1
|
Van Acker N, Frenois FX, Gravelle P, Tosolini M, Syrykh C, Laurent C, Brousset P. Spatial mapping of innate lymphoid cells in human lymphoid tissues and lymphoma at single-cell resolution. Nat Commun 2025; 16:4545. [PMID: 40374674 PMCID: PMC12081901 DOI: 10.1038/s41467-025-59811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Innate lymphoid cells (ILC) distribution and compartmentalization in human lymphoid tissues are incompletely described. Through combined multiplex immunofluorescence, multispectral imaging, and advanced computer vision methods, we provide a map of ILCs at the whole-slide single-cell resolution level, and study their proximity to T helper (Th) cells. The results show that ILC2 predominates in thymic medulla; by contrast, immature Th cells prevail in the cortex. Unexpectedly, we find that Th2-like and Th17-like phenotypes appear before complete T cell receptor gene rearrangements in these immature thymocytes. In the periphery, ILC2 are more abundant in lymph nodes and tonsils, penetrating lymphoid follicles. NK cells are uncommon in lymphoid tissues but abundant in the spleen, whereas ILC1 and ILC3 predominate in the ileum and appendix. Under pathogenic conditions, a deep perturbation of both ILC and Th populations is seen in follicular lymphoma compared with non-neoplastic conditions. Lastly, all ILCs are preferentially in close proximity to their Th counterparts. In summary, our histopathology tool help present a spatial mapping of human ILCs and Th cells, in normal and neoplastic lymphoid tissues.
Collapse
Affiliation(s)
- Nathalie Van Acker
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - François-Xavier Frenois
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pauline Gravelle
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Marie Tosolini
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Charlotte Syrykh
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Camille Laurent
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France
| | - Pierre Brousset
- Department of Pathology, CHU of Toulouse, Imag'IN Platform, IUCT-Oncopole, Toulouse, France.
- Cancer Research Center of Toulouse (INSERM), Team 9 NoLymIT and Labex TOUCAN, Toulouse, France.
| |
Collapse
|
2
|
Guichard V, Leão FB, Zhao J, Zhang Y, Ito T, Shirley S, Postler TS, Tian R, Huang Y, Ghosh S. Pre-existing epigenetic state and differential NF-κB activation shape type 2 immune cell responses. Immunity 2025:S1074-7613(25)00179-7. [PMID: 40367949 DOI: 10.1016/j.immuni.2025.04.016] [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: 03/04/2024] [Revised: 12/20/2024] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
CD4+ T helper 2 (Th2) cells and group 2 innate lymphoid cells (ILC2s) drive type 2 immune responses via similar effector molecules that are primarily induced by different signals-interleukin (IL)-33 in ILC2s and TCR engagement in Th2 cells. Here, we examined the transcriptional regulation of type 2 immunity, focusing on the NF-κB pathway, which is differentially activated by TCR engagement or cytokine signaling. Conditional deletion of the NF-κB subunits c-Rel and p65 limited the expression of key type 2 genes, including Il13 and Il5, in ILC2s but not in Th2 cells. Genome-wide analysis revealed that the regulatory regions of such genes exist in an open chromatin state in ILC2s, allowing NF-κB binding upon IL-33 stimulation. These regions are less accessible in unstimulated Th2 cells, where NFAT plays a dominant role. Accordingly, p65 deletion impaired ILC2 activation and function during airway inflammation and helminth infection. Thus, innate and adaptive lymphocytes leverage distinct epigenetic landscapes and transcriptional regulators to control shared effector genes.
Collapse
Affiliation(s)
- Vincent Guichard
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Felipe Batista Leão
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jingyao Zhao
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yingyu Zhang
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Takamasa Ito
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Simon Shirley
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Thomas S Postler
- Vaccine Design and Development Laboratory, IAVI, New York, NY 10004, USA
| | - Ruxiao Tian
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yuefeng Huang
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | - Sankar Ghosh
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
3
|
Gan H, Huang Z, Pan Q, Ye F, Zhu Z, Sun B. Recurrence of acute allergic asthma depends on the role of ILC2 driven by Il1rl1 signaling. Cell Commun Signal 2025; 23:215. [PMID: 40329299 PMCID: PMC12057255 DOI: 10.1186/s12964-025-02220-0] [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: 01/19/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Asthma is a chronic inflammatory airway disease characterized by recurrent episodes that significantly impair disease control and reduce patients' quality of life. Despite its clinical importance, the mechanisms underlying asthma relapse remain poorly understood, and effective strategies to prevent exacerbations are still lacking. METHODS An acute allergic asthma relapse mouse model was established using ovalbumin sensitization and challenge. Single-cell transcriptomics was employed to investigate the cellular and molecular mechanisms driving asthma relapse. Flow cytometry and gene knockout experiments were conducted to validate the findings. RESULTS We successfully established an acute allergic asthma relapse mouse model. Single-cell transcriptomic analysis revealed that T cells and type 2 innate lymphoid cells (ILC2s) are pivotal during asthma relapse, serving as the primary cellular sources of type 2 inflammatory cytokines. Further subcluster analysis identified T-cell subcluster 4 and ILC2 subcluster 0 as the predominant contributors to type 2 cytokine production. Complex intercellular communication networks were observed, with macrophages, natural killer (NK) cells, and dendritic cells functioning as central signaling hubs. Pseudo-time trajectory analysis highlighted the critical role of ILC2s and the Il1rl1 signaling pathway in asthma relapse. These findings were corroborated by flow cytometry. Il1rl1-deficient mice displayed similar pulmonary inflammation to wild-type mice during the initial asthma episode; however, asthma relapse was significantly attenuated. Mechanistically, Il1rl1 deficiency resulted in a substantial reduction in both the number and functional capacity of ILC2s. CONCLUSION The recurrence of acute allergic asthma is driven, at least in part, by ILC2s through Il1rl1 signaling. Genetic ablation of Il1rl1 significantly suppresses asthma relapse, suggesting that targeting Il1rl1 may represent a novel therapeutic strategy for preventing asthma exacerbations.
Collapse
Affiliation(s)
- Hui Gan
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhifeng Huang
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingjun Pan
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fei Ye
- Allergy Department/Otolaryngology Head and Neck Surgery, Department of Zhongshan People's Hospital, Zhongshan, 528403, PR China
| | - Zheng Zhu
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Allergy and Clinical Immunology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Baoqing Sun
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
4
|
Shirley S, Ichise H, Di Natale V, Jin J, Wu C, Zou R, Zhang W, Fang Y, Zhang Y, Chen M, Peng S, Basu U, Que J, Huang Y. A vasculature-resident innate lymphoid cell population in mouse lungs. Nat Commun 2025; 16:3718. [PMID: 40253407 PMCID: PMC12009297 DOI: 10.1038/s41467-025-58982-1] [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: 09/29/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Tissue-resident immune cells such as innate lymphoid cells (ILC) are known to reside in the parenchymal compartments of tissues and modulate local immune protection. Here we use intravascular cell labeling, parabiosis and multiplex 3D imaging to identify a population of group 3 ILCs in mice that are present within the intravascular space of lung blood vessels (vILC3). vILC3s are distributed broadly in alveolar capillary beds from which inhaled pathogens enter the lung parenchyma. By contrast, conventional ILC3s in tissue parenchyma are enriched in lymphoid clusters in proximity to large veins. In a mouse model of pneumonia, Pseudomonas aeruginosa infection results in rapid vILC3 expansion and production of chemokines including CCL4. Blocking CCL4 in vivo attenuates neutrophil recruitment to the lung at the early stage of infection, resulting in prolonged inflammation and delayed bacterial clearance. Our findings thus define the intravascular space as a site of ILC residence in mice, and reveal a unique immune cell population that interfaces with tissue alarmins and the circulating immune system for timely host defense.
