed the effects of Th1-type and Th2-type responses on germinal center reaction, ectopic expression of MHC-II molecules, and formation of tertiary lymphoid structures. CONCLUSIONS Synergistic effects of Th1- and Th2-type responses were involved in the pathogenesis of IgG4-RD via their influences on both acute inflammatory processes and the chronicity and complexity of IgG4-RD.
Collapse
Affiliation(s)
- Shaozhe Cai
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Hu
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Zhou
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yanli Huang
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shengyan Lin
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Rongfen Gao
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
37
|
Lim J, Lin EV, Hong JY, Vaidyanathan B, Erickson SA, Annicelli C, Medzhitov R. Induction of natural IgE by glucocorticoids. J Exp Med 2022; 219:213459. [PMID: 36098746 PMCID: PMC9475297 DOI: 10.1084/jem.20220903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/24/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
IgE mediates allergic responses by coating mast cell or basophil surfaces and inducing degranulation upon binding a specific allergen. IgE can also be spontaneously produced in the absence of foreign allergens; yet the origin, regulation, and functions of such "natural" IgE still remain largely unknown. Here, we find that glucocorticoids enhance the production of IgE in B cells both in vivo and ex vivo without antigenic challenge. Such IgE production is promoted by B cell-intrinsic glucocorticoid receptor signaling that reinforces CD40 signaling and synergizes with the IL-4/STAT6 pathway. In addition, we found that rare B cells in the mesenteric lymph nodes are responsible for the production of glucocorticoid-inducible IgE. Furthermore, locally produced glucocorticoids in the gut may induce natural IgE during perturbations of gut homeostasis, such as dysbiosis. Notably, mice preemptively treated with glucocorticoids were protected from subsequent pathogenic anaphylaxis. Together, our results suggest that glucocorticoids, classically considered to be broadly immunosuppressive, have a selective immunostimulatory role in B cells.
Collapse
Affiliation(s)
- Jaechul Lim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Erica V. Lin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Jun Young Hong
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea,Jun Young Hong:
| | - Bharat Vaidyanathan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Steven A. Erickson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Charles Annicelli
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT,Correspondence to Ruslan Medzhitov:
| |
Collapse
|
38
|
The role of chromatin loop extrusion in antibody diversification. Nat Rev Immunol 2022; 22:550-566. [PMID: 35169260 PMCID: PMC9376198 DOI: 10.1038/s41577-022-00679-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 12/15/2022]
Abstract
Cohesin mediates chromatin loop formation across the genome by extruding chromatin between convergently oriented CTCF-binding elements. Recent studies indicate that cohesin-mediated loop extrusion in developing B cells presents immunoglobulin heavy chain (Igh) variable (V), diversity (D) and joining (J) gene segments to RAG endonuclease through a process referred to as RAG chromatin scanning. RAG initiates V(D)J recombinational joining of these gene segments to generate the large number of different Igh variable region exons that are required for immune responses to diverse pathogens. Antigen-activated mature B cells also use chromatin loop extrusion to mediate the synapsis, breakage and end joining of switch regions flanking Igh constant region exons during class-switch recombination, which allows for the expression of different antibody constant region isotypes that optimize the functions of antigen-specific antibodies to eliminate pathogens. Here, we review recent advances in our understanding of chromatin loop extrusion during V(D)J recombination and class-switch recombination at the Igh locus.
Collapse
|
39
|
Cerutti A, Filipska M, Fa XM, Tachó-Piñot R. Impact of the mucosal milieu on antibody responses to allergens. J Allergy Clin Immunol 2022; 150:503-512. [PMID: 36075636 DOI: 10.1016/j.jaci.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
Respiratory and digestive mucosal surfaces are continually exposed to common environmental antigens, which include potential allergens. Although innocuous in healthy individuals, allergens cause allergy in predisposed subjects and do so by triggering a pathologic TH2 cell response that induces IgE class switching and somatic hypermutation in allergen-specific B cells. The ensuing affinity maturation and plasma cell differentiation lead to the abnormal release of high-affinity IgE that binds to powerful FcεRI receptors on basophils and mast cells. When cross-linked by allergen, FcεRI-bound IgE instigates the release of prestored and de novo-induced proinflammatory mediators. Aside from causing type I hypersensitivity reactions underlying allergy, IgE affords protection against nematodes or venoms from insects and snakes, which raises questions as to the fundamental differences between protective and pathogenic IgE responses. In this review, we discuss the impact of the mucosal environment, including the epithelial and mucus barriers, on the induction of protective IgE responses against environmental antigens. We further discuss how perturbations of these barriers may contribute to the induction of pathogenic IgE production.
