1
|
Sutanto H, Elisa E, Rachma B, Fetarayani D. Gut Microbiome Modulation in Allergy Treatment: The Role of Fecal Microbiota Transplantation. Am J Med 2025; 138:769-777.e3. [PMID: 39855612 DOI: 10.1016/j.amjmed.2025.01.005] [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: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
The prevalence of allergic diseases has been rising, paralleling lifestyle changes and environmental exposures that have altered human microbiome composition. This review article examines the intricate relationship between the gut microbiome and allergic diseases, emphasizing the potential of fecal microbiota transplantation as a promising novel treatment approach. It explains how reduced microbial exposure in modern societies contributes to immune dysregulation and the increasing incidence of allergies. The discussion also addresses immune homeostasis and its modulation by the gut microbiome, highlighting the shift from eubiosis to dysbiosis in allergic conditions. Furthermore, this article reviews existing studies and emerging research on the role of fecal microbiota transplantation in restoring microbial balance, providing insights into its mechanisms, efficacy, and safety.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Elisa Elisa
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Betty Rachma
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Deasy Fetarayani
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia; Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
2
|
Gomez RA, Hou J, Gersuk VH, Chow IT, Farrington ML, Robinson D, Kwok WW. Ara h 2 105-124-Specific TH2A Cells Drive Peanut Allergy in DRB1*15:01 Individuals: A Detailed Epitope Analysis. Clin Exp Allergy 2025. [PMID: 40308027 DOI: 10.1111/cea.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/03/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The IgE-mediated CD4 T-cell response to peanut (Arachis hypogaea) is heterogeneous, yet TH2 cells remain central drivers of pathology. This study aimed to dissect this complexity at the epitope level by focusing on the HLA-DRB1*15:01-DRB5*01:01 haplotype. Specifically, we examined how distinct epitope-specific T-cell subsets shape the immunological landscape of peanut allergy in peanut-allergic (PA) versus non-peanut-allergic (NPA) individuals. METHODS Using in vitro and ex vivo MHC-II tetramer approaches, the phenotype, frequency, function, and transcriptome of CD4 T-cell responses to novel Ara h epitopes were assessed. Bulk RNA sequencing further characterised these T cells, allowing identification of subsets associated with TH2 polarisation in PA individuals. RESULTS Eleven HLA-DRB1*15:01 and DRB5*01:01-restricted epitopes were identified in Ara h 1, 2, 3, 6, 7, and 8 using tetramer-guided epitope mapping on cell lines, followed by ex vivo validation in peripheral blood. T-cell phenotype was epitope-dependent, with a distinct TH2A population specific to the epitope Ara h 2105-124 (Ara h 2 p14) detected only in PA donors. These TH2A cells were phenotypically and transcriptionally distinct, marked by high CRTH2/CD161, low CD27, IL-5 production, and gene enrichment in cytokine signalling and lipid metabolism. Other epitope-specific T-cell subsets displayed more heterogeneous gene profiles related to immune activation, differentiation, and antigen presentation, underscoring the complexity of peanut-specific responses even within a single HLA haplotype. CONCLUSION These findings reveal that the strong TH2 bias in DRB1*15:01-DRB5*01:01 PA individuals arises from a distinct subset of Ara h 2 p14-specific TH2A cells characterised by a specialised metabolic and cytokine signalling program. At the same time, the functional diversity observed in non-Ara h 2 p14 subsets highlights the potential for leveraging these populations in tolerance-promoting therapies. Understanding the epitope-level heterogeneity of peanut-specific T-cells provides insight into the epitope-specific mechanisms driving peanut allergy and has potential implications for therapeutic interventions.
Collapse
Affiliation(s)
- Rebecca A Gomez
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Jue Hou
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Vivian H Gersuk
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - I-Ting Chow
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Immune Medicine Division, Adaptive Biotechnologies, Seattle, Washington, USA
| | - Mary L Farrington
- Allergy and Immunology, Virginia Mason Franciscan Health, Seattle, Washington, USA
| | - David Robinson
- Allergy and Immunology, Virginia Mason Franciscan Health, Seattle, Washington, USA
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Kato A, Kita H. The immunology of asthma and chronic rhinosinusitis. Nat Rev Immunol 2025:10.1038/s41577-025-01159-0. [PMID: 40240657 DOI: 10.1038/s41577-025-01159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Asthma and chronic rhinosinusitis (CRS) are common chronic inflammatory diseases of the respiratory tract that have increased in prevalence over the past five decades. The clinical relationship between asthma and CRS has been well recognized, suggesting a common pathogenesis between these diseases. Both diseases are driven by complex airway epithelial cell and immune cell interactions that occur in response to environmental triggers such as allergens, microorganisms and irritants. Advances, including a growing understanding of the biology of the cells involved in the disease, the application of multiomics technologies and the performance of large-scale clinical studies, have led to a better understanding of the pathophysiology and heterogeneity of asthma and CRS. This research has promoted the concept that these diseases consist of several endotypes, in which airway epithelial cells, innate lymphoid cells, T cells, B cells, granulocytes and their mediators are distinctly involved in the immunopathology. Identification of the disease heterogeneity and immunological markers has also greatly improved the protocols for biologic therapies and the clinical outcomes in certain subsets of patients. However, many clinical and research questions remain. In this Review, we discuss recent advances in characterizing the immunological mechanisms of asthma and CRS, with a focus on the main cell types and molecules involved in these diseases.
Collapse
Affiliation(s)
- Atsushi Kato
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hirohito Kita
- Division of Allergy, Asthma, and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ, USA.
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ, USA.
| |
Collapse
|
4
|
Pizzarello CR, Jackson CM, Herman K, Seppo AE, Rebhahn J, Scherzi T, Berin MC, Looney RJ, Mosmann TR, Järvinen KM. A Phenotypically Distinct Human Th2 Cell Subpopulation Is Associated With Development of Allergic Disorders in Infancy. Allergy 2025; 80:949-964. [PMID: 39899007 PMCID: PMC11971024 DOI: 10.1111/all.16489] [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: 03/21/2024] [Revised: 11/18/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Little is known about the ontogeny of T cell immunity during infancy in farming and urban lifestyles due to the lack of immunophenotyping in such birth cohorts. METHODS Two birth cohorts (farming and urban) at differing risks and rates of allergic diseases were compared. Blood mononuclear cells were collected from infants at birth, and 6 and 12 months of age. Full spectrum flow cytometry, followed by traditional gating and the Scalable Weighted Iterative Flow-clustering Technique (SWIFT) high-dimensional analysis, were used to identify cell populations that differed between farming and urban infants. Additionally, single-cell RNAseq and multiplex cytokine assays were used to assess the function of cell populations of interest. RESULTS Several regulatory T cell (Treg) subpopulations were elevated in farming lifestyles and in non-atopic infants. A unique effector memory CD25+CD127+CD161-CD49d+CCR4+CRTH2+ Th2 population was elevated at 6 months in urban infants and in those who developed atopic dermatitis and/or food allergy and allergic sensitization. Although this population shared Th2 and IL-9 skewing with Th2A cells, the population uniquely failed to express CD161, produced more IL-2 and TNF-α, and upregulated the differentially expressed genes (DEGs), FOXP3 and the cytokine inducible SH2-containing protein gene (CISH) relative to Th2A cells. This population has been termed Th2B cells. CONCLUSION We describe a unique effector memory Th2 population elevated in urban high-risk infants, potentially implicated in the development of allergic disease.
Collapse
Affiliation(s)
- Catherine R Pizzarello
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Courtney M Jackson
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Katherine Herman
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Jonathan Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Tyler Scherzi
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - R John Looney
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Tim R Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
5
|
Jaiswal YS, Williams LL. The Rising Incidence of Food Allergies and Infant Food Allergies. Annu Rev Food Sci Technol 2025; 16:269-287. [PMID: 39971351 DOI: 10.1146/annurev-food-111523-121952] [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] [Indexed: 02/21/2025]
Abstract
Infant food allergies have become a continually rising global health issue. There is a lack of global standardized recommendations on measures for prevention and treatment of infant food allergies because of the variations in ethnic, social, educational, and healthcare practices that affect the outcomes of research studies. Food allergies can cause mild to severe reactions and can affect social and emotional aspects of life up to the adolescent stage and are sometimes never outgrown. Maternal factors such as in utero supply of antibodies, dietary diversity, genetics, food allergen consumption during pregnancy, gut microbiota, and breastfeeding characteristics are the cornerstones of the development of an infant's immune system. In this review, we discuss how prenatal and postnatal factors affect the gut microbiota and development of an infant's immune system, and the current therapies available. The importance of food processing and education of stakeholders in the care of infants with food allergies is also discussed.
Collapse
Affiliation(s)
- Yogini S Jaiswal
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, Kannapolis, North Carolina, USA; ,
| | - Leonard L Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, Kannapolis, North Carolina, USA; ,
| |
Collapse
|
6
|
Rabin RL, Altman MC, Arshad SH, Beger RD, Frischmeyer-Guerrerio PA, Goleva E, Hamilton RG, Khurana Hershey GK, Shamji MH, Sampson HA, Santos AF, Shreffler WG, Togias A, Vieths S, Wambre E, Wenzel SE, Hise K, Lee J, Tripathi A, Slater JE. Biomarker-driven drug development for allergic diseases and asthma: An FDA public workshop. J Allergy Clin Immunol 2025:S0091-6749(25)00329-X. [PMID: 40154576 DOI: 10.1016/j.jaci.2025.03.014] [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: 10/10/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
The US Food and Drug Administration (FDA) hosted a workshop on February 22, 2024, to discuss the status of biomarkers in drug development for allergic asthma and food allergy. The workshop provided a forum for open discussion among regulators, academicians, National Institutes of Health staff and industry to inform stakeholders of the requirements for the FDA to adopt a biomarker as a surrogate end point for a clinical trial, and to inform FDA of the status of various biomarkers in development. The workshop was divided into 3 sessions: (1) FDA and European Union regulators discussing regulatory perspectives on use of biomarkers in drug development programs, (2) investigators discussing biomarkers for pediatric and adult asthma, and (3) investigators discussing biomarkers for food allergy. In this report, we review the information presented at the workshop and summarize the current status of potential biomarkers for these allergic diseases.
Collapse
Affiliation(s)
- Ronald L Rabin
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md.
| | | | - S Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Richard D Beger
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Ark
| | | | - Elena Goleva
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | | | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hugh A Sampson
- Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course and Population Sciences & Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, and the Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Wayne G Shreffler
- Food Allergy Center, Division of Pediatric Allergy and Immunology, and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Stefan Vieths
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Erik Wambre
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, Pa
| | - Kathleen Hise
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Joohee Lee
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Anubha Tripathi
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Jay E Slater
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| |
Collapse
|
7
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [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] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Khan M, Alteneder M, Reiter W, Krausgruber T, Dobnikar L, Madern M, Waldherr M, Bock C, Hartl M, Ellmeier W, Henriksson J, Boucheron N. Single-cell and chromatin accessibility profiling reveals regulatory programs of pathogenic Th2 cells in allergic asthma. Nat Commun 2025; 16:2565. [PMID: 40089475 PMCID: PMC11910648 DOI: 10.1038/s41467-025-57590-3] [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: 08/28/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Lung pathogenic T helper type 2 (pTh2) cells are important in mediating allergic asthma, but fundamental questions remain regarding their heterogeneity and epigenetic regulation. Here we investigate immune regulation in allergic asthma by single-cell RNA sequencing in mice challenged with house dust mite, in the presence and absence of histone deacetylase 1 (HDAC1) function. Our analyses indicate two distinct highly proinflammatory subsets of lung pTh2 cells and pinpoint thymic stromal lymphopoietin (TSLP) and Tumour Necrosis Factor Receptor Superfamily (TNFRSF) members as important drivers to generate pTh2 cells in vitro. Using our in vitro model, we uncover how signalling via TSLP and a TNFRSF member shapes chromatin accessibility at the type 2 cytokine gene loci by modulating HDAC1 repressive function. In summary, we have generated insights into pTh2 cell biology and establish an in vitro model for investigating pTh2 cells that proves useful for discovering molecular mechanisms involved in pTh2-mediated allergic asthma.