Collapse
Affiliation(s)
- Simon Shirley
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Vincenzo Di Natale
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jiacheng Jin
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Christine Wu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Raymond Zou
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Wanwei Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yinshan Fang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA
| | - Yingyu Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miao Chen
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Sophia Peng
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Uttiya Basu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA.
| | - Yuefeng Huang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
5
|
Graf LM, Radtke D, Voehringer D. Regulation of eosinophil recruitment and heterogeneity during allergic airway inflammation. FRONTIERS IN ALLERGY 2025; 6:1585142. [PMID: 40276331 PMCID: PMC12018390 DOI: 10.3389/falgy.2025.1585142] [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: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Eosinophils represent a granulocyte cell type that is strongly associated with type 2 inflammatory conditions. During steady state conditions few eosinophils are found in lung tissue, though they may contribute to homeostasis. In allergic airway inflammation, eosinophils are strongly increased and associated to disease severity. The underlying type 2 immune response tightly regulates eosinophil development, recruitment, survival, and heterogeneity. Inflammatory eosinophils in the lung are unfavourable, as they can cause tissue damage, amplify type 2 immunity and induce bronchial obstruction by expelling granular proteins and cytokines. In this review we provide an overview about mechanisms regulating development of eosinophils in the bone marrow and their extravasation into the lung including recent findings on induction and diversity of eosinophilia in allergic airway inflammation.
Collapse
Affiliation(s)
| | | | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
6
|
Waizman DA, Brown-Soler I, Martin AL, Ma Y, Zhou K, Israni-Winger K, Zhang C, Medzhitov R, Launay P, Michieletto MF, Henao-Mejia J, Palm NW, Craft J, Eisenstein A, Wang A. Skin damage signals mediate allergic sensitization to spatially unlinked antigen. Sci Immunol 2025; 10:eadn0688. [PMID: 40184440 PMCID: PMC12100540 DOI: 10.1126/sciimmunol.adn0688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/10/2024] [Accepted: 03/13/2025] [Indexed: 04/06/2025]
Abstract
Our current understanding of immunity to pathogens suggests that anatomic coupling of antigens with danger signals is a required feature for the formation of immune memory. However, in the context of pathogen-independent inflammation, the stringency of this anatomical coupling is unclear. Here, we demonstrate that multiple modes of skin injury were sufficient to induce a humoral response to antigens introduced in the gut. Skin damage induced a narrow subset of endocrine cytokines that were necessary and sufficient for the priming of antigens introduced at various distal tissues. Thus, in addition to "local priming" of antigen entering through damaged skin, there also exists another paradigm of "remote priming" where anatomical coupling is not essential because of the dissemination of damage-associated intermediaries. Our findings have implications for understanding the fundamental mechanisms of the formation of humoral memory with wide implications for diseases such as food allergy and in vaccinology.
Collapse
Affiliation(s)
- Daniel A. Waizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Isabela Brown-Soler
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anjelica L. Martin
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yifan Ma
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kenneth Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Cuiling Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Pierre Launay
- Centre de Recherche sur l’Inflammation, INSERM UMR1149, CNRS EMR8252, Université Paris Cité, Paris, France
| | - Michaël F. Michieletto
- Institute for Immunology and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge Henao-Mejia
- Institute for Immunology and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, Perelman School of Medicine, and Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joe Craft
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Internal Medicine (Rheumatology, Allergy, and Immunology), Yale School of Medicine, New Haven, CT 06510, USA
| | - Anna Eisenstein
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Internal Medicine (Rheumatology, Allergy, and Immunology), Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
7
|
Olsthoorn SEM, van Krimpen A, Hendriks RW, Stadhouders R. Chronic Inflammation in Asthma: Looking Beyond the Th2 Cell. Immunol Rev 2025; 330:e70010. [PMID: 40016948 PMCID: PMC11868696 DOI: 10.1111/imr.70010] [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: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Asthma is a common chronic inflammatory disease of the airways. A substantial number of patients present with severe and therapy-resistant asthma, for which the underlying biological mechanisms remain poorly understood. In most asthma patients, airway inflammation is characterized by chronic activation of type 2 immunity. CD4+ T helper 2 (Th2) cells are the canonical producers of the cytokines that fuel type 2 inflammation: interleukin (IL)-4, IL-5, IL-9, and IL-13. However, more recent findings have shown that other lymphocyte subsets, in particular group 2 innate lymphoid cells (ILC2s) and type 2 CD8+ cytotoxic T (Tc2) cells, can also produce large amounts of type 2 cytokines. Importantly, a substantial number of severe therapy-resistant asthma patients present with chronic type 2 inflammation, despite the high sensitivity of Th2 cells for suppression by corticosteroids-the mainstay drugs for asthma. Emerging evidence indicates that ILC2s and Tc2 cells are more abundant in severe asthma patients and can adopt corticosteroid-resistance states. Moreover, many severe asthma patients do not present with overt type 2 airway inflammation, implicating non-type 2 immunity as a driver of disease. In this review, we will discuss asthma pathophysiology and focus on the roles played by ILC2s, Tc2 cells, and non-type 2 lymphocytes, placing special emphasis on severe disease forms.
Collapse
Affiliation(s)
- Simone E. M. Olsthoorn
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Anneloes van Krimpen
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
8
|
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: 11] [Impact Index Per Article: 11.0] [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
|
9
|
León B. Type 2 conventional dendritic cell functional heterogeneity: ontogenically committed or environmentally plastic? Trends Immunol 2025; 46:104-120. [PMID: 39843310 PMCID: PMC11835539 DOI: 10.1016/j.it.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/24/2025]
Abstract
Conventional dendritic cells (cDCs) are sentinels of the mammalian immune system that sense a wide range of danger and homeostatic signals to induce appropriately targeted T cell immune responses. Traditionally classified into two main subsets, cDC1 and cDC2, recent research shows that cDC2s exhibit significant heterogeneity and can be further subdivided. Studies in mice and humans show that, beyond their ontogeny, cDC2s acquire dynamic and tissue-specific characteristics that are influenced by local environmental signals, which impact on their functions during homeostasis, inflammation, and infection. The novel concept is proposed that tissue-derived signals and tissue plasticity can override preestablished developmental programming, thereby redefining developmental trajectories and cDC2 functionality. Ultimately, understanding cDC2 heterogeneity and plasticity has important implications for modulating T cell immunity in health and disease.
Collapse
Affiliation(s)
- Beatriz León
- Innate Cells and Th2 Immunity Section, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Liu H, Wu M, Wang Q, Gao L, Jiang H, Shi K, Lin Y, Zhou J, Huang J, Qu S, Zhang Y, Zheng F, Huang Y, Han J. IL-33 released during challenge phase regulates allergic asthma in an age-dependent way. Cell Mol Immunol 2025; 22:191-207. [PMID: 39134801 PMCID: PMC11782506 DOI: 10.1038/s41423-024-01205-2] [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: 01/31/2024] [Accepted: 07/12/2024] [Indexed: 02/01/2025] Open
Abstract
Epithelial-derived cytokines, especially type 2 alarmins (TSLP, IL-25, and IL-33), have emerged as critical mediators of type 2 inflammation. IL-33 attracts more interest for its strong association with allergic asthma, especially in childhood asthma. However, the age-dependent role of IL-33 to the development of allergic asthma remains elusive. Here, using OVA-induced allergic asthma model in neonatal and adult mice, we report that IL-33 is the most important alarmin in neonatal lung both at steady state or inflammation. The deficiency of IL-33/ST2 abrogated the development of allergic asthma only in neonates, whereas in adults the effect was limited. Interestingly, the deficiency of IL-33/ST2 equally dampened the ILC2 responses in both neonatal and adult models. However, the effect of IL-33/ST2 deficiency on Th2 responses is age-dependent, which is only blocked in neonates. Furthermore, IL-33/ST2 signaling is dispensable for OVA sensitization. Following OVA challenge in adults, the deficiency of IL-33/ST2 results in compensational more TSLP, which in turn recruits and activates lung DCs and boosts Th2 responses. The enriched γδ T17 cells in IL-33/ST2 deficient neonatal lung suppress the expression of type 2 alarmins, CCL20 and GM-CSF via IL-17A, thus might confer the inhibition of allergic asthma. Finally, on the basis of IL-33 deficiency, the additive protective effects of TSLP blocking is much more pronounced than IL-25 blocking in adults. Our studies demonstrate that the role of IL-33 for ILC2 and Th2 responses varies among ages in OVA models and indicate that the factor of age should be considered for intervention of asthma.