Collapse
Affiliation(s)
- Andrea Cerutti
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain; Division of Clinical Immunology, Department of Medicine, Mount Sinai School of Medicine, New York.
| | - Martyna Filipska
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Xavi Marcos Fa
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Roser Tachó-Piñot
- Lydia Becher Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
40
|
Testera-Montes A, Palomares F, Jurado-Escobar R, Fernandez-Santamaria R, Ariza A, Verge J, Salas M, Campo P, Mayorga C, Torres MJ, Rondon C, Eguiluz-Gracia I. Sequential class switch recombination to IgE and allergen-induced accumulation of IgE + plasmablasts occur in the nasal mucosa of local allergic rhinitis patients. Allergy 2022; 77:2712-2724. [PMID: 35340036 DOI: 10.1111/all.15292] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND The involvement of allergen-specific (s)IgE in local allergic rhinitis (LAR) has been debated. Here, we investigate the effect of nasal allergen challenge with Dermatophagoides pteronyssinus (NAC-DP) in mucosal and peripheral B-cell subpopulations in LAR patients. METHODS Nine LAR, 5 allergic rhinitis (AR), and 5 non-atopic healthy control (HC) individuals were subjected to a 3-day NAC-DP protocol, and nasal biopsies and blood samples were collected before and after provocation. Nasal biopsies were used for immunohistochemistry and gene expression studies, whereas the frequency of lymphocyte subsets and basophil activation test (BAT) were analyzed in blood samples by flow cytometry. sIgG was measured in sera. RESULTS NAC-DP induced an increase in IgE+ CD38+ plasmablasts in the nasal mucosa of LAR patients, but not in AR or HC individuals. Markers of sequential recombination to IgE (εCSR) (from IgG) were observed in 33% of LAR, 20% of AR, and 0% of HC subjects. NAC-DP increased the proportion of peripheral CD19+ CD20+ CD38+ plasmablasts in AR and LAR patients, but not in HC. Expression of the mucosal homing receptor CXCR3 in peripheral CD19+ CD20+ CD38+ plasmablasts from LAR, AR, and HC individuals was 7%, 5%, and 0.5%, respectively. In vitro DP stimulation increased proliferating CD19+ CD20+ CD38+ plasmablasts in LAR and AR patients, but not in HC. Serum DP-sIgG was higher in LAR and AR patients as compared to HC. BAT was positive in 33%, 100%, and 0% of LAR, AR, and HC subjects, respectively. CONCLUSION These results suggest that allergen exposure induces the sequential εCSR of IgG+ CD19+ CD20+ CD38+ plasmablasts in the nasal mucosa of LAR patients.