Collapse
Affiliation(s)
- Matarr Khan
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Marlis Alteneder
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Wolfgang Reiter
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Lina Dobnikar
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Moritz Madern
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Monika Waldherr
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
- FH Campus Wien, Department of Applied Life Sciences/Bioengineering/Bioinformatics, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Markus Hartl
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Johan Henriksson
- Umeå University, Umeå Centre for Microbial Research (UCMR), Integrated Science Lab (Icelab), Department of Molecular Biology, Umeå, Sweden
| | - Nicole Boucheron
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria.
| |
Collapse
|
9
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
10
|
Zhou X, Dunham D, Sindher SB, Long A, Fernandes A, Chang I, Assa’ad A, Pongracic J, Spergel JM, Tam J, Tilles S, Wang J, Boyd SD, Chinthrajah RS, Nadeau KC. HLA-DR + regulatory T cells and IL-10 are associated with success or failure of desensitization outcomes. Allergy 2025; 80:762-774. [PMID: 39291303 PMCID: PMC11893263 DOI: 10.1111/all.16311] [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: 02/14/2024] [Revised: 07/14/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Omalizumab (XOLAIR®)-assisted multi-food oral immunotherapy (mOIT) has been shown to safely, effectively, and rapidly desensitize patients with multiple food allergies. In our clinical trial (NCT02626611) on omalizumab-assisted mOIT, different desensitization outcomes (success or failure of desensitization) were observed following a period of either continued or discontinued mOIT. However, the association between the immunological changes induced by omalizumab-assisted mOIT and desensitization outcomes has not yet been fully elucidated. In this study, due to the key roles of regulatory T (Treg) cells and the type 2 helper T cell (Th2) pathway in immune tolerance to food allergens, we aimed to characterize their association with the desensitization outcomes of omalizumab-assisted mOIT. METHODS Mass cytometry and multiplex cytokine assays were performed on blood samples obtained from participants with allergies to peanut, cashew, or milk in our phase 2 clinical study (NCT02626611). Comprehensive statistical and bioinformatic analyses were conducted on high-dimensional cytometry-based single-cell data and high-throughput multiplex cytokine data. RESULTS Our results demonstrated that the frequency of HLA-DR+ Treg cells, and the production of Th2 cytokines (IL-4, IL-5, IL-13, and IL-9) as well as the immunoregulatory cytokine IL-10 by peripheral blood mononuclear cells (PBMCs) was significantly increased in cultures with allergen compared to cultures with media alone at baseline (Week 0). We also observed increased frequency of allergen responsive HLA-DR+ Treg cells and enhanced production of IL-10 by PBMCs in participants who achieved successful desensitization compared to those with failure of desensitization. However, the production of Th2 cytokines by PBMCs did not show significant differences between participants with different desensitization outcomes (success vs. failure of desensitization), despite omalizumab-assisted mOIT inducing a significant reduction in the production of Th2 cytokines. CONCLUSIONS We demonstrated that the frequency of HLA-DR+ Treg cells and IL-10 cytokine production by PBMCs are associated with desensitization outcomes of omalizumab-assisted mOIT. These findings suggest potential immunological parameters that could be targeted to enhance desensitization success rates.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Diane Dunham
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sayantani B Sindher
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Long
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrea Fernandes
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris Chang
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Amal Assa’ad
- Division of Allergy and Immunology, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
| | - Jacqueline Pongracic
- Division of Allergy and Immunology, the Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Tam
- Division of Clinical Immunology and Allergy, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Stephen Tilles
- Seattle Allergy and Asthma Research Institute, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Julie Wang
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C. Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
Lozano-Ojalvo D, Chen X, Kazmi W, Menchén-Martínez D, Pérez-Rodríguez L, Fernandes-Braga W, Tyler S, Benkov K, Pittman N, Lai J, Sampson HA, Curotto de Lafaille M, Dunkin D, Berin MC. Differential T follicular helper cell phenotypes distinguish IgE-mediated milk allergy from eosinophilic esophagitis in children. J Allergy Clin Immunol 2025; 155:909-922. [PMID: 39389123 PMCID: PMC11895100 DOI: 10.1016/j.jaci.2024.09.024] [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: 10/25/2023] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND IgE-mediated food allergy and eosinophilic esophagitis (EoE) are diseases commonly triggered by milk. Milk-responsive CD4+ T cells producing type 2 cytokines are present in both diseases, yet the clinical manifestation of disease in milk allergy (MA) and EoE are distinct. OBJECTIVE We sought to identify differences in CD4+ T cells between EoE and MA that may be responsible for distinct disease manifestations. METHOD The total and milk-specific CD4+ T-cell phenotype of children with MA, children with EoE (active or in remission), and controls was measured using spectral flow cytometry of peripheral blood (all groups) or esophageal biopsies (EoE and control). RESULTS Circulating milk-responsive T cells could be identified in active EoE and MA. An increased frequency of TH2A cells was also noted in MA and EoE. In circulating T cells, type 2 cytokine production was elevated in MA, but not EoE. Within the milk-responsive T follicular helper (TFH) subset, a dichotomy of phenotype was noted: TFH13 cells predominated in MA, while IL-10-producing TFH cells predominated in EoE. In the esophagus, CD4+ T cells were constitutively activated and expressed not only type 2 cytokines, but also IL-10 and IL-21 in active EoE. IgG4 was produced from CD38+ plasma cells in close proximity to CD4+ T cells. In vitro activation studies demonstrated that IL-10 and IL-21 elicited strong IgG4 responses in B lymphocytes, while IL-4 and IL-13 promoted IgE production. CONCLUSIONS Our studies demonstrate a dichotomy of TFH responses that may be the basis for different clinical manifestations to milk in EoE and MA.
Collapse
Affiliation(s)
- Daniel Lozano-Ojalvo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Instituto de Investigación en Ciencias de la Alimentación, Madrid, Spain
| | - Xin Chen
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Wajiha Kazmi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David Menchén-Martínez
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Instituto de Investigación en Ciencias de la Alimentación, Madrid, Spain
| | | | | | - Scott Tyler
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Keith Benkov
- Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nanci Pittman
- Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joanne Lai
- Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hugh A Sampson
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - David Dunkin
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - M Cecilia Berin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Medicine, Feinberg School of Medicine at Northwestern University, Chicago, Ill.
| |
Collapse
|
12
|
Ogulur I, Mitamura Y, Yazici D, Pat Y, Ardicli S, Li M, D'Avino P, Beha C, Babayev H, Zhao B, Zeyneloglu C, Giannelli Viscardi O, Ardicli O, Kiykim A, Garcia-Sanchez A, Lopez JF, Shi LL, Yang M, Schneider SR, Skolnick S, Dhir R, Radzikowska U, Kulkarni AJ, Imam MB, Veen WVD, Sokolowska M, Martin-Fontecha M, Palomares O, Nadeau KC, Akdis M, Akdis CA. Type 2 immunity in allergic diseases. Cell Mol Immunol 2025; 22:211-242. [PMID: 39962262 PMCID: PMC11868591 DOI: 10.1038/s41423-025-01261-2] [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: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025] Open
Abstract
Significant advancements have been made in understanding the cellular and molecular mechanisms of type 2 immunity in allergic diseases such as asthma, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis (EoE), food and drug allergies, and atopic dermatitis (AD). Type 2 immunity has evolved to protect against parasitic diseases and toxins, plays a role in the expulsion of parasites and larvae from inner tissues to the lumen and outside the body, maintains microbe-rich skin and mucosal epithelial barriers and counterbalances the type 1 immune response and its destructive effects. During the development of a type 2 immune response, an innate immune response initiates starting from epithelial cells and innate lymphoid cells (ILCs), including dendritic cells and macrophages, and translates to adaptive T and B-cell immunity, particularly IgE antibody production. Eosinophils, mast cells and basophils have effects on effector functions. Cytokines from ILC2s and CD4+ helper type 2 (Th2) cells, CD8 + T cells, and NK-T cells, along with myeloid cells, including IL-4, IL-5, IL-9, and IL-13, initiate and sustain allergic inflammation via T cell cells, eosinophils, and ILC2s; promote IgE class switching; and open the epithelial barrier. Epithelial cell activation, alarmin release and barrier dysfunction are key in the development of not only allergic diseases but also many other systemic diseases. Recent biologics targeting the pathways and effector functions of IL4/IL13, IL-5, and IgE have shown promising results for almost all ages, although some patients with severe allergic diseases do not respond to these therapies, highlighting the unmet need for a more detailed and personalized approach.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Carina Beha
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Bingjie Zhao
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical and Diagnostic Science, School of Medicine, University of Salamanca, Salamanca, Spain
| | - Juan-Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Minglin Yang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, CA, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, CA, USA
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Abhijeet J Kulkarni
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mar Martin-Fontecha
- Departamento de Quimica Organica, Facultad de Optica y Optometria, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
13
|
Mannion JM, Rahimi RA. Tissue-Resident Th2 Cells in Type 2 Immunity and Allergic Diseases. Immunol Rev 2025; 330:e70006. [PMID: 39981858 PMCID: PMC11897987 DOI: 10.1111/imr.70006] [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: 08/23/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Type 2 immunity represents a unique immune module that provides host protection against macro-parasites and noxious agents such as venoms and toxins. In contrast, maladaptive type 2 immune responses cause allergic diseases. While multiple cell types play important roles in type 2 immunity, recent studies in humans and murine models of chronic allergic diseases have shown that a distinct population of tissue-resident, CD4+ T helper type 2 (Th2) cells play a critical role in chronic allergic inflammation. The rules regulating Th2 cell differentiation have remained less well defined than other T cell subsets, but recent studies have shed new light into the specific mechanisms controlling Th2 cell biology in vivo. Here, we review our current understanding of the checkpoints regulating the development and function of tissue-resident Th2 cells with a focus on chronic allergic diseases. We discuss evidence for a barrier tissue checkpoint in initial Th2 cell priming, including the role of neuropeptides, damage-associated molecular patterns, and dendritic cell macro-clusters. Furthermore, we review the evidence for a second barrier tissue checkpoint that instructs the development of multi-cytokine producing, tissue-resident Th2 cells that orchestrate allergic inflammation. Lastly, we discuss potential approaches to therapeutically target tissue-resident Th2 cells in chronic allergic diseases.
Collapse
Affiliation(s)
- Jenny M Mannion
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rod A Rahimi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Lai CL, Santner-Nanan B, Maltese PJ, Ong CKS, Palmer DJ, Campbell DE, Makrides M, Gold M, Nanan R, Prescott SL, Hsu PS. Impaired calcium influx underlies skewed T helper cell differentiation in children with IgE-mediated food allergies. Allergy 2025; 80:513-524. [PMID: 39250135 DOI: 10.1111/all.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Reasons for Th2 skewing in IgE-mediated food allergies remains unclear. Clinical observations suggest impaired T cell activation may drive Th2 responses evidenced by increased atopic manifestations in liver transplant patients on tacrolimus (a calcineurin inhibitor). We aimed to assess differentiation potential, T cell activation and calcium influx of naïve CD4+ T cells in children with IgE-mediated food allergies. METHODS Peripheral blood mononuclear cells from infants in the Starting Time for Egg Protein (STEP) Trial were analyzed by flow cytometry to assess Th1/Th2/Treg development. Naïve CD4+ T cells from children with and without food allergies were stimulated for 7 days to assess Th1/Th2/Treg transcriptional factors and cytokines. Store operated calcium entry (SOCE) was measured in children with and without food allergies. The effect of tacrolimus on CD4+ T cell differentiation was assessed by treating stimulated naïve CD4+ T cells from healthy volunteers with tacrolimus for 7 days. RESULTS Egg allergic infants had impaired development of IFNγ+ Th1 cells and FoxP3+ transitional CD4+ T cells compared with non-allergic infants. This parallels reduced T-bet, IFNγ and FoxP3 expression in naïve CD4+ T cells from food allergic children after in vitro culture. SOCE of naïve CD4+ T cells was impaired in food allergic children. Naïve CD4+ T cells treated with tacrolimus had reduced IFNγ, T-bet, and FoxP3, but preserved IL-4 expression. CONCLUSIONS In children with IgE-mediated food allergies, dysregulation of T helper cell development is associated with impaired SOCE, which underlies an intrinsic impairment in Th1 and Treg differentiation. Along with tacrolimus-induced Th2 skewing, this highlights an important role of SOCE/calcineurin pathway in T helper cell differentiation.