Collapse
Affiliation(s)
- Hangyu Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiangqiang Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuchuang Gao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ketai Shi
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yawen Lin
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ju Huang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Qu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwei Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Gurram RK, Li P, Oh J, Chen X, Spolski R, Yao X, Lin JX, Roy S, Liao MJ, Liu C, Yu ZX, Levine SJ, Zhu J, Leonard WJ. TSLP acts on regulatory T cells to maintain their identity and limit allergic inflammation. Sci Immunol 2025; 10:eadk0073. [PMID: 39792638 DOI: 10.1126/sciimmunol.adk0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 07/08/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025]
Abstract
Thymic stromal lymphopoietin (TSLP) is a type I cytokine that promotes allergic responses and mediates type 2 immunity. A balance between effector T cells (Teffs), which drive the immune response, and regulatory T cells (Tregs), which suppress the response, is required for proper immune homeostasis. Here, we report that TSLP differentially acts on Teffs versus Tregs to balance type 2 immunity. As expected, deletion of TSLP receptor (TSLPR) on all T cells (Cd4CreCrlf2fl/fl mice) resulted in lower numbers of T helper 2 (TH2) cells and diminished ovalbumin-induced airway inflammation, but selective deletion of TSLPR on Tregs (Foxp3YFP-Cre/YCrlf2fl/fl mice) resulted in increased interleukin-5 (IL-5)- and IL-13-secreting TH2 cells and lung eosinophilia. Moreover, TSLP augmented the expression of factors that stabilize Tregs. During type 2 immune responses, TSLPR-deficient Tregs acquired TH2-like properties, with augmented GATA3 expression and secretion of IL-13. TSLP not only is a driver of TH2 effector cells but also acts in a negative feedback loop, thus promoting the ability of Tregs to limit allergic inflammation.
Collapse
Affiliation(s)
- Rama K Gurram
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Peng Li
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jangsuk Oh
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xi Chen
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Rosanne Spolski
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xianglan Yao
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892-1674, USA
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Suyasha Roy
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Matthew J Liao
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Stewart J Levine
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892-1674, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Obata‐Ninomiya K, Jayaraman T, Ziegler SF. From the bench to the clinic: basophils and type 2 epithelial cytokines of thymic stromal lymphopoietin and IL-33. Clin Transl Immunology 2024; 13:e70020. [PMID: 39654685 PMCID: PMC11626414 DOI: 10.1002/cti2.70020] [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: 03/26/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 epithelial cytokines, including thymic stromal lymphopoietin and IL-33, play central roles in modulation of type 2 immune cells, such as basophils. Basophils are a small subset of granulocytes within the leukocyte population that predominantly exist in the blood. They have non-redundant roles in allergic inflammation in peripheral tissues such as the lung, skin and gut, where they increase and accumulate at inflammatory lesions and exclusively produce large amounts of IL-4, a type 2 cytokine. These inflammatory reactions are known to be, to some extent, phenocopies of infectious diseases of ticks and helminths. Recently, biologics related to both type 2 epithelial cytokines and basophils have been approved by the US Food and Drug Administration for treatment of allergic diseases. We summarised the roles of Type 2 epithelial cytokines and basophils in basic science to translational medicine, including recent findings.
Collapse
Affiliation(s)
| | | | - Steven F Ziegler
- Center of Fundamental ImmunologyBenaroya Research InstituteSeattleWAUSA
- Department of ImmunologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
13
|
Troch KF, Jakob MO, Forster PM, Jarick KJ, Schreiber J, Preusser A, Guerra GM, Durek P, Tizian C, Sterczyk N, Helfrich S, Duerr CU, Voehringer D, Witkowski M, Artis D, Rollenske T, Kruglov AA, Mashreghi MF, Klose CSN. Group 2 innate lymphoid cells are a non-redundant source of interleukin-5 required for development and function of murine B1 cells. Nat Commun 2024; 15:10566. [PMID: 39632879 PMCID: PMC11618303 DOI: 10.1038/s41467-024-54780-3] [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: 03/15/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Tissue-resident immune cells, such as innate lymphoid cells, mediate protective or detrimental immune responses at barrier surfaces. Upon activation by stromal or epithelial cell-derived alarmins, group 2 innate lymphoid cells (ILC2s) are a rapid source of type 2 cytokines, such as IL-5. However, due to the overlap in effector functions, it remains unresolved whether ILC2s are an essential component of the type 2 response or whether their function can be compensated by other cells, such as T cells. Here we show a non-redundant role of ILC2s in supporting the development and function of B1 cells. We demonstrate that B1 cells fail to develop properly in the absence of ILC2s and identify the IL-33 receptor on ILC2s as an essential cell-intrinsic regulator of IL-5 production. Further, conditional deletion of Il5 in ILC2s results in defective B1 cell development and immunoglobulin production. Consequently, B1 cells with phosphatidylcholine specific B cell receptor rearrangements are diminished in ILC2-deficient mice. Thus, our data establish an essential function of ILC2s in supporting B1 cells and antibody production at barrier surfaces.
Collapse
Affiliation(s)
- Karoline F Troch
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Manuel O Jakob
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Patrycja M Forster
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Katja J Jarick
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Jonathan Schreiber
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Alexandra Preusser
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Gabriela M Guerra
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Caroline Tizian
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Nele Sterczyk
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Sofia Helfrich
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - Claudia U Duerr
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Mario Witkowski
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tim Rollenske
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Andrey A Kruglov
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany
| | - Christoph S N Klose
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm 30, Berlin, Germany.
| |
Collapse
|
14
|
Kania AK, Kokkinou E, Pearce E, Pearce E. Metabolic adaptations of ILC2 and Th2 cells in type 2 immunity. Curr Opin Immunol 2024; 91:102503. [PMID: 39520759 DOI: 10.1016/j.coi.2024.102503] [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: 07/19/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Type 2 immune responses play a crucial role in host defense against parasitic infections but can also promote the development of allergies and asthma. This response is orchestrated primarily by group 2 innate lymphoid cells (ILC2) and helper type 2 (Th2) cells, both of which undergo substantial metabolic reprogramming as they transition from resting to activated states. Understanding these metabolic adaptations not only provides insights into the fundamental biology of ILC2 and Th2 cells but also opens up potential therapeutic avenues for the identification of novel metabolic targets that can extend the current treatment regimens for diseases in which type 2 immune responses play pivotal roles. By integrating recent findings, this review underscores the significance of cellular metabolism in orchestrating immune functions and highlights future directions for research in this evolving field.
Collapse
Affiliation(s)
- Anna K Kania
- Bloomberg Kimmel Institute of Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Efthymia Kokkinou
- Bloomberg Kimmel Institute of Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Erika Pearce
- Bloomberg Kimmel Institute of Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Edward Pearce
- Bloomberg Kimmel Institute of Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
15
|
Lopez Espinoza A, Christopher T, Tait Wojno ED. Epithelial-immune interactions govern type 2 immunity at barrier surfaces. Curr Opin Immunol 2024; 91:102501. [PMID: 39522453 PMCID: PMC11734749 DOI: 10.1016/j.coi.2024.102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Allergic diseases are acute and chronic inflammatory conditions resulting from disproportionate responses to environmental stimuli. Affecting approximately 40% of the global population, these diseases significantly contribute to morbidity and increasing health care costs. Allergic reactions are triggered by pollen, house dust mites, animal dander, mold, food antigens, venoms, toxins, and drugs. This review explores the pivotal role of the epithelium in the skin, lungs, and gastrointestinal tract in regulating the allergic response and delves into the mechanisms of tissue-specific epithelial-immune interactions in this context, with recent advances highlighting their roles in the initiation, elicitation, and resolution phases of allergy. Understanding these intricate interactions at epithelial barriers is essential for developing targeted therapies to manage and treat allergic diseases.