Collapse
Affiliation(s)
- Almudena Testera-Montes
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Francisca Palomares
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Raquel Jurado-Escobar
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Ruben Fernandez-Santamaria
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Adriana Ariza
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Jesus Verge
- ENT Unit, Hospital Clinico Virgen de la Victoria, Malaga, Spain
| | - Maria Salas
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Paloma Campo
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
| | - Cristobalina Mayorga
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND), Malaga, Spain
| | - Maria Jose Torres
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND), Malaga, Spain
- Universidad de Málaga (UMA), Málaga, Spain
| | - Carmen Rondon
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| | - Ibon Eguiluz-Gracia
- Allergy Unit, Hospital Regional Universitario de Malaga, Malaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), RICORS "Enfermedades inflamatorias", Málaga, Spain
| |
Collapse
|
41
|
Chen Q, Xie M, Liu H, Dent AL. Development of allergen-specific IgE in a food-allergy model requires precisely timed B cell stimulation and is inhibited by Fgl2. Cell Rep 2022; 39:110990. [PMID: 35767958 PMCID: PMC9271337 DOI: 10.1016/j.celrep.2022.110990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/12/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Immunoglobulin E (IgE) responses are a central feature of allergic disease. Using a well-established food-allergy model in mice, we show that two sensitizations with cognate B cell antigen (Ag) and adjuvant 7 days apart promotes optimal development of IgE+ germinal center (GC) B cells and high-affinity IgE production. Intervals of 3 or 14 days between Ag sensitizations lead to loss of IgE+ GC B cells and an undetectable IgE response. The immunosuppressive factors Fgl2 and CD39 are down-regulated in T follicular helper (TFH) cells under optimal IgE-sensitization conditions. Deletion of Fgl2 in TFH and T follicular regulatory (TFR) cells, but not from TFR cells alone, increase Ag-specific IgE levels and IgE-mediated anaphylactic responses. Overall, we find that Ag-specific IgE responses require precisely timed stimulation of IgE+ GC B cells by Ag. Furthermore, we show that Fgl2 is expressed by TFH cells and represses IgE. This work has implications for the development and treatment of food allergies. Using a mouse food-allergy model, Chen et al. find that allergen-specific IgE responses require precisely timed stimulation of IgE+ germinal center B cells. The authors further show that Fgl2 expressed by T follicular helper cells represses IgE. This work has implications for the development and treatment of food allergy.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Markus Xie
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hong Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
42
|
Seals MR, Moran MM, Leavenworth JD, Leavenworth JW. Contribution of Dysregulated B-Cells and IgE Antibody Responses to Multiple Sclerosis. Front Immunol 2022; 13:900117. [PMID: 35784370 PMCID: PMC9243362 DOI: 10.3389/fimmu.2022.900117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS), a debilitating autoimmune inflammatory disease that affects the brain and spinal cord, causes demyelination of neurons, axonal damage, and neurodegeneration. MS and the murine experimental autoimmune encephalomyelitis (EAE) model have been viewed mainly as T-cell-mediated diseases. Emerging data have suggested the contribution of B-cells and autoantibodies to the disease progression. However, the underlying mechanisms by which dysregulated B-cells and antibody response promote MS and EAE remain largely unclear. Here, we provide an updated review of this specific subject by including B-cell biology and the role of B-cells in triggering autoimmune neuroinflammation with a focus on the regulation of antibody-producing B-cells. We will then discuss the role of a specific type of antibody, IgE, as it relates to the potential regulation of microglia and macrophage activation, autoimmunity and MS/EAE development. This knowledge can be utilized to develop new and effective therapeutic approaches to MS, which fits the scope of the Research Topic "Immune Mechanism in White Matter Lesions: Clinical and Pathophysiological Implications".
Collapse
Affiliation(s)
- Malik R. Seals
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Multidisciplinary Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Monica M. Moran
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jonathan D. Leavenworth
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
43
|
Colas L, Magnan A, Brouard S. Immunoglobulin E response in health and disease beyond allergic disorders. Allergy 2022; 77:1700-1718. [PMID: 35073421 DOI: 10.1111/all.15230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/13/2021] [Accepted: 01/16/2022] [Indexed: 12/24/2022]
Abstract
Immunoglobulin E is the latest discovered of immunoglobulin family and has been long associated with anaphylaxis and worm expulsion. Immunoglobulin E, along with mast cells, basophils, and eosinophils, is also a hallmark of type 2 immunity which is dysregulated in numerous diseases such as asthma, rhinitis, atopic dermatitis, and eosinophilic esophagitis in addition to anaphylaxis as aforementioned. However, recent advances have shed light on IgE regulation and memory explaining the low level of free IgE, the scarcity of IgE plasma cells that are mainly short live and the absence of IgE memory B cells in homeostatic conditions. Furthermore, IgE was implicated in inflammatory conditions beyond allergic disorders where IgE-mediated facilitated antigen presentation can enhance cellular and humoral response against autoantigens in systemic lupus or chronic urticaria leading to more severe disease and even against neoantigen facilitating tumor cell lysis. At last, IgE was unexpectedly associated with allograft rejection or atheromatous cardiovascular diseases where precise mechanisms remain to be deciphered. The purpose of this review is to summarize these recent advances in IgE regulation, biology, and physiopathology beyond allergic diseases opening whole new fields of IgE biology to explore.