Collapse
Affiliation(s)
- C L Lai
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - B Santner-Nanan
- Sydney Medical School Nepean and Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales, Australia
| | - P J Maltese
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
| | - C K S Ong
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
| | - D J Palmer
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Medicine, The University of Western Australia, Crawley, Western Australia, Australia
| | - D E Campbell
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - M Makrides
- South Australian Health and Medical Research Institute, SAHMRI Women and Kids, Adelaide, South Australia, Australia
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - M Gold
- Discipline of Paediatrics, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - R Nanan
- Sydney Medical School Nepean and Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales, Australia
| | - S L Prescott
- School of Medicine, The University of Western Australia, Crawley, Western Australia, Australia
- The ORIGINS Project, Telethon Kids Institute, The University of Western Australia, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
| | - P S Hsu
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Schärli S, Luther F, Di Domizio J, Hillig C, Radonjic-Hoesli S, Thormann K, Simon D, Rønnstad ATM, Ruge IF, Fritz BG, Bjarnsholt T, Vallone A, Kezic S, Menden MP, Roesner LM, Werfel T, Thyssen JP, Eyerich S, Gilliet M, Bertschi NL, Schlapbach C. IL-9 sensitizes human T H2 cells to proinflammatory IL-18 signals in atopic dermatitis. J Allergy Clin Immunol 2025; 155:491-504.e9. [PMID: 39521283 DOI: 10.1016/j.jaci.2024.10.027] [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: 06/27/2024] [Revised: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND TH2 cells crucially contribute to the pathogenesis of atopic dermatitis (AD) by secreting high levels of IL-13 and IL-22. Yet the upstream regulators that activate TH2 cells in AD skin remain unclear. IL-18 is a putative upstream regulator of TH2 cells because it is implicated in AD pathogenesis and has the capacity to activate T cells. OBJECTIVE We sought to decipher the role of IL-18 in TH2 responses in blood and skin of AD patients. METHODS Peripheral blood mononuclear cells and skin biopsy samples from AD patients and healthy donors were used. Functional assays were performed ex vivo using stimulation or blocking experiments. Analysis was performed by flow cytometry, bead-based multiplex assays, RT-qPCR, RNA-Seq, Western blot, and spatial sequencing. RESULTS IL-18Rα+ TH2 cells were enriched in blood and lesional skin of AD patients. Of all the cytokines for which TH2 cells express the receptor, only IL-9 was able to induce IL-18R via an IL-9R-JAK1/JAK3-STAT1 signaling pathway. Functionally, stimulation of circulating TH2 cells with IL-18 induced secretion of IL-13 and IL-22, an effect that was enhanced by costimulation with IL-9. Mechanistically, IL-18 induced TH2 cytokines via activation of IRAK4, NF-κB, and AP-1 signaling in TH2 cells, and neutralization of IL-18 inhibited these cytokines in cultured explants of AD skin lesions. Finally, IL-18 protein levels correlated positively with disease severity in lesional AD skin. CONCLUSION Our data identify a novel IL-9/IL-18 axis that contributes to TH2 responses in AD. Our findings suggest that both IL-9 and IL-18 could represent upstream targets for future treatment of AD.
Collapse
Affiliation(s)
- Stefanie Schärli
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Fabian Luther
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jeremy Di Domizio
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Christina Hillig
- Computational Health Center, Institute of Computational Biology, Helmholtz Munich, Munich, Germany
| | - Susanne Radonjic-Hoesli
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Kathrin Thormann
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Iben Frier Ruge
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Blaine G Fritz
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Angela Vallone
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sanja Kezic
- Department of Public and Occupational Health, Amsterdam University Medical Center, University of Amsterdam, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Michael P Menden
- Computational Health Center, Institute of Computational Biology, Helmholtz Munich, Munich, Germany; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School (MHH), Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School (MHH), Hannover, Germany
| | - Jacob P Thyssen
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Stefanie Eyerich
- Center for Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Michel Gilliet
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Nicole L Bertschi
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Boyd H, Santos AF. Novel diagnostics in food allergy. J Allergy Clin Immunol 2025; 155:275-285. [PMID: 39710304 DOI: 10.1016/j.jaci.2024.12.1071] [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: 10/22/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Food allergy is increasing in prevalence, posing significant challenges for individuals and their families and adversely affecting their quality of life. Misdiagnosis can lead to unnecessary dietary and social limitations and increased food allergy risk, whereas failure to diagnose may result in life-threatening anaphylaxis. Therefore, a precise diagnosis is of the utmost importance; however, barriers exist at every stage of the diagnostic process. Diagnosis of food allergy relies on clinical history, IgE sensitization tests, and oral food challenge. Component testing and identification of optimal cutoffs have improved diagnostic accuracy. Nevertheless, many patients still require an oral food challenge, and better tests are needed to reduce this need. Novel ways of detecting biomarkers, such as the basophil activation test and peptide-specific IgE level, are transitioning into clinical practice. Future approaches may include the use of alternative biologic samples, novel laboratory technologies, and analytic tools (including artificial intelligence) to integrate test results and clinical information. Conscientious use of existing tests, access to tests with superior diagnostic accuracy, and combination of tests, can lead to improved precision of diagnosis of food allergy and timely introduction of tolerated foods into the diet. This review summarizes recent advances in novel approaches to food allergy diagnosis that can enhance clinical decision making both now and in the future.
Collapse
Affiliation(s)
- Holly Boyd
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom.
| |
Collapse
|
17
|
Alkon N, Chennareddy S, Cohenour ER, Ruggiero JR, Stingl G, Bangert C, Rindler K, Bauer WM, Weninger W, Griss J, Jonak C, Brunner PM. Single-cell sequencing delineates T-cell clonality and pathogenesis of the parapsoriasis disease group. J Allergy Clin Immunol 2025; 155:461-478. [PMID: 39278361 DOI: 10.1016/j.jaci.2024.09.004] [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: 03/27/2024] [Revised: 07/30/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Mycosis fungoides (MF), the most common cutaneous T-cell lymphoma, is often underdiagnosed in early stages because of similarities with benign dermatoses such as atopic dermatitis (AD). Furthermore, the delineation from what is called "parapsoriasis en plaque", a disease that can appear either in a small- or large-plaque form, is still controversial. OBJECTIVE We sought to characterize the parapsoriasis disease spectrum. METHODS We performed single-cell RNA sequencing of skin biopsies from patients within the parapsoriasis-to-early-stage MF spectrum, stratified for small and large plaques, and compared them to AD, psoriasis, and healthy control skin. RESULTS Six of 8 large-plaque lesions harbored either an expanded alpha/beta or gamma/delta T-cell clone with downregulation of CD7 expression, consistent with a diagnosis of early-stage MF. In contrast, 6 of 7 small-plaque lesions were polyclonal in nature, thereby lacking a lymphomatous phenotype, and also revealed a less inflammatory microenvironment than early-stage MF or AD. Of note, polyclonal small- and large-plaque lesions characteristically harbored a population of NPY+ innate lymphoid cells and displayed a stromal signature of complement upregulation and antimicrobial hyperresponsiveness in fibroblasts and sweat gland cells, respectively. These conditions were clearly distinct from AD or psoriasis, which uniquely harbored CD3+CRTH2+ IL-13 expressing "TH2A" cells, or strong type 17 inflammation, respectively. CONCLUSION These data position polyclonal small- and large-plaque parapsoriasis lesions as a separate disease entity that characteristically harbors a so far undescribed innate lymphoid cell population. We thus propose a new term, "polyclonal parapsoriasis en plaque", for this kind of lesion because they can be clearly differentiated from early- and advanced-stage MF, psoriasis, and AD on several cellular and molecular levels.
Collapse
Affiliation(s)
- Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emry R Cohenour
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John R Ruggiero
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
18
|
Oishi K, Nakano N, Ota M, Inage E, Izawa K, Kaitani A, Ando T, Hara M, Ohtsuka Y, Nishiyama C, Ogawa H, Kitaura J, Okumura K, Shimizu T. MHC Class II-Expressing Mucosal Mast Cells Promote Intestinal Mast Cell Hyperplasia in a Mouse Model of Food Allergy. Allergy 2025. [PMID: 39868907 DOI: 10.1111/all.16477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND IgE-mediated food allergy is accompanied by mucosal mast cell (MMC) hyperplasia in the intestinal mucosa. Intestinal MMC numbers correlate with the severity of food allergy symptoms. However, the mechanisms by which MMCs proliferate excessively are poorly understood. Here, we clarify the role of newly identified MHC class II (MHCII)-expressing MMCs in the effector phase of IgE-mediated food allergy. METHODS Mice reconstituted with MHCII-deficient or wild-type MMCs were used to generate a mouse mode of IgE-mediated food allergy. We assessed the extent of intestinal MMC hyperplasia and the severity of hypothermia in these mice. In addition, we performed in vitro antigen presentation assay using induced MHCII-expressing MMCs generated from bone marrow cells to evaluate the effect of CD4+ T cell activation on MMC proliferation. RESULTS In food-allergic mice, we identified the appearance of MHCII-expressing MMCs in the intestinal mucosa and showed that MMC hyperplasia was suppressed in mice with MHCII-deficient MMCs compared to mice with wild-type MMCs. In vitro assays demonstrated that MHCII-expressing MMCs incorporate food antigens directly and through the high-affinity IgE receptor FcεRI-mediated endocytosis and activate antigen-specific CD4+ T cells from food-allergic mice by antigen presentation. Activated CD4+ T cells secrete IL-4 and large amounts of IL-5, which enhance production of the mast cell growth factor IL-9 by IL-33-activated MMCs. Excess IL-9 causes excessive MMC proliferation, leading to the development of MMC hyperplasia. CONCLUSION Antigen presentation to CD4+ T cells by MHCII-expressing MMCs triggers intestinal MMC hyperplasia and exacerbates IgE-mediated food allergy.
Collapse
Affiliation(s)
- Kenji Oishi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masamu Ota
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Eisuke Inage
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kumi Izawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ayako Kaitani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mutsuko Hara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshikazu Ohtsuka
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chiharu Nishiyama
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jiro Kitaura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Dantzer JA, Lewis SA, Psoter KJ, Sutherland A, Frazier A, Richardson E, Maiche S, Seumois G, Peters B, Wood RA. Clinical and immunological outcomes after randomized trial of baked milk oral immunotherapy for milk allergy. JCI Insight 2025; 10:e184301. [PMID: 39782691 PMCID: PMC11721308 DOI: 10.1172/jci.insight.184301] [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: 07/01/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUNDCow's milk (CM) allergy is the most common food allergy in young children. Treatment with oral immunotherapy (OIT) has shown efficacy, but high rates of adverse reactions. The aim of this study was to determine whether baked milk OIT (BMOIT) could reduce adverse reactions while still inducing desensitization, and to identify immunological correlates of successful BMOIT.METHODSThis phase II, randomized trial evaluated the safety and efficacy of BMOIT in milk-allergic children 3-18 years old. After the initial placebo-controlled first year of treatment, placebo-treated participants crossed over to active BMOIT. Double-blind, placebo-controlled oral food challenges (OFCs) were conducted with BM after year 1 and to both BM and unheated milk (UM) after year 2. IgG and IgE antibodies were measured along with CM-specific (CM+) CD4+ memory T cell populations, profiled using flow cytometry and scRNA-Seq.RESULTSTwenty-one of 30 (70%) reached the primary endpoint of tolerating 4044 mg of BM protein at month 24, and 11 of 30 tolerated 2000 mg or more of UM protein. Dosing symptoms were common, but more than 98% were mild, with no severe reactions. Immunological changes associated with desensitization included increased CM IgG4, CM+ FOXP3+ cells, and Tregs and corresponding decreases in CM IgE, CM+ Th2A cells, and CD154+ cells. T cell and antibody measurements were combined to build a model that predicted UM OFC outcomes.CONCLUSIONBMOIT was well tolerated and induced desensitization to BM and UM. This desensitization corresponded to redistribution within antigen-specific antibody and T cell compartments that provided insight into the mechanistic changes that occur with OIT treatment.TRIAL REGISTRATIONClinicalTrials.gov NCT03462030.FUNDING: Myra Reinhardt Family Foundation (grant number 128388), NIH/NIAID (U19AI135731, T32AI125179, S10OD025052).