Collapse
Affiliation(s)
| | - Tighe Christopher
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elia D Tait Wojno
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
16
|
Kabil A, Nayyar N, Brassard J, Li Y, Chopra S, Hughes MR, McNagny KM. Microbial intestinal dysbiosis drives long-term allergic susceptibility by sculpting an ILC2-B1 cell-innate IgE axis. J Allergy Clin Immunol 2024; 154:1260-1276.e9. [PMID: 39134158 DOI: 10.1016/j.jaci.2024.07.023] [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: 02/27/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND The abundance and diversity of intestinal commensal bacteria influence systemic immunity with impact on disease susceptibility and severity. For example, loss of short chain fatty acid (SCFA)-fermenting bacteria in early life (humans and mice) is associated with enhanced type 2 immune responses in peripheral tissues including the lung. OBJECTIVE Our goal was to reveal the microbiome-dependent cellular and molecular mechanisms driving enhanced susceptibility to type 2 allergic lung disease. METHODS We used low-dose vancomycin to selectively deplete SCFA-fermenting bacteria in wild-type mice. We then examined the frequency and activation status of innate and adaptive immune cell lineages with and without SCFA supplementation. Finally, we used ILC2-deficient and signal transducer and activator of transcription 6 (STAT6)-deficient transgenic mouse strains to delineate the cellular and cytokine pathways leading to enhanced allergic disease susceptibility. RESULTS Mice with vancomycin-induced dysbiosis exhibited a 2-fold increase in lung ILC2 primed to produce elevated levels of IL-2, -5, and -13. In addition, upon IL-33 inhalation, mouse lung ILC2 displayed a novel ability to produce high levels of IL-4. These expanded and primed ILC2s drove B1 cell expansion and IL-4-dependent production of IgE that in turn led to exacerbated allergic inflammation. Importantly, these enhanced lung inflammatory phenotypes in mice with vancomycin-induced dysbiosis were reversed by administration of dietary SCFA (specifically butyrate). CONCLUSION SCFAs regulate an ILC2-B1 cell-IgE axis. Early-life administration of vancomycin, an antibiotic known to deplete SCFA-fermenting gut bacteria, primes and amplifies this axis and leads to lifelong enhanced susceptibility to type 2 allergic lung disease.
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalia Nayyar
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yicong Li
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sameeksha Chopra
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael R Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada; Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
17
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [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: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
18
|
Liu J, Wang M, Tian X, Wu S, Peng H, Zhu Y, Liu Y. New insights into allergic rhinitis treatment: MSC nanovesicles targeting dendritic cells. J Nanobiotechnology 2024; 22:575. [PMID: 39294599 PMCID: PMC11411834 DOI: 10.1186/s12951-024-02748-2] [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: 01/30/2024] [Accepted: 08/02/2024] [Indexed: 09/20/2024] Open
Abstract
Allergic rhinitis (AR) is a condition with limited treatment options. This study investigates the potential use of mesenchymal stem cell (MSC) nanovesicles as a novel therapy for AR. Specifically, the study explores the underlying mechanisms of MSC nanovesicle therapy by targeting dendritic cells (DCs). The researchers fabricated DC-targeted P-D2-EVs nanovesicles and characterized their properties. Transcriptomic sequencing and single-cell sequencing analyses were performed to study the impact of P-D2-EVs on AR mice, identifying core genes involved in the treatment. In vitro cell experiments were conducted to validate the effects of P-D2-EVs on DC metabolism, Th2 differentiation, and ILC2 activation. The results showed that P-D2-EVs efficiently targeted DCs. Transcriptomic sequencing analysis revealed differential expression of 948 genes in nasal tissue DCs of mice treated with P-D2-EVs. Single-cell sequencing further revealed that P-D2-EVs had inhibitory effects on DC activation, Th2 differentiation, and ILC2 activation, with Fut1 identified as the core gene. Validation experiments demonstrated that P-D2-EVs improved IL10 metabolism in DCs by downregulating Fut1 expression, thereby suppressing Th2 differentiation and ILC2 activation. Animal experiments confirmed the inhibitory effects of P-D2-EVs and their ability to ameliorate AR symptoms in mice. The study suggests that P-D2-EVs reshape DC metabolism and suppress Th2 differentiation and ILC2 activation through the inhibition of the Fut1/ICAM1/P38 MAPK signaling pathway, providing a potential therapeutic approach for AR.
Collapse
Affiliation(s)
- Jianyu Liu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Meiqun Wang
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Xiaoyan Tian
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Shuhong Wu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Haisen Peng
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Yaqiong Zhu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Yuehui Liu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
19
|
Gu S, Wang R, Zhang W, Wen C, Chen C, Liu S, Lei Q, Zhang P, Zeng S. The production, function, and clinical applications of IL-33 in type 2 inflammation-related respiratory diseases. Front Immunol 2024; 15:1436437. [PMID: 39301028 PMCID: PMC11410612 DOI: 10.3389/fimmu.2024.1436437] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Epithelial-derived IL-33 (Interleukin-33), as a member of alarm signals, is a chemical substance produced under harmful stimuli that can promote innate immunity and activate adaptive immune responses. Type 2 inflammation refers to inflammation primarily mediated by Type 2 helper T cells (Th2), Type 2 innate lymphoid cells (ILC2), and related cytokines. Type 2 inflammation manifests in various forms in the lungs, with diseases such as asthma and chronic obstructive pulmonary disease chronic obstructive pulmonary disease (COPD) closely associated with Type 2 inflammation. Recent research suggests that IL-33 has a promoting effect on Type 2 inflammation in the lungs and can be regarded as an alarm signal for Type 2 inflammation. This article provides an overview of the mechanisms and related targets of IL-33 in the development of lung diseases caused by Type 2 inflammation, and summarizes the associated treatment methods. Analyzing lung diseases from a new perspective through the alarm of Type 2 inflammation helps to gain a deeper understanding of the pathogenesis of these related lung diseases. This, in turn, facilitates a better understanding of the latest treatment methods and potential therapeutic targets for diseases, with the expectation that targeting lL-33 can propose new strategies for disease prevention.
Collapse
Affiliation(s)
- Shiyao Gu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruixuan Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wantian Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Cen Wen
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
20
|
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
|
21
|
Qin M, Fang Y, Zheng Q, Peng M, Wang L, Sang X, Cao G. Tissue microenvironment induces tissue specificity of ILC2. Cell Death Discov 2024; 10:324. [PMID: 39013890 PMCID: PMC11252336 DOI: 10.1038/s41420-024-02096-y] [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: 03/24/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Type 2 innate lymphoid cells were found to be members of the innate immune cell family, which is involved in innate and adaptive immunity to resist the invasion of foreign antigens and induce allergic reactions caused by allergens. The advancement of ILC2 research has pointed out that ILC2s have a high degree of diversity, challenging the notion of their homogeneity as a cellular population. An increasing number of studies indicate that ILC2 is a cell population with tissue specificity which can be induced by the tissue microenvironment. In addition, crosstalk between tissues can change ILC2 functions of migration and activation. Here, we emphasize that ILC2 undergoes adaptive changes under the regulation of the tissue microenvironment and distant tissues, thereby coordinating the organization's operation. In addition, ILC2 alterations induced by the tissue microenvironment are not limited to the ILC2 cell population, and ILC2 can also transdifferentiate into another class of ILC cell population (ILC1 or ILC3). In this review, we summarized the tissue-specific effects of ILC2 by tissue microenvironment and focused on the function of ILC2 in inter-tissue crosstalk. Lastly, we discussed the transdifferentiations of ILC2 caused by the abnormal change in tissue environment.
Collapse
Affiliation(s)
- Minjing Qin
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuanyuan Fang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qitong Zheng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia'nan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
22
|
Kumagai T, Iwata A, Furuya H, Kato K, Okabe A, Toda Y, Kanai M, Fujimura L, Sakamoto A, Kageyama T, Tanaka S, Suto A, Hatano M, Kaneda A, Nakajima H. A distal enhancer of GATA3 regulates Th2 differentiation and allergic inflammation. Proc Natl Acad Sci U S A 2024; 121:e2320727121. [PMID: 38923989 PMCID: PMC11228505 DOI: 10.1073/pnas.2320727121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Asthma is a widespread airway disorder where GATA3-dependent Type-2 helper T (Th2) cells and group 2 innate lymphoid cells (ILC2s) play vital roles. Asthma-associated single nucleotide polymorphisms (SNPs) are enriched in a region located 926-970 kb downstream from GATA3 in the 10p14 (hG900). However, it is unknown how hG900 affects the pathogenesis of allergic airway inflammation. To investigate the roles of the asthma-associated GATA3 enhancer region in experimental allergic airway inflammation, we first examined the correlation between GATA3 expression and the activation of the hG900 region was analyzed by flow cytometry and ChIP-qPCR. We found that The activation of enhancers in the hG900 region was strongly correlated to the levels of GATA3 in human peripheral T cell subsets. We next generated mice lacking the mG900 region (mG900KO mice) were generated by the CRISPR-Cas9 system, and the development and function of helper T cells and ILCs in mG900KO mice were analyzed in steady-state conditions and allergic airway inflammation induced by papain or house dust mite (HDM). The deletion of the mG900 did not affect the development of lymphocytes in steady-state conditions or allergic airway inflammation induced by papain. However, mG900KO mice exhibited reduced allergic inflammation and Th2 differentiation in the HDM-induced allergic airway inflammation. The analysis of the chromatin conformation around Gata3 by circular chromosome conformation capture coupled to high-throughput sequencing (4C-seq) revealed that the mG900 region interacted with the transcription start site of Gata3 with an influencing chromatin conformation in Th2 cells. These findings indicate that the mG900 region plays a pivotal role in Th2 differentiation and thus enhances allergic airway inflammation.