Collapse
Affiliation(s)
- Luc Colas
- Plateforme Transversale d'Allergologie et d'immunologie Clinique PFTA Clinique dermatologique CHU de Nantes Nantes France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology Nantes France
| | - Antoine Magnan
- Hôpital Foch, Suresnes; Université de Versailles Saint‐Quentin Paris‐Saclay; INRAe Paris France
| | - Sophie Brouard
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology Nantes France
- Labex IGO Nantes France
- Centre d’Investigation Clinique en Biothérapie Centre de ressources biologiques (CRB) Nantes France
| |
Collapse
|
44
|
Papp K, Kovács Á, Orosz A, Hérincs Z, Randek J, Liliom K, Pfeil T, Prechl J. Absolute Quantitation of Serum Antibody Reactivity Using the Richards Growth Model for Antigen Microspot Titration. SENSORS (BASEL, SWITZERLAND) 2022; 22:3962. [PMID: 35632371 PMCID: PMC9147899 DOI: 10.3390/s22103962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
In spite of its pivotal role in the characterization of humoral immunity, there is no accepted method for the absolute quantitation of antigen-specific serum antibodies. We devised a novel method to quantify polyclonal antibody reactivity, which exploits protein microspot assays and employs a novel analytical approach. Microarrays with a density series of disease-specific antigens were treated with different serum dilutions and developed for IgG and IgA binding. By fitting the binding data of both dilution series to a product of two generalized logistic functions, we obtained estimates of antibody reactivity of two immunoglobulin classes simultaneously. These estimates are the antigen concentrations required for reaching the inflection point of thermodynamic activity coefficient of antibodies and the limiting activity coefficient of antigen. By providing universal chemical units, this approach may improve the standardization of serological testing, the quality control of antibodies and the quantitative mapping of the antibody-antigen interaction space.
Collapse
Affiliation(s)
- Krisztián Papp
- R&D Laboratory, Diagnosticum Zrt, 1047 Budapest, Hungary; (K.P.); (Z.H.)
| | - Ágnes Kovács
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, 1117 Budapest, Hungary; (Á.K.); (T.P.)
| | - Anita Orosz
- Department of Immunology, Eötvös Loránd University, 1117 Budapest, Hungary;
| | - Zoltán Hérincs
- R&D Laboratory, Diagnosticum Zrt, 1047 Budapest, Hungary; (K.P.); (Z.H.)
| | - Judit Randek
- Budapest University of Technology and Economics, 1111 Budapest, Hungary;
| | - Károly Liliom
- Department of Biophysics and Radiation Biology, Semmelweis University, 1085 Budapest, Hungary;
| | - Tamás Pfeil
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, 1117 Budapest, Hungary; (Á.K.); (T.P.)
- ELKH-ELTE Numerical Analysis and Large Networks Research Group, 1117 Budapest, Hungary
| | - József Prechl
- R&D Laboratory, Diagnosticum Zrt, 1047 Budapest, Hungary; (K.P.); (Z.H.)
| |
Collapse
|
45
|
Kawakami T, Kasakura K, Kawakami Y, Ando T. Immunoglobulin E-Dependent Activation of Immune Cells in Rhinovirus-Induced Asthma Exacerbation. FRONTIERS IN ALLERGY 2022; 3:835748. [PMID: 35386658 PMCID: PMC8974681 DOI: 10.3389/falgy.2022.835748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/24/2022] [Indexed: 11/26/2022] Open
Abstract
Acute exacerbation is the major cause of asthma morbidity, mortality, and health-care costs. Respiratory viral infections, particularly rhinovirus (RV) infections, are associated with the majority of asthma exacerbations. The risk for bronchoconstriction with RV is associated with allergic sensitization and type 2 airway inflammation. The efficacy of the humanized anti-IgE monoclonal antibody omalizumab in treating asthma and reducing the frequency and severity of RV-induced asthma exacerbation is well-known. Despite these clinical data, mechanistic details of omalizumab's effects on RV-induced asthma exacerbation have not been well-defined for years due to the lack of appropriate animal models. In this Perspective, we discuss potential IgE-dependent roles of mast cells and dendritic cells in asthma exacerbations.