Collapse
Affiliation(s)
- Jennifer A. Dantzer
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sloan A. Lewis
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Kevin J. Psoter
- Division of General Pediatrics, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - April Frazier
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Eve Richardson
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Synaida Maiche
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Gregory Seumois
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert A. Wood
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
20
|
De Vlieger L, Nuyttens L, Keppens C, Ieven T, Matton C, Diels M, Verelst S, Raes M, Leus J, Coppens K, Sauer K, Dilissen E, Cremer J, Coorevits L, Frans G, Schrijvers R, Bullens DMA. Egg allergen-specific T-cell and cytokine responses in healthy and egg-allergic children naturally tolerating baked egg. Pediatr Allergy Immunol 2025; 36:e70018. [PMID: 39803990 DOI: 10.1111/pai.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND Type 1 regulatory T (Tr1) cells are critical players in maintaining peripheral tolerance, by producing high IL-10 levels in association with inducible T-cell co-stimulator (ICOS) expression. Whether these cells play a role in naturally acquired baked egg tolerance is unknown. OBJECTIVES Evaluate frequencies of egg-responsive Tr1 and Th2 cells in egg-allergic children that naturally acquired baked egg tolerance (BET) versus non-egg-allergic (NEA) children. METHODS Peripheral blood mononuclear cells from 70 natural BET and 15 NEA children were stimulated for 7 days with ovalbumin and ovomucoid. By flowcytometry, egg-responsive Tr1 cells were identified by co-expression of CD49b and LAG3, and Th2 cells by expression of CD49b but absence of LAG3. Seven-day cultured supernatant was analyzed for Th1, Th2, Tr1, and Th17 cytokines by MSD. RESULTS Natural BET children had a higher percentage of egg-responsive Th2 cells vs. NEA children (6.75% vs. 10.35%, p = .006). No significant difference was found in frequencies of egg-responsive Tr1 cells between NEA and natural BET children (11.40% vs. 12.55%, p = .42), although Tr1-related IL-10 and IL-21 production was higher in BET children. Interestingly, egg-responsive Tr1 cells from NEA children expressed higher ICOS levels vs. natural BET children (97.90 vs. 88.20, p < .0001). Supernatant from natural BET children showed elevated levels of Th2 cytokines IL-5, IL-9 and IL-13 and Th17 cytokine IL-17A. CONCLUSION Natural BET children maintain increased egg-specific Th2 responses, along with comparable proportions of egg-responsive Tr1 cells exhibiting higher IL-10 but lower ICOS expression in comparison with NEA children.
Collapse
Affiliation(s)
- Liselot De Vlieger
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Lisa Nuyttens
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Cheyenne Keppens
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Toon Ieven
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Division of Allergy and Clinical Immunology, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | - Marianne Diels
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Sophie Verelst
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Pediatrics, Jessa Hospital, Hasselt, Belgium
| | - Marc Raes
- Department of Pediatrics, Jessa Hospital, Hasselt, Belgium
| | - Jasmine Leus
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Pediatrics, AZ Maria Middelares, Ghent, Belgium
| | - Katrien Coppens
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Pediatrics, Imelda Hospital, Bonheiden, Belgium
| | - Kate Sauer
- Department of Pediatrics, AZ Sint-Jan Hospital, Bruges, Belgium
| | - Ellen Dilissen
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Lieve Coorevits
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Division of Allergy and Clinical Immunology, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Glynis Frans
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | - Rik Schrijvers
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Division of Allergy and Clinical Immunology, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Dominique M A Bullens
- Department of Microbiology, Immunology and Transplantation, Allergy and Immunology Research Group, KU Leuven, Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Sindher SB, Nadeau KC, Chinthrajah RS, Leflein JG, Bégin P, Ohayon JA, Ponda P, Wambre E, Liu J, Khokhar FA, Akinlade B, Maloney J, Orengo JM, Hamilton JD, Kamal MA, Hooper AT, Patel N, Patel K, Laws E, Mannent LP, Radin AR. Efficacy and Safety of Dupilumab in Children With Peanut Allergy: A Multicenter, Open-Label, Phase II Study. Allergy 2025; 80:227-237. [PMID: 39673452 PMCID: PMC11724241 DOI: 10.1111/all.16404] [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: 12/13/2023] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Peanut allergy is a potentially life-threatening food allergy in children. This study explored whether dupilumab, a human monoclonal immunoglobulin (Ig)G4 antibody that blocks the activity of interleukin (IL)-4/IL-13, improved safety and desensitization to peanut exposure in children with peanut allergy. METHODS A Phase II, 24-week, multicenter, single-arm, open-label, proof-of-concept study was conducted in the USA and Canada (NCT03793608). Children/adolescents with peanut allergy received subcutaneous dupilumab 300 mg (≥ 60 kg) or 200 mg (≥ 20 to < 60 kg) every 2 weeks. The primary endpoint was the proportion of participants who passed a double-blind placebo-controlled food challenge (DBPCFC) with ≥ 444 mg (cumulative) of peanut protein at week 24. Secondary endpoints included safety measures (Consortium of Food Allergy Research grading system) and change from baseline in peanut-specific (ps)-IgG4, total IgE, and ps-IgE. RESULTS Twenty-four participants enrolled and received dupilumab: 75.0% were male, 79.2% were white, mean (standard deviation) age was 11.7 (3.3) years. Most (95.8%) participants had not received allergen immunotherapy. Two participants (8.3%) achieved the primary endpoint and passed the DBPCFC at week 24. Fifteen participants (62.5%) reported 66 treatment-emergent adverse events, all being mild or in moderate intensity. At the week 24 DBPCFC, 8 participants (33.3%) had a grade 2 allergic reaction (no grade 3 or above); 10 (41.7%) used adrenaline as a rescue medication. Dupilumab treatment resulted in a median reduction of total and ps-IgE of -54% and -49%, respectively, and a 0% change in ps-IgG4. CONCLUSIONS Dupilumab monotherapy treatment for 24 weeks did not improve desensitization to peanut exposure after food challenge.
Collapse
Affiliation(s)
- Sayantani B. Sindher
- Sean N. Parker Center for Allergy and Asthma Research at Stanford UniversityStanfordCaliforniaUSA
| | | | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford UniversityStanfordCaliforniaUSA
| | | | - Philippe Bégin
- Centre Hospitalier Universitaire Sainte‐JustineMontrealQuebecCanada
| | - Jason A. Ohayon
- Department of PediatricsMcMaster UniversityHamiltonOntarioCanada
| | - Punita Ponda
- Division of Allergy and ImmunologyChildren's Hospital at MontefioreBronxNew YorkUSA
| | - Erik Wambre
- Translational Research ProgramBenaroya Research Institute at Virginia MasonSeattleWashingtonUSA
| | - Jinzhong Liu
- Regeneron Pharmaceuticals Inc.TarrytownNew YorkUSA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Kraft M, Saleh S, Heine G, Scheffold A, Bacher P, Worm M. Detection and Characterisation of Wasp Venom-Specific T Cells Using the ARTE Method in Allergic Patients. Clin Exp Allergy 2024. [PMID: 39675967 DOI: 10.1111/cea.14606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Affiliation(s)
- Magdalena Kraft
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Samira Saleh
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Guido Heine
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Margitta Worm
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
24
|
Zou X, Wang K, Deng Y, Guan P, Pu Q, Wang Y, Mou J, Du Y, Lou X, Wang S, Jiang N, Zhou S, Wang H, Du D, Liu X, Hu H, Zhang H. Hypoxia-inducible factor 2α promotes pathogenic polarization of stem-like Th2 cells via modulation of phospholipid metabolism. Immunity 2024; 57:2808-2826.e8. [PMID: 39609127 DOI: 10.1016/j.immuni.2024.11.001] [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: 03/10/2023] [Revised: 07/10/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
T helper 2 (Th2) cells orchestrate immunity against parasite infection and promote tissue repair but promote pathology in asthma and tissue fibrosis. Here, we examined the mechanisms driving pathogenic differentiation of Th2 cells. Single-cell analyses of CD4+ T cells from asthma and chronic rhinosinusitis patients revealed high expression of the hypoxia-inducible factor (HIF)2α in Th2 cells. In mice, HIF2α deficiency impaired Th2 differentiation and alleviated asthmatic inflammation. Single-cell and lineage tracing approaches delineated a differentiation trajectory from TCF1+Ly108+ stem-like Th2 cells to the ST2+CD25+ pathogenic progeny, depending on a HIF2α-GATA3 circuit that modulated phospholipid metabolism and T cell receptor (TCR)-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) activation via transcriptional regulation of the inositol polyphosphate multikinase (IPMK). Overexpression of IPMK in HIF2α-deficient cells promoted Phosphatidylinositol (3,4,5)-trisphosphate (PIP3) synthesis and pathogenic Th2 cell differentiation, whereas pharmacological inhibition of HIF2α impaired pathogenic differentiation of Th2 cells and mitigated airway inflammation. Our findings provide insight into the contextual cues that promote Th2-mediated pathology and suggest HIF2α as a therapeutic target in asthma.
Collapse
Affiliation(s)
- Xinkai Zou
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Keyue Wang
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujun Deng
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pengbo Guan
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qianlun Pu
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuemeng Wang
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jun Mou
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Infectious Diseases and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yizhou Du
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoxian Lou
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sijiao Wang
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Jiang
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
| | - Dan Du
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing 401329, China.
| | - Hongbo Hu
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Chongqing International Institute for Immunology, Chongqing, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610041, China.
| | - Huiyuan Zhang
- Center for Immunology and Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
25
|
Sen Chaudhuri A, Sun J. Lung-resident lymphocytes and their roles in respiratory infections and chronic respiratory diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:214-223. [PMID: 39834580 PMCID: PMC11742555 DOI: 10.1016/j.pccm.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Indexed: 01/22/2025]
Abstract
Recent scientific breakthroughs have blurred traditional boundaries between innate and adaptive immunity, revealing a sophisticated network of tissue-resident cells that deliver immediate, localized immune responses. These lymphocytes not only provide rapid frontline defense but also present a paradoxical role in the pathogenesis of respiratory diseases such as asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and the long-term tissue consequences of viral infections including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This review traverses the intricate landscape of lung-resident lymphocytes, delving into their origins, diverse functions, and their dualistic impact on pulmonary health. We dissect their interactions with the microenvironment and the regulatory mechanisms guiding their activity, with an emphasis on their contribution to both immune protection and immunopathology. This review aims to elucidate the complex narrative of these cells, enhancing our understanding of the development of precise therapeutic strategies to combat acute and chronic pulmonary diseases. Through this exploration, the review aspires to shed light on the potential of harnessing lung-resident lymphocytes for the treatment of respiratory conditions.