Collapse
Affiliation(s)
- Takashi Kumagai
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Hiroki Furuya
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Kodai Kato
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Atsushi Okabe
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Health and Disease Omics Center, Chiba University, Chiba260-8670, Japan
| | - Yosuke Toda
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Mizuki Kanai
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Lisa Fujimura
- Biomedical Research Center, Chiba University, Chiba260-8670, Japan
| | - Akemi Sakamoto
- Biomedical Research Center, Chiba University, Chiba260-8670, Japan
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Masahiko Hatano
- Biomedical Research Center, Chiba University, Chiba260-8670, Japan
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Health and Disease Omics Center, Chiba University, Chiba260-8670, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba260-8670, Japan
| |
Collapse
|
23
|
Szeto AC, Clark PA, Ferreira AC, Heycock M, Griffiths EL, Jou E, Mannion J, Luan SL, Storrar S, Knolle MD, Kozik P, Jolin HE, Fallon PG, McKenzie AN. Mef2d potentiates type-2 immune responses and allergic lung inflammation. Science 2024; 384:eadl0370. [PMID: 38935708 PMCID: PMC7616247 DOI: 10.1126/science.adl0370] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/02/2024] [Indexed: 06/29/2024]
Abstract
Innate lymphoid cells (ILCs) and adaptive T lymphocytes promote tissue homeostasis and protective immune responses. Their production depends on the transcription factor GATA3, which is further elevated specifically in ILC2s and T helper 2 cells to drive type-2 immunity during tissue repair, allergic disorders, and anti-helminth immunity. The control of this crucial up-regulation is poorly understood. Using CRISPR screens in ILCs we identified previously unappreciated myocyte-specific enhancer factor 2d (Mef2d)-mediated regulation of GATA3-dependent type-2 lymphocyte differentiation. Mef2d-deletion from ILC2s and/or T cells specifically protected against an allergen lung challenge. Mef2d repressed Regnase-1 endonuclease expression to enhance IL-33 receptor production and IL-33 signaling and acted downstream of calcium-mediated signaling to translocate NFAT1 to the nucleus to promote type-2 cytokine-mediated immunity.
Collapse
Affiliation(s)
- Aydan C.H. Szeto
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Paula A. Clark
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Ana C.F. Ferreira
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Morgan Heycock
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Emma L. Griffiths
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Eric Jou
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Jonathan Mannion
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
- Cambridge
University Hospitals,
Cambridge, CB2 0QQ, United Kingdom
| | - Shi-Lu Luan
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Sophie Storrar
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Martin D. Knolle
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
- Cambridge
University Hospitals,
Cambridge, CB2 0QQ, United Kingdom
| | - Patrycja Kozik
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | - Helen E. Jolin
- MRC Laboratory
of Molecular Biology,
Cambridge, CB2 0QH, United Kingdom
| | | | | |
Collapse
|
24
|
Son A, Baral I, Falduto GH, Schwartz DM. Locus of (IL-9) control: IL9 epigenetic regulation in cellular function and human disease. Exp Mol Med 2024; 56:1331-1339. [PMID: 38825637 PMCID: PMC11263352 DOI: 10.1038/s12276-024-01241-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 06/04/2024] Open
Abstract
Interleukin-9 (IL-9) is a multifunctional cytokine with roles in a broad cross-section of human diseases. Like many cytokines, IL-9 is transcriptionally regulated by a group of noncoding regulatory elements (REs) surrounding the IL9 gene. These REs modulate IL-9 transcription by forming 3D loops that recruit transcriptional machinery. IL-9-promoting transcription factors (TFs) can bind REs to increase locus accessibility and permit chromatin looping, or they can be recruited to already accessible chromatin to promote transcription. Ample mechanistic and genome-wide association studies implicate this interplay between IL-9-modulating TFs and IL9 cis-REs in human physiology, homeostasis, and disease.
Collapse
Affiliation(s)
- Aran Son
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Ishita Baral
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guido H Falduto
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniella M Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Jou E. Clinical and basic science aspects of innate lymphoid cells as novel immunotherapeutic targets in cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:1-60. [PMID: 39461748 DOI: 10.1016/bs.pmbts.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade, demonstrating remarkable efficacy across a broad range of cancer types. However, not all patients or cancer types respond to contemporary clinically-utilised immunotherapeutic strategies, which largely focus on harnessing adaptive immune T cells for cancer treatment. Accordingly, it is increasingly recognised that upstream innate immune pathways, which govern and orchestrate the downstream adaptive immune response, may prove critical in overcoming cancer immunotherapeutic resistance. Innate lymphoid cells (ILCs) are the most recently discovered major innate immune cell population. They have overarching roles in homeostasis and orchestrating protective immunity against pathogens. As innate immune counterparts of adaptive immune T cells, ILCs exert effector functions through the secretion of cytokines and direct cell-to-cell contact, with broad influence on the overall immune response. Importantly, dysregulation of ILC subsets have been associated with a range of diseases, including immunodeficiency disorders, allergy, autoimmunity, and more recently, cancer. ILCs may either promote or inhibit cancer initiation and progression depending on the cancer type and the specific ILC subsets involved. Critically, therapeutic targeting of ILCs and their associated cytokines shows promise against a wide range of cancer types in both preclinical models and early phase oncology clinical trials. This chapter provides a comprehensive overview of the current understanding of ILC subsets and the associated cytokines they produce in cancer pathogenesis, with specific focus on how these innate pathways are, or can be targeted, therapeutically to overcome therapeutic resistance and ultimately improve patient care.
Collapse
Affiliation(s)
- Eric Jou
- Department of Oncology, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom; Kellogg College, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
26
|
Zaiss DMW, Pearce EJ, Artis D, McKenzie ANJ, Klose CSN. Cooperation of ILC2s and T H2 cells in the expulsion of intestinal helminth parasites. Nat Rev Immunol 2024; 24:294-302. [PMID: 37798539 DOI: 10.1038/s41577-023-00942-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
Type 2 immune responses form a critical defence against enteric worm infections. In recent years, mouse models have revealed shared and unique functions for group 2 innate lymphoid cells and T helper 2 cells in type 2 immune response to intestinal helminths. Both cell types use similar innate effector functions at the site of infection, whereas each population has distinct roles during different stages of infection. In this Perspective, we review the underlying mechanisms used by group 2 innate lymphoid cells and T helper 2 cells to cooperate with each other and suggest an overarching model of the interplay between these cell types over the course of a helminth infection.
Collapse
Affiliation(s)
- Dietmar M W Zaiss
- Department of Immune Medicine, University Regensburg, Regensburg, Germany.
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany.
| | - Edward J Pearce
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
27
|
Englebert K, Taquin A, Azouz A, Acolty V, Vande Velde S, Vanhollebeke M, Innes H, Boon L, Keler T, Leo O, Goriely S, Moser M, Oldenhove G. The CD27/CD70 pathway negatively regulates visceral adipose tissue-resident Th2 cells and controls metabolic homeostasis. Cell Rep 2024; 43:113824. [PMID: 38386557 DOI: 10.1016/j.celrep.2024.113824] [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: 05/09/2023] [Revised: 12/11/2023] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
Adipose tissue homeostasis relies on the interplay between several regulatory lineages, such as type 2 innate lymphoid cells (ILC2s), T helper 2 (Th2) cells, regulatory T cells, eosinophils, and type 2 macrophages. Among them, ILC2s are numerically the dominant source of type 2 cytokines and are considered as major regulators of adiposity. Despite the overlap in immune effector molecules and sensitivity to alarmins (thymic stromal lymphopoietin and interleukin-33) between ILC2s and resident memory Th2 lymphocytes, the role of the adaptive axis of type 2 immunity remains unclear. We show that mice deficient in CD27, a member of the tumor necrosis factor receptor superfamily, are more resistant to obesity and associated disorders. A comparative analysis of the CD4 compartment of both strains revealed higher numbers of fat-resident memory Th2 cells in the adipose tissue of CD27 knockout mice, which correlated with decreased programmed cell death protein 1-induced apoptosis. Our data point to a non-redundant role for Th2 lymphocytes in obesogenic conditions.