Collapse
Affiliation(s)
- Toshiaki Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Toshiaki Kawakami
| | - Kazumi Kasakura
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Yu Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Phelps A, Bruton K, Grydziuszko E, Koenig JFE, Jordana M. The Road Toward Transformative Treatments for Food Allergy. FRONTIERS IN ALLERGY 2022; 3:826623. [PMID: 35386642 PMCID: PMC8974751 DOI: 10.3389/falgy.2022.826623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/11/2022] [Indexed: 12/28/2022] Open
Abstract
A series of landmark studies have provided conclusive evidence that the early administration of food allergens dramatically prevents the emergence of food allergy. One of the greatest remaining challenges is whether patients with established food allergy can return to health. This challenge is particularly pressing in the case of allergies against peanut, tree nuts, fish, and shellfish which are lifelong in most patients and may elicit severe reactions. The standard of care for food allergy is allergen avoidance and the timely administration of epinephrine upon accidental exposure. Epinephrine, and other therapeutic options like antihistamines provide acute symptom relief but do not target the underlying pathology of the disease. In principle, any transformative treatment for established food allergy would require the restoration of a homeostatic immunological state. This may be attained through either an active, non-harmful immune response (immunological tolerance) or a lack of a harmful immune response (e.g., anergy), such that subsequent exposures to the allergen do not elicit a clinical reaction. Importantly, such a state must persist beyond the course of the treatment and exert its protective effects permanently. In this review, we will discuss the immunological mechanisms that maintain lifelong food allergies and are, consequently, those which must be dismantled or reprogrammed to instate a clinically non-reactive state. Arguably, the restoration of such a state in the context of an established food allergy would require a reprogramming of the immune response against a given food allergen. We will discuss existing and experimental therapeutic strategies to eliminate IgE reactivity and, lastly, will propose outstanding questions to pave the road to the development of novel, transformative therapeutics in food allergy.
Collapse
Affiliation(s)
- Allyssa Phelps
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute, McMaster University, Hamilton, ON, Canada
| | | | | | | | | |
Collapse
|
47
|
Jin J, Sunusi S, Lu H. Group 2 innate lymphoid cells (ILC2s) are important in typical type 2 immune-mediated diseases and an essential therapeutic target. J Int Med Res 2022; 50:3000605211053156. [PMID: 35048721 PMCID: PMC8796086 DOI: 10.1177/03000605211053156] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The prevalence rate of allergic diseases, such as asthma, atopic rhinitis (AR), and atopic dermatitis (AD), has been significantly increasing over the years because of environmental changes. Type 2 immunity is mediated by allergic inflammation initiated by an innate immune response. This response is orchestrated by type 2 cytokines (interleukin [IL]-4, IL-5, IL-9, and IL-13) together with other cells. The dendritic cell [DC]-T helper 2 (Th2) cell axis is the conventional type 2 immune pathway, and is currently known as the group 2 innate lymphoid cell (ILC2)-DC-Th2 axis that mediates type 2 inflammation. ILC2s strongly mediate type 2 inflammation in allergic diseases. ILC2s are activated by epithelial cell-derived cytokines, such as IL-25 and IL-33, and thymic stromal lymphopoietin. Additionally, ILC2s are activated by mast cell lipid inflammatory mediators, such as cysteinyl leukotrienes and prostaglandin D2. ILC2s produce a large amount of type 2 cytokines. The important role of ILC2s in the pathogenesis of type 2-mediated disease has resulted in ILC2-derived cytokines being a target for therapeutic development. In this review, we discuss type 2 immunity, mainly the ILC2-DC-Th2 axis, and other immune cells, the dominant role of ILC2s in asthma, AR, and AD, and therapeutic targets against type 2 cytokines.