Collapse
Affiliation(s)
- Arka Sen Chaudhuri
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
26
|
Feng X, Flüchter P, De Tenorio JC, Schneider C. Tuft cells in the intestine, immunity and beyond. Nat Rev Gastroenterol Hepatol 2024; 21:852-868. [PMID: 39327439 DOI: 10.1038/s41575-024-00978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Tuft cells have gained substantial attention over the past 10 years due to numerous reports linking them with type 2 immunity and microorganism-sensing capacity in many mucosal tissues. This heightened interest is fuelled by their unique ability to produce an array of biological effector molecules, including IL-25, allergy-related eicosanoids, and the neurotransmitter acetylcholine, enabling downstream responses in diverse cell types. Operating through G protein-coupled receptor-mediated signalling pathways reminiscent of type II taste cells in oral taste buds, tuft cells emerge as chemosensory sentinels that integrate luminal conditions, eliciting appropriate responses in immune, epithelial and neuronal populations. How tuft cells promote tissue alterations and adaptation to the variety of stimuli at mucosal surfaces has been explored in multiple studies in the past few years. Since the initial recognition of the role of tuft cells, the discovery of diverse tuft cell effector functions and associated feedback loops have also revealed the complexity of tuft cell biology. Although earlier work largely focused on extraintestinal tissues, novel genetic tools and recent mechanistic studies on intestinal tuft cells established fundamental concepts of tuft cell activation and functions. This Review is an overview of intestinal tuft cells, providing insights into their development, signalling and interaction modules in immunity and other states.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Flüchter
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
27
|
Huang Y, Wang S, Huang J, Shen Y, Zou L, Liu H. Investigating the Causal Relationship Between Gut Microbiota and Allergic Conjunctivitis: A Two-Sample Mendelian Randomization Study. Ocul Immunol Inflamm 2024; 32:2411-2420. [PMID: 39353056 DOI: 10.1080/09273948.2024.2388202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE To investigate the causal association between gut microbiota and allergic conjunctivitis. METHODS A two-sample Mendelian randomization (MR) analysis was performed using the summary statistics of gut microbiota (18,340) from MiBio-Gen consortium and allergic conjunctivitis data (n = 218,792) obtained from the IEU Open GWAS project. F-statistics and sensitivity analyses were used to address potential biases and ensure the reliability of our findings. Reverse MR analysis was conducted to assess the possible of reverse causal relationships. RESULTS The inverse variance weighted estimates revealed the protective potential of the phylum Euryarchaeota against allergic conjunctivitis (OR = 0.87, p = 6.17 × 10-4). On the other hand, the genus Christensenellaceae R.7 group (OR = 0.75, p = 2.89 × 10-3), family Peptostreptococcaceae (OR = 0.83, p = 6.22 × 10-3), genus Lachnospiraceae FCS020 group (OR = 0.82, p = 0.02) all showed a suggestive protective association with allergic conjunctivitis. Additionally, sensitivity analysis confirmed the robustness of the above associations. In the reverse MR analysis, no significant causal association was found between gut microbiota and allergic conjunctivitis. CONCLUSION This study has revealed a potential causal correlation between the phylum Euryarchaeota and allergic conjunctivitis, offering new insights to improve prevention and treatment of this condition.
Collapse
Affiliation(s)
- Yuanyang Huang
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Wang
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinfang Huang
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yining Shen
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leilei Zou
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Liu
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Zhou X, Simonin EM, Jung YS, Galli SJ, Nadeau KC. Role of allergen immunotherapy and biologics in allergic diseases. Curr Opin Immunol 2024; 91:102494. [PMID: 39357079 PMCID: PMC11609009 DOI: 10.1016/j.coi.2024.102494] [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: 04/16/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
The rise in the prevalence of allergic diseases has become a global health burden. Allergic diseases are a group of immune-mediated disorders characterized by IgE-mediated conditions resulting from a type 2 helper T cell (Th2)-skewed immune response. This review aims to comprehensively summarize recent research on the roles of allergen immunotherapy (AIT) and biologics in allergic diseases. Specifically, we review the mechanisms of AIT and biologics in modulating innate and adaptive immunity involved in allergic disease pathogenesis, as well as their safety and efficacy in the treatment of allergic diseases. We also discuss current new AIT strategies such as recombinant allergen-based vaccines and allergen extract nanoencapsulation. Further research is needed to understand immune tolerance mechanisms beyond the Th2 pathway and to characterize immunological changes in responders and nonresponders to AIT or biologics. This additional research may uncover new targets for monitoring treatment responses and developing personalized treatment strategies for allergic diseases.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Elisabeth M Simonin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Youn S Jung
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Stephen J Galli
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
29
|
Pogorelov D, Bode SFN, He X, Ramiro-Garcia J, Hedin F, Ammerlaan W, Konstantinou M, Capelle CM, Zeng N, Poli A, Domingues O, Montamat G, Hunewald O, Ciré S, Baron A, Longworth J, Demczuk A, Bazon ML, Casper I, Klimek L, Neuberger-Castillo L, Revets D, Guyonnet L, Delhalle S, Zimmer J, Benes V, Codreanu-Morel F, Lehners-Weber C, Weets I, Alper P, Brenner D, Gutermuth J, Guerin C, Morisset M, Hentges F, Schneider R, Shamji MH, Betsou F, Wilmes P, Glaab E, Cosma A, Goncalves J, Hefeng FQ, Ollert M. Multiomics approaches disclose very-early molecular and cellular switches during insect-venom allergen-specific immunotherapy: an observational study. Nat Commun 2024; 15:10266. [PMID: 39592626 PMCID: PMC11599746 DOI: 10.1038/s41467-024-54684-2] [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: 02/01/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Allergen-specific immunotherapy (AIT) induces immune tolerance, showing the highest success rate (>95%) for insect venom while a much lower chance for pollen allergy. However, the molecular switches leading to successful durable tolerance restoration remain elusive. The primary outcome of this observational study is the comprehensive immunological cellular characterization during the AIT initiation phase, whereas the secondary outcomes are the serological and Th2-cell-type-specific transcriptomic analyses. Here we apply a multilayer-omics approach to reveal dynamic peripheral immune landscapes during the AIT-initiation phase in venom allergy patients (VAP) versus pollen-allergic and healthy controls. Already at baseline, VAP exhibit altered abundances of several cell types, including classical monocytes (cMono), CD4+ hybrid type 1-type 17 cells (Th1-Th17 or Th1/17) and CD8+ counterparts (Tc1-Tc17 or Tc1/17). At 8-24 h following AIT launch in VAP, we identify a uniform AIT-elicited pulse of late-transitional/IL-10-producing B cells, IL-6 signaling within Th2 cells and non-inflammatory serum-IL-6 levels. Sequential induction of activation and survival protein markers also immediately occur. A disequilibrium between serum IL-6 and cMono in VAP baseline is restored at day seven following AIT launch. Our longitudinal analysis discovers molecular switches during initiation-phase insect-venom AIT that secure long-term outcomes. Trial number: NCT02931955.
Collapse
Affiliation(s)
- Dimitrii Pogorelov
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Center of Allergy & Environment, Technical University of Munich, Munich, Germany
| | - Sebastian Felix Nepomuk Bode
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Centre-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Xin He
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Javier Ramiro-Garcia
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fanny Hedin
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Wim Ammerlaan
- Integrated BioBank of Luxembourg, Luxembourg Institute of Health, Dudelange, Luxembourg
| | - Maria Konstantinou
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Christophe M Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Olivia Domingues
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Oliver Hunewald
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Séverine Ciré
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Alexandre Baron
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Agnieszka Demczuk
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Murilo Luiz Bazon
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Ingrid Casper
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | | | - Dominique Revets
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Lea Guyonnet
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Cytometry Platform, Institut Curie; Innovative Therapies in Haemostasis, INSERM, Université de Paris, Paris, France
| | - Sylvie Delhalle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Françoise Codreanu-Morel
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Christiane Lehners-Weber
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Ilse Weets
- Department of Clinical Biology/ Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Pinar Alper
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Jan Gutermuth
- Department of Dermatology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Coralie Guerin
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Cytometry Platform, Institut Curie; Innovative Therapies in Haemostasis, INSERM, Université de Paris, Paris, France
| | - Martine Morisset
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
- Allergy Unit, Angers University Hospital, Angers, France
| | - François Hentges
- National Unit of Immunology-Allergology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, UK
| | - Fay Betsou
- Integrated BioBank of Luxembourg, Luxembourg Institute of Health, Dudelange, Luxembourg
- CRBIP, Institut Pasteur, Université Paris Cité, Paris, France
| | - Paul Wilmes
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Antonio Cosma
- National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jorge Goncalves
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Feng Q Hefeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
30
|
Wong DSH, Santos AF. The future of food allergy diagnosis. FRONTIERS IN ALLERGY 2024; 5:1456585. [PMID: 39575109 PMCID: PMC11578968 DOI: 10.3389/falgy.2024.1456585] [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: 06/28/2024] [Accepted: 10/07/2024] [Indexed: 11/24/2024] Open
Abstract
Food allergy represents an increasing global health issue, significantly impacting society on a personal and on a systems-wide level. The gold standard for diagnosing food allergy, the oral food challenge, is time-consuming, expensive, and carries risks of allergic reactions, with unpredictable severity. There is, therefore, an urgent need for more accurate, scalable, predictive diagnostic techniques. In this review, we discuss possible future directions in the world of food allergy diagnosis. We start by describing the current clinical approach to food allergy diagnosis, highlighting novel diagnostic methods recommended for use in clinical practice, such as the basophil activation test and molecular allergology, and go on to discuss tests that require more research before they can be applied to routine clinical use, including the mast cell activation test and bead-based epitope assay. Finally, we consider exploratory approaches, such as IgE glycosylation, IgG4, T and B cell assays, microbiome analysis, and plasma cytokines. Artificial intelligence is assessed for potential integrated interpretation of panels of diagnostic tests. Overall, a framework is proposed suggesting how combining established and emerging technologies can effectively enhance the accuracy of food allergy diagnosis in the future.
Collapse
Affiliation(s)
- Dominic S. H. Wong
- King's College London GKT School of Medical Education, London, United Kingdom
| | - Alexandra F. Santos
- Department of Women and Children’s Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| |
Collapse
|
31
|
Virkud YV, Styles JN, Kelly RS, Patil SU, Ruiter B, Smith NP, Clish C, Wheelock CE, Celedón JC, Litonjua AA, Bunyavanich S, Weiss ST, Baker ES, Lasky-Su JA, Shreffler WG. Immunomodulatory metabolites in IgE-mediated food allergy and oral immunotherapy outcomes based on metabolomic profiling. Pediatr Allergy Immunol 2024; 35:e14267. [PMID: 39530396 PMCID: PMC11756372 DOI: 10.1111/pai.14267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The immunometabolic mechanisms underlying variable responses to oral immunotherapy (OIT) in patients with IgE-mediated food allergy are unknown. OBJECTIVE To identify novel pathways associated with tolerance in food allergy, we used metabolomic profiling to find pathways important for food allergy in multiethnic cohorts and responses to OIT. METHODS Untargeted plasma metabolomics data were generated from the VDAART healthy infant cohort (N = 384), a Costa Rican cohort of children with asthma (N = 1040), and a peanut OIT trial (N = 20) evaluating sustained unresponsiveness (SU, protection that lasts after therapy) versus transient desensitization (TD, protection that ends immediately afterward). Generalized linear regression modeling and pathway enrichment analysis identified metabolites associated with food allergy and OIT outcomes. RESULTS Compared with unaffected children, those with food allergy were more likely to have metabolomic profiles with altered histidines and increased bile acids. Eicosanoids (e.g., arachidonic acid derivatives) (q = 2.4 × 10-20) and linoleic acid derivatives (q = 3.8 × 10-5) pathways decreased over time on OIT. Comparing SU versus TD revealed differing concentrations of bile acids (q = 4.1 × 10-8), eicosanoids (q = 7.9 × 10-7), and histidine pathways (q = .015). In particular, the bile acid lithocholate (4.97 [1.93, 16.14], p = .0027), the eicosanoid leukotriene B4 (3.21 [1.38, 8.38], p = .01), and the histidine metabolite urocanic acid (22.13 [3.98, 194.67], p = .0015) were higher in SU. CONCLUSIONS We observed distinct profiles of bile acids, histidines, and eicosanoids that vary among patients with food allergy, over time on OIT and between SU and TD. Participants with SU had higher levels of metabolites such as lithocholate and urocanic acid, which have immunomodulatory roles in key T-cell subsets, suggesting potential mechanisms of tolerance in immunotherapy.