Collapse
Affiliation(s)
- Kevin Englebert
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Anaelle Taquin
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Abdulkader Azouz
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Institute for Medical Immunology (IMI), ULB, Gosselies, Belgium
| | - Valérie Acolty
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Sylvie Vande Velde
- Interuniversity Institute of Bioinformatics in Brussels (ULB-VUB), Brussels, Belgium; Machine Learning Group, ULB, Brussels, Belgium
| | - Marie Vanhollebeke
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Hadrien Innes
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | | | | | - Oberdan Leo
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Stanislas Goriely
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium; Institute for Medical Immunology (IMI), ULB, Gosselies, Belgium
| | - Muriel Moser
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium
| | - Guillaume Oldenhove
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium; Immunobiology Lab, ULB, Gosselies, Belgium.
| |
Collapse
|
28
|
Dai Z, Gong Z, Wang C, Long W, Liu D, Zhang H, Lei A. The role of hormones in ILC2-driven allergic airway inflammation. Scand J Immunol 2024; 99:e13357. [PMID: 39008023 DOI: 10.1111/sji.13357] [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: 08/22/2023] [Revised: 11/23/2023] [Accepted: 01/05/2024] [Indexed: 07/16/2024]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a type of innate immune cells that produce a large amount of IL-5 and IL-13 and two cytokines that are crucial for various processes such as allergic airway inflammation, tissue repair and tissue homeostasis. It is known that damaged epithelial-derived alarmins, such as IL-33, IL-25 and thymic stromal lymphopoietin (TSLP), are the predominant ILC2 activators that mediate the production of type 2 cytokines. In recent years, abundant studies have found that many factors can regulate ILC2 development and function. Hormones synthesized by the body's endocrine glands or cells play an important role in immune response. Notably, ILC2s express hormone receptors and their proliferation and function can be modulated by multiple hormones during allergic airway inflammation. Here, we summarize the effects of multiple hormones on ILC2-driven allergic airway inflammation and discuss the underlying mechanisms and potential therapeutic significance.
Collapse
Affiliation(s)
- Zhongling Dai
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhande Gong
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - WeiXiang Long
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Duo Liu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Haijun Zhang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
29
|
Cha J, Kim TG, Bhae E, Gwak HJ, Ju Y, Choe YH, Jang IH, Jung Y, Moon S, Kim T, Lee W, Park JS, Chung YW, Yang S, Kang YK, Hyun YM, Hwang GS, Lee WJ, Rho M, Ryu JH. Skin microbe-dependent TSLP-ILC2 priming axis in early life is co-opted in allergic inflammation. Cell Host Microbe 2024; 32:244-260.e11. [PMID: 38198924 DOI: 10.1016/j.chom.2023.12.006] [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: 11/13/2022] [Revised: 09/17/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Although early life colonization of commensal microbes contributes to long-lasting immune imprinting in host tissues, little is known regarding the pathophysiological consequences of postnatal microbial tuning of cutaneous immunity. Here, we show that postnatal exposure to specific skin commensal Staphylococcus lentus (S. lentus) promotes the extent of atopic dermatitis (AD)-like inflammation in adults through priming of group 2 innate lymphoid cells (ILC2s). Early postnatal skin is dynamically populated by discrete subset of primed ILC2s driven by microbiota-dependent induction of thymic stromal lymphopoietin (TSLP) in keratinocytes. Specifically, the indole-3-aldehyde-producing tryptophan metabolic pathway, shared across Staphylococcus species, is involved in TSLP-mediated ILC2 priming. Furthermore, we demonstrate a critical contribution of the early postnatal S. lentus-TSLP-ILC2 priming axis in facilitating AD-like inflammation that is not replicated by later microbial exposure. Thus, our findings highlight the fundamental role of time-dependent neonatal microbial-skin crosstalk in shaping the threshold of innate type 2 immunity co-opted in adulthood.
Collapse
Affiliation(s)
- Jimin Cha
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Euihyun Bhae
- Department of Artificial Intelligence, Hanyang University, Seoul 04763, Korea
| | - Ho-Jin Gwak
- Department of Computer Science, Hanyang University, Seoul 04763, Korea
| | - Yeajin Ju
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Korea
| | - Young Ho Choe
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - In-Hwan Jang
- National Creative Research Initiative Center for Hologenomics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Korea
| | - Sungmin Moon
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Taehyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Wuseong Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jung Sun Park
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Youn Wook Chung
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Korea
| | - Yong-Kook Kang
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Young-Min Hyun
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Won-Jae Lee
- National Creative Research Initiative Center for Hologenomics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Mina Rho
- Department of Computer Science, Hanyang University, Seoul 04763, Korea; Department of Biomedical Informatics, Hanyang University, Seoul 04763, Korea
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea.
| |
Collapse
|
30
|
Zhu J. A pathway linking atopic dermatitis to skin microbes. Cell Host Microbe 2024; 32:154-155. [PMID: 38359797 DOI: 10.1016/j.chom.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/17/2024]
Abstract
Interactions between microbiota and host skin have an important impact on cutaneous immunity and inflammation. In this issue of Cell Host & Microbe, Cha et al. report that skin commensal bacteria-mediated priming of group 2 innate lymphoid cells in early life predisposes the mice to atopic dermatitis-like inflammation in adulthood.
Collapse
Affiliation(s)
- Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
31
|
Kral M, van der Vorst EP, Surnov A, Weber C, Döring Y. ILC2-mediated immune crosstalk in chronic (vascular) inflammation. Front Immunol 2023; 14:1326440. [PMID: 38179045 PMCID: PMC10765502 DOI: 10.3389/fimmu.2023.1326440] [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: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Abstract
Crosstalk between innate and adaptive immunity is pivotal for an efficient immune response and to maintain immune homeostasis under steady state conditions. As part of the innate immune system, type 2 innate lymphoid cells (ILC2s) have emerged as new important regulators of tissue homeostasis and repair by fine-tuning innate-adaptive immune cell crosstalk. ILC2s mediate either pro- or anti-inflammatory immune responses in a context dependent manner. Inflammation has proven to be a key driver of atherosclerosis, resembling the key underlying pathophysiology of cardiovascular disease (CVD). Notably, numerous studies point towards an atheroprotective role of ILC2s e.g., by mediating secretion of type-II cytokines (IL-5, IL-13, IL-9). Boosting these protective responses may be suitable for promising future therapy, although these protective cues are currently incompletely understood. Additionally, little is known about the mechanisms by which chemokine/chemokine receptor signaling shapes ILC2 functions in vascular inflammation and atherosclerosis. Hence, this review will focus on the latest findings regarding the protective and chemokine/chemokine receptor guided interplay between ILC2s and other immune cells like T and B cells, dendritic cells and macrophages in atherosclerosis. Further, we will elaborate on potential therapeutic implications which result or could be distilled from the dialogue of ILC2s with cells of the immune system in cardiovascular diseases.
Collapse
Affiliation(s)
- Maria Kral
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Emiel P.C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Munich, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Alexey Surnov
- Type 1 Diabetes Immunology (TDI), Helmholtz Diabetes Center (HDC), Helmholtz Center Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR) Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
32
|
Ding Z, Mulder J, Robinson MJ. The origins and longevity of IgE responses as indicated by serological and cellular studies in mice and humans. Allergy 2023; 78:3103-3117. [PMID: 37417548 PMCID: PMC10952832 DOI: 10.1111/all.15799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
The existence of long-lived IgE antibody-secreting cells (ASC) is contentious, with the maintenance of sensitization by the continuous differentiation of short-lived IgE+ ASC a possibility. Here, we review the epidemiological profile of IgE production, and give an overview of recent discoveries made on the mechanisms regulating IgE production from mouse models. Together, these data suggest that for most individuals, in most IgE-associated diseases, IgE+ ASC are largely short-lived cells. A subpopulation of IgE+ ASC in humans is likely to survive for tens of months, although due to autonomous IgE B cell receptor (BCR) signaling and antigen-driven IgE+ ASC apoptosis, in general IgE+ ASC probably do not persist for the decades that other ASC are inferred to do. We also report on recently identified memory B cell transcriptional subtypes that are the likely source of IgE in ongoing responses, highlighting the probable importance of IL-4Rα in their regulation. We suggest the field should look at dupilumab and other drugs that prohibit IgE+ ASC production as being effective treatments for IgE-mediated aspects of disease in most individuals.