Collapse
Affiliation(s)
- Jie Jin
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Sadik Sunusi
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hongyan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
48
|
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: 5.0] [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
|
49
|
Gertie JA, Zhang B, Liu EG, Hoyt LR, Yin X, Xu L, Long LL, Soldatenko A, Gowthaman U, Williams A, Eisenbarth SC. Oral anaphylaxis to peanut in a mouse model is associated with gut permeability but not with Tlr4 or Dock8 mutations. J Allergy Clin Immunol 2022; 149:262-274. [PMID: 34051223 PMCID: PMC8626534 DOI: 10.1016/j.jaci.2021.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The etiology of food allergy is poorly understood; mouse models are powerful systems to discover immunologic pathways driving allergic disease. C3H/HeJ mice are a widely used model for the study of peanut allergy because, unlike C57BL/6 or BALB/c mice, they are highly susceptible to oral anaphylaxis. However, the immunologic mechanism of this strain's susceptibility is not known. OBJECTIVE We aimed to determine the mechanism underlying the unique susceptibility to anaphylaxis in C3H/HeJ mice. We tested the role of deleterious Toll-like receptor 4 (Tlr4) or dedicator of cytokinesis 8 (Dock8) mutations in this strain because both genes have been associated with food allergy. METHODS We generated C3H/HeJ mice with corrected Dock8 or Tlr4 alleles and sensitized and challenged them with peanut. We then characterized the antibody response to sensitization, anaphylaxis response to both oral and systemic peanut challenge, gut microbiome, and biomarkers of gut permeability. RESULTS In contrast to C3H/HeJ mice, C57BL/6 mice were resistant to anaphylaxis after oral peanut challenge; however, both strains undergo anaphylaxis with intraperitoneal challenge. Restoring Tlr4 or Dock8 function in C3H/HeJ mice did not protect from anaphylaxis. Instead, we discovered enhanced gut permeability resulting in ingested allergens in the bloodstream in C3H/HeJ mice compared to C57BL/6 mice, which correlated with an increased number of goblet cells in the small intestine. CONCLUSIONS Our work highlights the potential importance of gut permeability in driving anaphylaxis to ingested food allergens; it also indicates that genetic loci outside of Tlr4 and Dock8 are responsible for the oral anaphylactic susceptibility of C3H/HeJ mice.
Collapse
Affiliation(s)
- Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Singapore Immunology Network (SIgN), Singapore
| | - Elise G Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn
| | - Laura R Hoyt
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Xiangyun Yin
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lauren L Long
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn
| | - Arielle Soldatenko
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Department of Pathology, University of Massachusetts Medical School, Worcester, Mass
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Conn.
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn.
| |
Collapse
|
50
|
Xiong S, Jia Y, Liu C. IgE-expressing long-lived plasma cells in persistent sensitization. Front Pediatr 2022; 10:979012. [PMID: 36545659 PMCID: PMC9760851 DOI: 10.3389/fped.2022.979012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Persistent allergies affect the quality of life of patients and increase economic burdens. Many clinical observations indicate the presence of IgE+ long-lived plasma cells (LLPCs), which account for the persistent secretion of specific IgE; however, the characteristics of the IgE+ LLPCs have yet to be identified clearly. In this review, we summarized the generation of IgE+ PCs, discussed the prosurvival factors in the microenvironment, and reviewed the unique IgE-BCR signaling, which may bring insights into understanding the survival mechanisms of IgE+ LLPCs.
Collapse
Affiliation(s)
- Shiqiu Xiong
- Department of Allergy, Center for Asthma Prevention and Lung Function Laboratory, Children's Hospital of Capital Institute of Pediatrics, Beijing, China.,Department of Pediatrics, Graduate School of Peking Union Medical College, Beijing, China
| | - Yang Jia
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuanhe Liu
- Department of Allergy, Center for Asthma Prevention and Lung Function Laboratory, Children's Hospital of Capital Institute of Pediatrics, Beijing, China.,Department of Pediatrics, Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|