Collapse
Affiliation(s)
- Yamini V. Virkud
- Department of Pediatrics, Division of Allergy and Immunology, Food Allergy Initiative, University of North Carolina, Chapel Hill, North Carolina, USA
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Jennifer N. Styles
- Department of Pediatrics, Division of Allergy and Immunology, Food Allergy Initiative, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Rachel S. Kelly
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Sarita U. Patil
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Bert Ruiter
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Neal P. Smith
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clary Clish
- Broad Institute, Cambridge, Massachusetts, USA
| | - Craig E. Wheelock
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Juan C. Celedón
- Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center. Division of Pulmonary Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Augusto A. Litonjua
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Supinda Bunyavanich
- Icahn School of Medicine at Mount Sinai, Department of Genetics & Genomic Sciences and Department of Pediatrics, New York, New York, USA
| | - Scott T. Weiss
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Erin S. Baker
- University of North Carolina, Chapel Hill, Department of Chemistry, Chapel Hill, North Carolina, USA
| | - Jessica A. Lasky-Su
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Wayne G. Shreffler
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
32
|
Vázquez-Mera S, Martelo-Vidal L, Miguéns-Suárez P, Bravo SB, Saavedra-Nieves P, Arias P, Ferreiro-Posse A, Vázquez-Lago J, Salgado FJ, González-Barcala FJ, Nieto-Fontarigo JJ. Exploring CD26 -/lo subpopulations of lymphocytes in asthma phenotype and severity: A novel CD4 + T cell subset expressing archetypical granulocyte proteins. Allergy 2024; 79:3005-3021. [PMID: 39319599 DOI: 10.1111/all.16327] [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: 06/27/2024] [Revised: 08/20/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Asthma pathology may induce changes in naïve/memory lymphocyte proportions assessable through the evaluation of surface CD26 (dipeptidyl peptidase 4/DPP4) levels. Our aim was to investigate the association of asthma phenotype/severity with the relative frequency of CD26-/lo, CD26int and CD26hi subsets within different lymphocyte populations. METHODS The proportion of CD26-/lo, CD26int and CD26hi subsets within CD4+ effector T cells (Teff), total CD4- lymphocytes, γδ-T cells, NK cells and NKT cells was measured in peripheral blood samples from healthy (N = 30) and asthma (N = 119) donors with different phenotypes/severities by flow cytometry. We performed K-means clustering analysis and further characterised the CD4+CD26-/lo Teff cell subset by LC-MS/MS and immunofluorescence. RESULTS Cluster analysis including clinical and flow cytometry data resulted in four groups, two of them with opposite inflammatory profiles (neutrophilic vs. eosinophilic). Neutrophilic asthma presented reduced CD4-CD26hi cells, which negatively correlated with systemic inflammation. Eosinophilic asthma displayed a general expansion of CD26-/lo subsets. Specifically, CD4+CD26-/lo Teff expansion was confirmed in asthma, especially in atopic patients. Proteomic characterisation of this subset with a TEM/TEMRA phenotype revealed upregulated levels of innate (e.g. MPO and RNASE2) and cytoskeleton/extracellular matrix (e.g. MMP9 and ACTN1) proteins. Immunofluorescence assays confirmed the presence of atypical proteins for CD4+ T cells, and an enrichment in 'flower-like' nuclei and MMP9/RNASE2 levels in CD4+CD26-/lo Teff compared to CD4+ T lymphocytes. CONCLUSION There is an association between CD26 levels in different lymphocyte subsets and asthma phenotype/severity. CD4+CD26-/loTEMRA cells expressing innate proteins specific to eosinophils/neutrophils could be determinant in sustaining long-term inflammation in adult allergic asthma.
Collapse
Affiliation(s)
- Sara Vázquez-Mera
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Laura Martelo-Vidal
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Miguéns-Suárez
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Susana Belén Bravo
- Proteomic Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Paula Saavedra-Nieves
- Department of Statistics, Mathematical Analysis and Optimization, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Pilar Arias
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Antía Ferreiro-Posse
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan Vázquez-Lago
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisco Javier Salgado
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Javier González-Barcala
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Medicine, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan José Nieto-Fontarigo
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
33
|
Ben-Baruch Morgenstern N, Rochman Y, Caldwell JM, Collins MH, Mukkada VA, Putnam PE, Bolton SM, Kliewer KL, Rothenberg ME. T H2 cell compensatory effect following benralizumab treatment for eosinophilic gastritis. J Allergy Clin Immunol 2024; 154:1325-1332.e2. [PMID: 39089335 DOI: 10.1016/j.jaci.2024.07.018] [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: 03/22/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Eosinophil accumulation is a main feature of eosinophilic gastritis (EoG) and is associated with its histologic diagnosis and pathology. However, a recent clinical trial has demonstrated that EoG endoscopic, noneosinophil histologic, and clinical features remain persistent despite complete eosinophil depletion. OBJECTIVE Our aim was to examine gastric T-cell composition and associated cytokine levels of patients with EoG following benralizumab-induced eosinophil depletion versus following administration of placebo. METHODS A cohort of subjects with EoG from a subset of subjects who participated in a recent phase 2 benralizumab trial was treated for 12 weeks with administration of 3 doses of benralizumab (anti-IL-5 receptor α antibody [n = 5]) or placebo (n = 4). Single-cell suspensions obtained by gastric biopsy were stimulated with phorbol 12,13-dibutyrate and ionomycin in the presence of brefeldin A and monensin. Harvested cells were fixed, stained, and analyzed by flow cytometry to examine T-cell populations and associated cytokines. RESULTS Following benralizumab treatment but not placebo, blood and gastric eosinophil levels decreased 16-fold and 10-fold, respectively. Whereas histologic score and features were significantly decreased, no change was observed in endoscopic score and features. Following complete eosinophil depletion with benralizumab, gastric TH2 cell levels were 3-fold higher than the levels in the patients with EoG who were given placebo; and the levels of associated type 2 cytokine production of IL-4, IL-5, and IL-13 in the benralizumab-treated patients were, respectively, 4-, 5.5-, and 2.5-fold, higher than those in the placebo-treated patients. CONCLUSION We have identified a putative positive feedback loop whereby eosinophil depletion results in a paradoxic increase in levels of TH2 cells and derived cytokines; this finding suggests an explanation for the limited success of eosinophil depletion as monotherapy in eosinophil-associated gastrointestinal disorders.
Collapse
Affiliation(s)
- Netali Ben-Baruch Morgenstern
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yrina Rochman
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julie M Caldwell
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Margaret H Collins
- Division of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Vincent A Mukkada
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip E Putnam
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Scott M Bolton
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kara L Kliewer
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
34
|
Ilangovan J, Neves JF, Santos AF. Innate lymphoid cells in immunoglobulin E-mediated food allergy. Curr Opin Allergy Clin Immunol 2024; 24:419-425. [PMID: 39132724 PMCID: PMC11356679 DOI: 10.1097/aci.0000000000001018] [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] [Indexed: 08/13/2024]
Abstract
PURPOSE OF REVIEW Recognition of the importance of innate lymphoid cells (ILCs) in the immune mechanisms of food allergy has grown in recent years. This review summarizes recent findings of ILCs in immunoglobulin E (IgE)-mediated food allergy. New research on ILCs in the context of the microbiome and other atopic diseases are also considered with respect to how they can inform understanding of the role of ILCs in food allergy. RECENT FINDINGS ILCs can mediate allergic and tolerogenic responses through multiple pathways. A novel subset of interleukin (IL)-10 producing ILC2s are associated with tolerance following immunotherapy to grass pollen, house dust mite allergy and lipid transfer protein allergy. ILC2s can drive food allergen-specific T cell responses in an antigen-specific manner. A memory subset of ILC2s has been identified through studies of other atopic diseases and is associated with effectiveness of response to therapy. SUMMARY The role of ILCs in food allergy and oral tolerance is relatively understudied compared to other diseases. ILCs can modulate immune responses through several mechanisms, and it is likely that these are of importance in the context of food allergy. Better understanding of theses pathways may help to answer fundamental questions regarding the development of food allergy and lead to novel therapeutic targets and treatment.
Collapse
Affiliation(s)
- Janarthanan Ilangovan
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine
- Centre for Host Microbiome Interactions
| | | | - Alexandra F. Santos
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London
- Children's Allergy Service, Guy's and St Thomas’ Hospital, London, UK
| |
Collapse
|
35
|
Ulrich BJ, Zhang W, Kenworthy BT, Kharwadkar R, Olson MR, Kaplan MH. Activin A Promotes Differentiation of a Pathogenic Multicytokine IL-9-secreting CD4+ T Cell Population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:823-830. [PMID: 39058312 PMCID: PMC11371476 DOI: 10.4049/jimmunol.2300635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024]
Abstract
The development of Th subsets results from cellular and cytokine cues that are present in the inflammatory environment. The developing T cell integrates multiple signals from the environment that sculpt the cytokine-producing capacity of the effector T cell. Importantly, T cells can discriminate similar cytokine signals to generate distinct outcomes, and that discrimination is critical in Th subset development. IL-9-secreting Th9 cells regulate multiple immune responses, including immunity to pathogens and tumors, allergic inflammation, and autoimmunity. In combination with IL-4, TGF-β or activin A promotes IL-9 production; yet, it is not clear if both TGF-β family members generate Th9 cells with identical phenotype and function. We observed that in contrast to TGF-β that efficiently represses Th2 cytokines in murine Th9 cultures, differentiation with activin A produced a multicytokine T cell phenotype with secretion of IL-4, IL-5, IL-13, and IL-10 in addition to IL-9. Moreover, multicytokine secreting cells are more effective at promoting allergic inflammation. These observations suggest that although TGF-β and IL-4 were identified as cytokines that stimulate optimal IL-9 production, they might not be the only cytokines that generate optimal function from IL-9-producing T cells in immunity and disease.
Collapse
Affiliation(s)
- Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Wenwu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Blake T Kenworthy
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Rakshin Kharwadkar
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Matthew R Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
36
|
Lang HP, Osum KC, Friedenberg SG. A review of CD4 + T cell differentiation and diversity in dogs. Vet Immunol Immunopathol 2024; 275:110816. [PMID: 39173398 PMCID: PMC11421293 DOI: 10.1016/j.vetimm.2024.110816] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
CD4+ T cells are an integral component of the adaptive immune response, carrying out many functions to combat a diverse range of pathogenic challenges. These cells exhibit remarkable plasticity, differentiating into specialized subsets such as T helper type 1 (TH1), TH2, TH9, TH17, TH22, regulatory T cells (Tregs), and follicular T helper (TFH) cells. Each subset is capable of addressing a distinct immunological need ranging from pathogen eradication to regulation of immune homeostasis. As the immune response subsides, CD4+ T cells rest down into long-lived memory phenotypes-including central memory (TCM), effector memory (TEM), resident memory (TRM), and terminally differentiated effector memory cells (TEMRA) that are localized to facilitate a swift and potent response upon antigen re-encounter. This capacity for long-term immunological memory and rapid reactivation upon secondary exposure highlights the role CD4+ T cells play in sustaining both adaptive defense mechanisms and maintenance. Decades of mouse, human, and to a lesser extent, pig T cell research has provided the framework for understanding the role of CD4+ T cells in immune responses, but these model systems do not always mimic each other. Although our understanding of pig immunology is not as extensive as mouse or human research, we have gained valuable insight by studying this model. More akin to pigs, our understanding of CD4+ T cells in dogs is much less complete. This disparity exists in part because canine immunologists depend on paradigms from mouse and human studies to characterize CD4+ T cells in dogs, with a fraction of available lineage-defining antibody markers. Despite this, every major CD4+ T cell subset has been described to some extent in dogs. These subsets have been studied in various contexts, including in vitro stimulation, homeostatic conditions, and across a range of disease states. Canine CD4+ T cells have been categorized according to lineage-defining characteristics, trafficking patterns, and what cytokines they produce upon stimulation. This review addresses our current understanding of canine CD4+ T cells from a comparative perspective by highlighting both the similarities and differences from mouse, human, and pig CD4+ T cell biology. We also discuss knowledge gaps in our current understanding of CD4+ T cells in dogs that could provide direction for future studies in the field.