Collapse
Affiliation(s)
- Zhoujie Ding
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | - Jesse Mulder
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | | |
Collapse
|
33
|
Chopp LB, Zhu X, Gao Y, Nie J, Singh J, Kumar P, Young KZ, Patel S, Li C, Balmaceno-Criss M, Vacchio MS, Wang MM, Livak F, Merchant JL, Wang L, Kelly MC, Zhu J, Bosselut R. Zfp281 and Zfp148 control CD4 + T cell thymic development and T H2 functions. Sci Immunol 2023; 8:eadi9066. [PMID: 37948511 DOI: 10.1126/sciimmunol.adi9066] [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: 05/24/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
How CD4+ lineage gene expression is initiated in differentiating thymocytes remains poorly understood. Here, we show that the paralog transcription factors Zfp281 and Zfp148 control both this process and cytokine expression by T helper cell type 2 (TH2) effector cells. Genetic, single-cell, and spatial transcriptomic analyses showed that these factors promote the intrathymic CD4+ T cell differentiation of class II major histocompatibility complex (MHC II)-restricted thymocytes, including expression of the CD4+ lineage-committing factor Thpok. In peripheral T cells, Zfp281 and Zfp148 promoted chromatin opening at and expression of TH2 cytokine genes but not of the TH2 lineage-determining transcription factor Gata3. We found that Zfp281 interacts with Gata3 and is recruited to Gata3 genomic binding sites at loci encoding Thpok and TH2 cytokines. Thus, Zfp148 and Zfp281 collaborate with Gata3 to promote CD4+ T cell development and TH2 cell responses.
Collapse
Affiliation(s)
- Laura B Chopp
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Immunology Graduate Group, University of Pennsylvania Medical School, Philadelphia, PA 19104, USA
| | - Xiaoliang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yayi Gao
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jia Nie
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jatinder Singh
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Parimal Kumar
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly Z Young
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shil Patel
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- University of Maryland Medical School, Baltimore, MD 21201, USA
| | - Caiyi Li
- Flow Cytometry Core, Laboratory of Genomic Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariah Balmaceno-Criss
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Melanie S Vacchio
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael M Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Ferenc Livak
- Flow Cytometry Core, Laboratory of Genomic Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juanita L Merchant
- Department of Gastroenterology and Hepatology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Lie Wang
- Institute of Immunology, and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Michael C Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
Shilovskiy IP, Kovchina VI, Timotievich ED, Nikolskii AA, Khaitov MR. Role and Molecular Mechanisms of Alternative Splicing of Th2-Cytokines IL-4 and IL-5 in Atopic Bronchial Asthma. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1608-1621. [PMID: 38105028 DOI: 10.1134/s0006297923100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 12/19/2023]
Abstract
Bronchial asthma (BA) is a heterogeneous chronic inflammatory disease of the respiratory tract. Allergic (atopic) asthma is the most common (up to 80% of cases) phenotype developing through the Th2-dependent mechanisms involving cytokines: IL-4, IL-5, IL-9, and IL-13. The genes encoding Th2-cytokines have a mosaic structure (encode exons and introns). Therefore, several mature mRNA transcripts and protein isoforms can be derived from a single mRNA precursor through alternative splicing, and they may contribute to BA pathogenesis. Analysis of the published studies and databases revealed existence of the alternative mRNA transcripts for IL-4, IL-5, and IL-13. The alternative transcripts of IL-4 and IL-5 carry open reading frames and therefore can encode functional proteins. It was shown that not only alternative mRNA transcripts exist for IL-4, but alternative protein isoforms, as well. Natural protein isoform (IL-4δ2) lacking the part encoded by exon-2 was identified. Similarly, alternative mRNA transcript with deleted exon-2 (IL-5δ2) was also identified for IL-5. In this review, we summarize current knowledge about the identified alternative mRNA transcripts and protein isoforms of Th2-cytokinins, first of all IL-4 and IL-5. We have analyzed biological properties of the alternative variants of these cytokines, their possible role in the allergic asthma pathogenesis, and considered their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Igor P Shilovskiy
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522, Russia.
| | - Valeriya I Kovchina
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522, Russia
| | - Ekaterina D Timotievich
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522, Russia
| | - Alexander A Nikolskii
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522, Russia
| | - Musa R Khaitov
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117997, Russia
| |
Collapse
|
35
|
Affiliation(s)
- Irina Tsymala
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| |
Collapse
|
36
|
Meloun A, León B. Sensing of protease activity as a triggering mechanism of Th2 cell immunity and allergic disease. FRONTIERS IN ALLERGY 2023; 4:1265049. [PMID: 37810200 PMCID: PMC10552645 DOI: 10.3389/falgy.2023.1265049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
CD4 T-helper cell type 2 (Th2) cells mediate host defense against extracellular parasites, like helminths. However, Th2 cells also play a pivotal role in the onset and progression of allergic inflammatory diseases such as atopic dermatitis, allergic rhinitis, asthma, and food allergy. This happens when allergens, which are otherwise harmless foreign proteins, are mistakenly identified as "pathogenic." Consequently, the encounter with these allergens triggers the activation of specific Th2 cell responses, leading to the development of allergic reactions. Understanding the molecular basis of allergen sensing is vital for comprehending how Th2 cell responses are erroneously initiated in individuals with allergies. The presence of protease activity in allergens, such as house dust mites (HDM), pollen, fungi, or cockroaches, has been found to play a significant role in triggering robust Th2 cell responses. In this review, we aim to examine the significance of protease activity sensing in foreign proteins for the initiation of Th2 cell responses, highlighting how evolving a host protease sensor may contribute to detect invading helminth parasites, but conversely can also trigger unwanted reactions to protease allergens. In this context, we will explore the recognition receptors activated by proteolytic enzymes present in major allergens and their contribution to Th2-mediated allergic responses. Furthermore, we will discuss the coordinated efforts of sensory neurons and epithelial cells in detecting protease allergens, the subsequent activation of intermediary cells, including mast cells and type 2 innate lymphoid cells (ILC2s), and the ultimate integration of all signals by conventional dendritic cells (cDCs), leading to the induction of Th2 cell responses. On the other hand, the review highlights the role of monocytes in the context of protease allergen exposure and their interaction with cDCs to mitigate undesirable Th2 cell reactions. This review aims to provide insights into the innate functions and cell communications triggered by protease allergens, which can contribute to the initiation of detrimental Th2 cell responses, but also promote mechanisms to effectively suppress their development.
Collapse
Affiliation(s)
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
37
|
Jan-Abu SC, Kabil A, McNagny KM. Parallel origins and functions of T cells and ILCs. Clin Exp Immunol 2023; 213:76-86. [PMID: 37235977 PMCID: PMC10324547 DOI: 10.1093/cei/uxad056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 05/28/2023] Open
Abstract
Innate lymphoid cells (ILCs) are tissue resident cells that are triggered through a relatively broad spectrum of alarmins, inflammatory cues, neuropeptides, and hormones. Functionally, ILCs are akin to subsets of helper T cells and are characterized by a similar effector cytokine profile. They also share a dependency on many of the same essential transcription factors identified for the maintenance and survival of T cells. The key distinguishing factor between the ILC family and T cells is the lack of antigen-specific T cell receptor (TCR) on ILCs and, thus, they can be considered the "ultimate invariant T cells". ILCs, like T cells, orchestrate downstream effector inflammatory responses by adjusting the cytokine microenvironment in a fashion that promotes protection, health, and homeostasis at mucosal barrier sites. But also, like T cells, ILCs have recently been implicated in several pathological inflammatory disease states. This review focuses on the selective role of ILCs in the development of allergic airway inflammation (AAI) and fibrosis in the gut where a complex ILC interplay has been shown to either attenuate or worsen disease. Finally, we discuss new data on TCR gene rearrangements in subsets of ILCs that challenge the current dogma linking their origin to committed bone marrow progenitors and instead propose a thymic origin for at least some ILCs. In addition, we highlight how naturally occurring TCR rearrangements and the expression of major histocompatibility (MHC) molecules in ILCs provide a useful natural barcode for these cells and may prove instrumental in studying their origins and plasticity.