Collapse
Affiliation(s)
- Haeree P Lang
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| | - Kevin C Osum
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA.
| | - Steven G Friedenberg
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| |
Collapse
|
37
|
Hung L, Zientara B, Berin MC. Contribution of T cell subsets to different food allergic diseases. Immunol Rev 2024; 326:35-47. [PMID: 39054597 DOI: 10.1111/imr.13368] [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: 07/27/2024]
Abstract
Food allergies occur due to a lack of tolerance to the proteins found in foods. While IgE- and non-IgE-mediated food allergies have different clinical manifestations, epidemiology, pathophysiology, and management, they share dysregulated T cell responses. Recent studies have shed light on the contributions of different T cell subsets to the development and persistence of different food allergic diseases. This review discusses the role of T cells in both IgE- and non-IgE-mediated food allergies and considers the potential future investigations in this context.
Collapse
Affiliation(s)
- Lisa Hung
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brianna Zientara
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
38
|
Niese ML, Pajulas AL, Rostron CR, Cheung CCL, Krishnan MS, Zhang J, Cannon AM, Kaplan MH. TL1A priming induces a multi-cytokine Th9 cell phenotype that promotes robust allergic inflammation in murine models of asthma. Mucosal Immunol 2024; 17:537-553. [PMID: 38493956 PMCID: PMC11354665 DOI: 10.1016/j.mucimm.2024.03.006] [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: 10/18/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Multi-cytokine-producing Th9 cells secrete IL-9 and type 2 cytokines and mediate mouse and human allergic inflammation. However, the cytokines that promote a multi-cytokine secreting phenotype have not been defined. Tumor necrosis factor superfamily member TL1A signals through its receptor DR3 to increase IL-9. Here we demonstrate that TL1A increases expression of IL-9 and IL-13 co-expressing cells in murine Th9 cell cultures, inducing a multi-cytokine phenotype. Mechanistically, this is linked to histone modifications allowing for increased accessibility at the Il9 and Il13 loci. We further show that TL1A alters the transcription factor network underlying expression of IL-9 and IL-13 in Th9 cells and increases binding of transcription factors to Il9 and Il13 loci. TL1A-priming enhances the pathogenicity of Th9 cells in murine models of allergic airway disease through the increased expression of IL-9 and IL-13. Lastly, in both chronic and memory-recall models of allergic airway disease, blockade of TL1A signaling decreases the multi-cytokine Th9 cell population and attenuates the allergic phenotype. Taken together, these data demonstrate that TL1A promotes the development of multi-cytokine Th9 cells that drive allergic airway diseases and that targeting pathogenic T helper cell-promoting cytokines could be an effective approach for modifying disease.
Collapse
Affiliation(s)
- Michelle L Niese
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abigail L Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cameron R Rostron
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cherry C L Cheung
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maya S Krishnan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony M Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
39
|
Patil SU, Dougan SK, Dougan M. Leaping toward Tolerance. NEJM EVIDENCE 2024; 3:EVIDe2400127. [PMID: 38804783 DOI: 10.1056/evide2400127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Affiliation(s)
- Sarita U Patil
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - Michael Dougan
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
40
|
Burk CM, Shreffler WG. Triggers for eosinophilic esophagitis (EoE): The intersection of food allergy and EoE. J Allergy Clin Immunol 2024; 153:1500-1509. [PMID: 38849185 PMCID: PMC11414349 DOI: 10.1016/j.jaci.2024.04.010] [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: 01/31/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024]
Abstract
Eosinophilic esophagitis and IgE-mediated food allergy are both food-triggered diseases that are increasing in prevalence. They share many clinical links, including significant comorbidity and similar food triggers, and as atopic diseases, they likely share upstream mechanisms related to barrier function and signals leading to TH2 skewing. In this review, we focus on links between eosinophilic esophagitis and IgE-mediated food allergy with an emphasis on what insights may be derived from overlapping food triggers and immune phenotypes. Through further investigation of these connections, we may be able to better understand not only IgE-mediated food allergy and eosinophilic esophagitis but also general atopic response to food proteins and evolution of allergic response to food.
Collapse
Affiliation(s)
- Caitlin M Burk
- Food Allergy Center, Division of Pediatric Allergy and Immunology, and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Wayne G Shreffler
- Food Allergy Center, Division of Pediatric Allergy and Immunology, and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| |
Collapse
|
41
|
Keswani T, LaHood NA, Marini-Rapoport O, Karmakar B, Andrieux L, Reese B, Sneed SL, Pedersen LC, Mueller GA, Patil SU. Neutralizing IgG 4 antibodies are a biomarker of sustained efficacy after peanut oral immunotherapy. J Allergy Clin Immunol 2024; 153:1611-1620.e7. [PMID: 38460677 PMCID: PMC11894811 DOI: 10.1016/j.jaci.2024.02.017] [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: 08/11/2023] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Clinical efficacy of oral immunotherapy (OIT) has been associated with the induction of blocking antibodies, particularly those capable of disrupting IgE-allergen interactions. Previously, we identified mAbs to Ara h 2 and structurally characterized their epitopes. OBJECTIVE We investigated longitudinal changes during OIT in antibody binding to conformational epitopes and correlated the results with isotype and clinical efficacy. METHODS We developed an indirect inhibitory ELISA using mAbs to block conformational epitopes on immobilized Ara h 2 from binding to serum immunoglobulins from peanut-allergic patients undergoing OIT. We tested the functional blocking ability of mAbs using passive cutaneous anaphylaxis in mice with humanized FcεRI receptors. RESULTS Diverse serum IgE recognition of Ara h 2 conformational epitopes are similar before and after OIT. Optimal inhibition of serum IgE occurs with the combination of 2 neutralizing mAbs (nAbs) recognizing epitopes 1.2 and 3, compared to 2 nonneutralizing mAbs (non-nAbs). After OIT, IgG4 nAbs, but not IgG1 or IgG2 nAbs, increased in sustained compared to transient outcomes. Induction of IgG4 nAbs occurs after OIT only in those with sustained efficacy. Murine passive cutaneous anaphylaxis after sensitization with pooled human sera is significantly inhibited by nAbs compared to non-nAbs. CONCLUSIONS Serum IgE conformational epitope diversity remains unchanged during OIT. However, IgG4 nAbs capable of uniquely disrupting IgE-allergen interactions to prevent effector cell activation are selectively induced in OIT-treated individuals with sustained clinical efficacy. Therefore, the induction of neutralizing IgG4 antibodies to Ara h 2 are clinically relevant biomarkers of durable efficacy in OIT.
Collapse
Affiliation(s)
- Tarun Keswani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Nicole A LaHood
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Orlee Marini-Rapoport
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Bijoya Karmakar
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Léna Andrieux
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass; Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Brian Reese
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Sunny L Sneed
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Lars C Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC
| | - Geoffrey A Mueller
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC
| | - Sarita U Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass.
| |
Collapse
|
42
|
Virkud YV, Styles JN, Kelly RS, Patil SU, Ruiter B, Smith NP, Clish C, Wheelock CE, Celedón JC, Litonjua AA, Bunyavanich S, Weiss ST, Baker ES, Lasky-Su JA, Shreffler WG. Metabolomics of IgE-Mediated Food Allergy and Oral Immunotherapy Outcomes based on Metabolomic Profiling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.31.24308233. [PMID: 38952781 PMCID: PMC11216533 DOI: 10.1101/2024.05.31.24308233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Background The immunometabolic mechanisms underlying variable responses to oral immunotherapy (OIT) in patients with IgE-mediated food allergy are unknown. Objective To identify novel pathways associated with tolerance in food allergy, we used metabolomic profiling to find pathways important for food allergy in multi-ethnic cohorts and responses to OIT. Methods Untargeted plasma metabolomics data were generated from the VDAART healthy infant cohort (N=384), a Costa Rican cohort of children with asthma (N=1040), and a peanut OIT trial (N=20) evaluating sustained unresponsiveness (SU, protection that lasts after therapy) versus transient desensitization (TD, protection that ends immediately afterwards). Generalized linear regression modeling and pathway enrichment analysis identified metabolites associated with food allergy and OIT outcomes. Results Compared with unaffected children, those with food allergy were more likely to have metabolomic profiles with altered histidines and increased bile acids. Eicosanoids (e.g., arachidonic acid derivatives) (q=2.4×10 -20 ) and linoleic acid derivatives (q=3.8×10 -5 ) pathways decreased over time on OIT. Comparing SU versus TD revealed differing concentrations of bile acids (q=4.1×10 -8 ), eicosanoids (q=7.9×10 -7 ), and histidine pathways (q=0.015). In particular, the bile acid lithocholate (4.97[1.93,16.14], p=0.0027), the eicosanoid leukotriene B4 (3.21[1.38,8.38], p=0.01), and the histidine metabolite urocanic acid (22.13[3.98,194.67], p=0.0015) were higher in SU. Conclusions We observed distinct profiles of bile acids, histidines, and eicosanoids that vary among patients with food allergy, over time on OIT and between SU and TD. Participants with SU had higher levels of metabolites such as lithocholate and urocanic acid, which have immunomodulatory roles in key T-cell subsets, suggesting potential mechanisms of tolerance in immunotherapy. Key Messages - Compared with unaffected controls, children with food allergy demonstrated higher levels of bile acids and distinct histidine/urocanic acid profiles, suggesting a potential role of these metabolites in food allergy. - In participants receiving oral immunotherapy for food allergy, those who were able to maintain tolerance-even after stopping therapyhad lower overall levels of bile acid and histidine metabolites, with the exception of lithocholic acid and urocanic acid, two metabolites that have roles in T cell differentiation that may increase the likelihood of remission in immunotherapy. Capsule summary This is the first study of plasma metabolomic profiles of responses to OIT in individuals with IgE-mediated food allergy. Identification of immunomodulatory metabolites in allergic tolerance may help identify mechanisms of tolerance and guide future therapeutic development.
Collapse
|
43
|
Ghouri H, Habib A, Nazir Z, Lohana N, Akilimali A. Omalizumab for the reduction of allergic reactions to foods: a narrative review. FRONTIERS IN ALLERGY 2024; 5:1409342. [PMID: 38873398 PMCID: PMC11172673 DOI: 10.3389/falgy.2024.1409342] [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: 04/05/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
The frequency of food allergies varies between 2% and 10%, depending on characteristics including age, region, race, and method of diagnosis self-reported by patients or oral food challenges (OFCs). The most common allergies reported are tree nuts (1.2%), milk (1.9%), peanuts (2.2%), and shellfish (1.3%). Omalizumab injection has now been approved by the FDA for the treatment of immunoglobulin E-mediated food allergies in specific adults and children aged one year or older. This medication reduces the risk of allergic reactions (Type I), which can include anaphylaxis, when an individual accidentally encounters one or more food allergens. Omalizumab functions by binding to IgE and altering IgE-mediated pathways, which lessens IgE's capacity to cause allergic reactions. Promising outcomes from clinical trials and case studies include lowered anaphylactic risk and enhanced tolerance to allergens. Omalizumab, however, may have adverse effects; thus, close observation is required. Overall, this review sheds light on the efficacy, safety, and clinical implications of omalizumab, highlighting its potential as a useful intervention for IgE-mediated food allergies.
Collapse
Affiliation(s)
- Hafsa Ghouri
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ashna Habib
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Zainab Nazir
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Nimerta Lohana
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
| | | |
Collapse
|
44
|
Chen Z, Suo Y, Du X, Zhao X. Genetically predicted N-methylhydroxyproline levels mediate the association between naive CD8+ T cells and allergic rhinitis: a mediation Mendelian randomization study. Front Immunol 2024; 15:1396246. [PMID: 38846949 PMCID: PMC11153669 DOI: 10.3389/fimmu.2024.1396246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Background Allergic rhinitis (AR), a prevalent chronic inflammatory condition triggered by immunoglobulin E (IgE), involves pivotal roles of immune and metabolic factors in its onset and progression. However, the intricacies and uncertainties in clinical research render current investigations into their interplay somewhat inadequate. Objective To elucidate the causal relationships between immune cells, metabolites, and AR, we conducted a mediation Mendelian randomization (MR) analysis. Methods Leveraging comprehensive publicly accessible summary-level data from genome-wide association studies (GWAS), this study employed the two-sample MR research method to investigate causal relationships among 731 immune cell phenotypes, 1400 metabolite levels, and AR. Additionally, employing the mediation MR approach, the study analyzed potential mediated effect of metabolites in the relationships between immune cells and AR. Various sensitivity analysis methods were systematically employed to ensure the robustness of the results. Results Following false discovery rate (FDR) correction, we identified three immune cell phenotypes as protective factors for AR: Naive CD8br %CD8br (odds ratio (OR): 0.978, 95% CI = 0.966-0.990, P = 4.5×10-4), CD3 on CD39+ activated Treg (OR: 0.947, 95% CI = 0.923-0.972, P = 3×10-5), HVEM on CD45RA- CD4+ (OR: 0.967, 95% CI = 0.948-0.986, P = 4×10-5). Additionally, three metabolite levels were identified as risk factors for AR: N-methylhydroxyproline levels (OR: 1.219, 95% CI = 1.104-1.346, P = 9×10-5), N-acetylneuraminate levels (OR: 1.133, 95% CI = 1.061-1.211, P = 1.7×10-4), 1-stearoyl-2-arachidonoyl-gpc (18:0/20:4) levels (OR: 1.058, 95% CI = 1.029-1.087, P = 5×10-5). Mediation MR analysis indicated a causal relationship between Naive CD8br %CD8br and N-methylhydroxyproline levels, acting as a protective factor (OR: 0.971, 95% CI = 0.950-0.992, P = 8.31×10-3). The mediated effect was -0.00574, accounting for 26.1% of the total effect, with a direct effect of -0.01626. Naive CD8+ T cells exert a protective effect on AR by reducing N-methylhydroxyproline levels. Conclusion Our study, delving into genetic information, has substantiated the intricate connection between immune cell phenotypes and metabolite levels with AR. This reveals a potential pathway to prevent the onset of AR, providing guiding directions for future clinical investigations.
Collapse
Affiliation(s)
- Zhengjie Chen
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Ying Suo
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Xintao Du
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Xiaoyun Zhao
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
- Department of Respiratory and Critical Care Medicine, Chest Hospital of Tianjin University, Tianjin, China
- DeepinBreath Union Laboratory, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
45
|
Kratchmarov R, Djeddi S, Dunlap G, He W, Jia X, Burk CM, Ryan T, McGill A, Allegretti JR, Kataru RP, Mehrara BJ, Taylor EM, Agarwal S, Bhattacharyya N, Bergmark RW, Maxfield AZ, Lee S, Roditi R, Dwyer DF, Boyce JA, Buchheit KM, Laidlaw TM, Shreffler WG, Rao DA, Gutierrez-Arcelus M, Brennan PJ. TCF1-LEF1 co-expression identifies a multipotent progenitor cell (T H2-MPP) across human allergic diseases. Nat Immunol 2024; 25:902-915. [PMID: 38589618 PMCID: PMC11849131 DOI: 10.1038/s41590-024-01803-2] [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: 01/21/2024] [Accepted: 03/06/2024] [Indexed: 04/10/2024]
Abstract
Repetitive exposure to antigen in chronic infection and cancer drives T cell exhaustion, limiting adaptive immunity. In contrast, aberrant, sustained T cell responses can persist over decades in human allergic disease. To understand these divergent outcomes, we employed bioinformatic, immunophenotyping and functional approaches with human diseased tissues, identifying an abundant population of type 2 helper T (TH2) cells with co-expression of TCF7 and LEF1, and features of chronic activation. These cells, which we termed TH2-multipotent progenitors (TH2-MPP) could self-renew and differentiate into cytokine-producing effector cells, regulatory T (Treg) cells and follicular helper T (TFH) cells. Single-cell T-cell-receptor lineage tracing confirmed lineage relationships between TH2-MPP, TH2 effectors, Treg cells and TFH cells. TH2-MPP persisted despite in vivo IL-4 receptor blockade, while thymic stromal lymphopoietin (TSLP) drove selective expansion of progenitor cells and rendered them insensitive to glucocorticoid-induced apoptosis in vitro. Together, our data identify TH2-MPP as an aberrant T cell population with the potential to sustain type 2 inflammation and support the paradigm that chronic T cell responses can be coordinated over time by progenitor cells.
Collapse
Affiliation(s)
- Radomir Kratchmarov
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Djeddi
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenqin He
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaojiong Jia
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caitlin M Burk
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tessa Ryan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna McGill
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica R Allegretti
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raghu P Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Babak J Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin M Taylor
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Shailesh Agarwal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Neil Bhattacharyya
- Massachusetts Eye & Ear Institute, Harvard Medical School, Boston, MA, USA
| | - Regan W Bergmark
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Alice Z Maxfield
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Lee
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel Roditi
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel F Dwyer
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua A Boyce
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Buchheit
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick J Brennan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Kidane FA, Müller L, Rocha-Hasler M, Tu A, Stanek V, Campion N, Bartosik T, Zghaebi M, Stoshikj S, Gompelmann D, Spittler A, Idzko M, Eckl-Dorna J, Schneider S. Deep immune profiling of chronic rhinosinusitis in allergic and non-allergic cohorts using mass cytometry. Clin Immunol 2024; 262:110174. [PMID: 38462155 DOI: 10.1016/j.clim.2024.110174] [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: 01/19/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
Chronic rhinosinusitis (CRS) is a persistent nasal and paranasal sinus mucosa inflammation comprising two phenotypes, namely CRS with nasal polyps (CRSwNP) and without (CRSsNP). CRSwNP can be associated with asthma and hypersensitivity to non-steroidal anti-inflammatory drug (NSAID) in a syndrome known as NSAID-exacerbated respiratory disease (N-ERD). Furthermore, CRS frequently intertwines with respiratory allergies. This study investigated levels of 33 different nasal and serum cytokines and phenotypic characteristics of peripheral blood mononuclear cells (PBMCs) within cohorts of CRS patients (n = 24), additionally examining the influence of comorbid respiratory allergies by mass cytometry. N-ERD patients showed heightened type 2 nasal cytokine levels. Mass cytometry revealed increased activated naive B cell levels in CRSwNP and N-ERD, while resting naive B cells were higher in CRSsNP. Th2a cell levels were significantly elevated in allergic subjects, but not in CRS groups. In conclusion, there are distinct immunological features in PBMCs of CRS phenotypes and allergy.
Collapse
Affiliation(s)
- Fana Alem Kidane
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Lena Müller
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | | | - Aldine Tu
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Victoria Stanek
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Nicholas Campion
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Tina Bartosik
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Mohammed Zghaebi
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Slagjana Stoshikj
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Daniela Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Core Facilities, Medical University of Vienna, Vienna, Austria; Department of Surgery, Research Lab, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria.
| | - Sven Schneider
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Identification of a human multipotent T H2 progenitor. Nat Immunol 2024; 25:735-736. [PMID: 38632340 DOI: 10.1038/s41590-024-01818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
|
48
|
Kennedy KV, Muir AB, Ruffner MA. Pathophysiology of Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:119-128. [PMID: 38575212 DOI: 10.1016/j.iac.2023.12.001] [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: 04/06/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, progressive immune-mediated disease associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Research over the last 2 decades has dramatically furthered our understanding of the complex interplay between genetics, environmental exposures, and cellular and molecular interactions involved in EoE. This review provides an overview of our current understanding of EoE pathogenesis.
Collapse
Affiliation(s)
- Kanak V Kennedy
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Melanie A Ruffner
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Division of Pediatric Allergy and Immunology, Department of Pediatrics, The Children's Hospital of Philadelphia
| |
Collapse
|
49
|
Gazzinelli-Guimaraes PH, Dulek B, Swanson P, Lack J, Roederer M, Nutman TB. Single-cell molecular signature of pathogenic T helper subsets in type 2-associated disorders in humans. JCI Insight 2024; 9:e177720. [PMID: 38587077 PMCID: PMC11128205 DOI: 10.1172/jci.insight.177720] [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: 11/16/2023] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
To unravel the heterogeneity and molecular signature of effector memory Th2 cells (Tem2), we analyzed 23 individuals' PBMCs of filaria-infected (Filaria+) and 24 healthy volunteers (Filaria-), with or without coincident house dust mite (HDM) allergic sensitization. Flow cytometry revealed 3 CD4+ Tem subsets - CCR4+CCR6+CRTH2- Tem17, CCR4+CCR6-CRTH2+ Tem2, and CCR6+CCR4+CRTH2+ Tem17.2 - markedly enriched in Filaria+ individuals. These subsets were sorted and analyzed by multiomic single-cell RNA immunoprofiling. SingleR-annotated Th2 cells from Tem2 and Tem17.2 cell subsets had features of pathogenic Th2 effector cells based on their transcriptional signatures, with downregulated CD27 and elevated expression levels of ITGA4, IL17RB, HPGDS, KLRB1, PTGDR2, IL9R, IL4, IL5, and IL13 genes. When the Filaria+ individuals were subdivided based on their allergic status, Tem2 cells in HDM+Filaria+ individuals showed an overall reduction in TCR diversity, suggesting the occurrence of antigen-driven clonal expansion. Moreover, HDM+Filaria+ individuals showed not only an expansion in the frequency of both Tem2 and Tem17.2 cell subsets, but also a change in their molecular program by overexpressing GATA3, IL17RB, CLRF2, and KLRB1, as well as increased antigen-induced IL-4, IL-5, and IL-13 production, suggesting that aeroallergens reshape the transcriptional and functional programming of Th2 cell subsets in human filarial infection toward a pathogenic immunophenotype.
Collapse
Affiliation(s)
| | | | - Phillip Swanson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
50
|
Li JC, Rotter NS, Stieb ES, Stockbridge JL, Theodorakakis MD, Shreffler WG. Utility of food allergy thresholds. Ann Allergy Asthma Immunol 2024; 132:321-327. [PMID: 38114041 DOI: 10.1016/j.anai.2023.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
Food allergy is a prevalent disease worldwide that is a significant quality-of-life burden, and accidental exposures to food allergens may elicit severe, life-threatening reactions such as anaphylaxis. The threshold level, or the dose that triggers an allergic reaction determined by oral food challenges, varies considerably among individuals suffering from food allergies. Moreover, IgE concentration, diversity, or function can only partially explain this variation in threshold; pathogenic effector TH2 cells have also been found to contribute to the eliciting dose. Though very sensitive to cofactors such as physical activity/stress, the threshold is a stable and reproducible feature of an individual's allergy over periods of many months, made clear in the past several years from treatment studies in which repeated threshold determination has been used as a treatment outcome; however, there also seem to be age-related changes at a population level. More routine determination of food allergy thresholds may help patients stratify risk to improve the management of their food allergy. Precautionary allergen labeling, such as "may contain" labels, often causes confusion since they are inconsistent and regularly contain little to trace allergen residues; thus, food products with such labeling may be unnecessarily avoided. Population-based eliciting dose levels have been determined in the literature; patients at lower risk with higher thresholds may be more confident with introducing foods with precautionary allergen labels. Understanding a patient's threshold level could aid in shared decision-making to determine the most suitable treatment options for patients, including the starting dose for oral immunotherapy and/or the use of biologics.
Collapse
Affiliation(s)
- Jennifer C Li
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts
| | - Nancy S Rotter
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Elisabeth S Stieb
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer L Stockbridge
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts
| | - Maria D Theodorakakis
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Wayne G Shreffler
- Food Allergy Center, Massachusetts General Hospital, Boston, Massachusetts; Division of Pediatric Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|