Collapse
Affiliation(s)
- Sia C Jan-Abu
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart and Lung Innovation (HLI), St Paul’s Hospital, Vancouver, BC, Canada
| |
Collapse
|
38
|
Butcher MJ, Gurram RK, Zhu X, Chen X, Hu G, Lazarevic V, Zhao K, Zhu J. GATA3 induces the pathogenicity of Th17 cells via regulating GM-CSF expression. Front Immunol 2023; 14:1186580. [PMID: 37449212 PMCID: PMC10337884 DOI: 10.3389/fimmu.2023.1186580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
T-bet-expressing Th17 (T-bet+RORγt+) cells are associated with the induction of pathology during experimental autoimmune encephalomyelitis (EAE) and the encephalitic nature of these Th17 cells can be explained by their ability to produce GM-CSF. However, the upstream regulatory mechanisms that control Csf2 (gene encoding GM-CSF) expression are still unclear. In this study, we found that Th17 cells dynamically expressed GATA3, the master transcription factor for Th2 cell differentiation, during their differentiation both in vitro and in vivo. Early deletion of Gata3 in three complimentary conditional knockout models by Cre-ERT2, hCd2 Cre and Tbx21 Cre, respectively, limited the pathogenicity of Th17 cells during EAE, which was correlated with a defect in generating pathogenic T-bet-expressing Th17 cells. These results indicate that early GATA3-dependent gene regulation is critically required to generate a de novo encephalitogenic Th17 response. Furthermore, a late deletion of Gata3 via Cre-ERT2 in the adoptive transfer EAE model resulted in a cell intrinsic failure to induce EAE symptoms which was correlated with a substantial reduction in GM-CSF production without affecting the generation and/or maintenance of T-bet-expressing Th17 cells. RNA-Seq analysis of Gata3-sufficient and Gata3-deficient CNS-infiltrating CD4+ effector T cells from mixed congenic co-transfer recipient mice revealed an important, cell-intrinsic, function of GATA3 in regulating the expression of Egr2, Bhlhe40, and Csf2. Thus, our data highlights a novel role for GATA3 in promoting and maintaining the pathogenicity of T-bet-expressing Th17 cells in EAE, via putative regulation of Egr2, Bhlhe40, and GM-CSF expression.
Collapse
Affiliation(s)
- Matthew J Butcher
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rama Krishna Gurram
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Xiaoliang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Xi Chen
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Gangqing Hu
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Vanja Lazarevic
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
Son A, Meylan F, Gomez-Rodriguez J, Kaul Z, Sylvester M, Falduto GH, Vazquez E, Haque T, Kitakule MM, Wang C, Manthiram K, Qi CF, Cheng J, Gurram RK, Zhu J, Schwartzberg P, Milner JD, Frischmeyer-Guerrerio PA, Schwartz DM. Dynamic chromatin accessibility licenses STAT5- and STAT6-dependent innate-like function of T H9 cells to promote allergic inflammation. Nat Immunol 2023; 24:1036-1048. [PMID: 37106040 PMCID: PMC10247433 DOI: 10.1038/s41590-023-01501-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023]
Abstract
Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.
Collapse
Affiliation(s)
- Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francoise Meylan
- Office of Science and Technology, National Institute of Arthritis, Musculoskeletal, and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julio Gomez-Rodriguez
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- TCR Therapeutics, Cambridge, MA, USA
| | - Zenia Kaul
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Guido H Falduto
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Estefania Vazquez
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tamara Haque
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Moses M Kitakule
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Pediatric Allergy Immunology and Rheumatology, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Chujun Wang
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kalpana Manthiram
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen-Feng Qi
- Pathology Core, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jun Cheng
- Embryonic Stem Cell and Transgenic Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rama K Gurram
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinfang Zhu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Schwartzberg
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joshua D Milner
- Division of Pediatric Allergy Immunology and Rheumatology, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Pamela A Frischmeyer-Guerrerio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
40
|
Cui K, Chen Z, Cao Y, Liu S, Ren G, Hu G, Fang D, Wei D, Liu C, Zhu J, Wu C, Zhao K. Restraint of IFN-γ expression through a distal silencer CNS-28 for tissue homeostasis. Immunity 2023; 56:944-958.e6. [PMID: 37040761 PMCID: PMC10175192 DOI: 10.1016/j.immuni.2023.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 01/13/2023] [Accepted: 03/13/2023] [Indexed: 04/13/2023]
Abstract
Interferon-γ (IFN-γ) is a key cytokine in response to viral or intracellular bacterial infection in mammals. While a number of enhancers are described to promote IFN-γ responses, to the best of our knowledge, no silencers for the Ifng gene have been identified. By examining H3K4me1 histone modification in naive CD4+ T cells within Ifng locus, we identified a silencer (CNS-28) that restrains Ifng expression. Mechanistically, CNS-28 maintains Ifng silence by diminishing enhancer-promoter interactions within Ifng locus in a GATA3-dependent but T-bet-independent manner. Functionally, CNS-28 restrains Ifng transcription in NK cells, CD4+ cells, and CD8+ T cells during both innate and adaptive immune responses. Moreover, CNS-28 deficiency resulted in repressed type 2 responses due to elevated IFN-γ expression, shifting Th1 and Th2 paradigm. Thus, CNS-28 activity ensures immune cell quiescence by cooperating with other regulatory cis elements within the Ifng gene locus to minimize autoimmunity.
Collapse
Affiliation(s)
- Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yaqiang Cao
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Shuai Liu
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Gang Ren
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Gangqing Hu
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Difeng Fang
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danping Wei
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chengyu Liu
- Transgenic Core Facility, DIR, NHLBI, NIH, Bethesda, MD, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA.
| |
Collapse
|
41
|
Topczewska PM, Rompe ZA, Jakob MO, Stamm A, Leclère PS, Preußer A, Duerr CU, Thole LML, Kotsch K, Artis D, Klose CSN. ILC2 require cell-intrinsic ST2 signals to promote type 2 immune responses. Front Immunol 2023; 14:1130933. [PMID: 37063913 PMCID: PMC10104602 DOI: 10.3389/fimmu.2023.1130933] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/08/2023] [Indexed: 04/03/2023] Open
Abstract
The initiation of type 2 immune responses at mucosal barriers is regulated by rapidly secreted cytokines called alarmins. The alarmins IL-33, IL-25 and TSLP are mainly secreted by stromal and epithelial cells in tissues and were linked to chronic inflammatory diseases, such as allergic lung inflammation, or to resistance against worm infections. Receptors for alarmins are expressed by a variety of immune cells, including group 2 innate lymphoid cells (ILC2s), an early source of the type 2 cytokines, such as IL-5 and IL-13, which have been linked to atopic diseases and anti-worm immunity as well. However, the precise contribution of the IL-33 receptor signals for ILC2 activation still needs to be completed due to limitations in targeting genes in ILC2. Using the newly established Nmur1 iCre-eGFP mouse model, we obtained specific conditional genetic ablation of the IL-33 receptor subunit ST2 in ILC2s. ST2-deficient ILC2s were unresponsive to IL-33 but not to stimulation with the alarmin IL-25. As a result of defective ST2 signals, ILC2s produced limited amounts of IL-5 and IL-13 and failed to support eosinophil homeostasis. Further, ST2-deficient ILC2s were unable to expand and promote the recruitment of eosinophils during allergic lung inflammation provoked by papain administration. During infection with Nippostrongylus brasiliensis, ILC2-intrinsic ST2 signals were required to mount an effective type 2 immune response against the parasite leading to higher susceptibility against worm infection in conditional knockout mice. Therefore, this study argues for a non-redundant role of cell-intrinsic ST2 signals triggering proper activation of ILC2 for initiation of type 2 immunity.
Collapse
Affiliation(s)
- Patrycja M. Topczewska
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Zoe A. Rompe
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Manuel O. Jakob
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Anton Stamm
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Pierre S. Leclère
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Alexandra Preußer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Claudia U. Duerr
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Linda Marie Laura Thole
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for General and Visceral Surgery, Hindenburgdamm, Berlin, Germany
| | - Katja Kotsch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for General and Visceral Surgery, Hindenburgdamm, Berlin, Germany
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Christoph S. N. Klose
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| |
Collapse
|