51
|
Mindt BC, Krisna SS, Duerr CU, Mancini M, Richer L, Vidal SM, Gerondakis S, Langlais D, Fritz JH. The NF-κB Transcription Factor c-Rel Modulates Group 2 Innate Lymphoid Cell Effector Functions and Drives Allergic Airway Inflammation. Front Immunol 2021; 12:664218. [PMID: 34867937 PMCID: PMC8635195 DOI: 10.3389/fimmu.2021.664218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/27/2021] [Indexed: 01/03/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of early type 2 immune responses. Upon tissue damage, ILC2s are activated by alarmins such as IL-33 and rapidly secrete large amounts of type 2 signature cytokines. ILC2 activation is governed by a network of transcriptional regulators including nuclear factor (NF)-κB family transcription factors. While it is known that activating IL-33 receptor signaling results in downstream NF-κB activation, the underlying molecular mechanisms remain elusive. Here, we found that the NF-κB subunit c-Rel is required to mount effective innate pulmonary type 2 immune responses. IL-33-mediated activation of ILC2s in vitro as well as in vivo was found to induce c-Rel mRNA and protein expression. In addition, we demonstrate that IL-33-mediated activation of ILC2s leads to nuclear translocation of c-Rel in pulmonary ILC2s. Although c-Rel was found to be a critical mediator of innate pulmonary type 2 immune responses, ILC2-intrinsic deficiency of c-Rel did not have an impact on the developmental capacity of ILC2s nor affected homeostatic numbers of lung-resident ILC2s at steady state. Moreover, we demonstrate that ILC2-intrinsic deficiency of c-Rel alters the capacity of ILC2s to upregulate the expression of ICOSL and OX40L, key stimulatory receptors, and the expression of type 2 signature cytokines IL-5, IL-9, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Collectively, our data using Rel−/− mice suggest that c-Rel promotes acute ILC2-driven allergic airway inflammation and suggest that c-Rel may contribute to the pathophysiology of ILC2-mediated allergic airway disease. It thereby represents a promising target for the treatment of allergic asthma, and evaluating the effect of established c-Rel inhibitors in this context would be of great clinical interest.
Collapse
Affiliation(s)
- Barbara C. Mindt
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Sai Sakktee Krisna
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Claudia U. Duerr
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Mathieu Mancini
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Lara Richer
- Department of Pathology, McGill University, Montréal, QC, Canada
| | - Silvia M. Vidal
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Steven Gerondakis
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - David Langlais
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Jörg H. Fritz
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
- *Correspondence: Jörg H. Fritz,
| |
Collapse
|
52
|
Kim Y, Ma C, Park S, Shin- Y, Lee T, Paek J, Hoon Kim K, Jang G, Cho H, Son S, Son SH, Yong Lee K, Lee K, Woo Jung Y, Ho Jeon Y, Byun Y. Rational Design, Synthesis and Evaluation of Oxazolo[4,5-c]-quinolinone Analogs as Novel Interleukin-33 Inhibitors. Chem Asian J 2021; 16:3702-3712. [PMID: 34553505 DOI: 10.1002/asia.202100896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/14/2021] [Indexed: 12/30/2022]
Abstract
Interleukin-33 (IL-33) is an epithelial-derived cytokine that plays an important role in immune-mediated diseases such as asthma, atopic dermatitis, and rheumatoid arthritis. Although IL-33 is considered a potential target for the treatment of allergy-related diseases, no small molecule that inhibits IL-33 has been reported. Based on the structure-activity relationship and in vitro 2D NMR studies employing 15 N-labeled IL-33, we identified that the oxazolo[4,5-c]-quinolinone analog 7 c binds to the interface region of IL-33 and IL-33 receptor (ST2), an orphan receptor of the IL-1 receptor family. Compound 7 c effectively inhibited the production of IL-6 in human mast cells in a dose-dependent manner. Compound 7 c is the first low molecular weight IL-33 inhibitor and may be used as a prototype molecule for structural optimization and investigation of the IL-33/ST2 signaling pathway.
Collapse
Affiliation(s)
- Yujin Kim
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Chao Ma
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea.,Current address, College of Food & Pharmaceutical Engineering, Guizhou Institute of Technology, Guizhou, 550003, P. R. China
| | - Seonghu Park
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Yujin Shin-
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Taeyun Lee
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Jiwon Paek
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Kyong Hoon Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, South Korea
| | - Geonhee Jang
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Haelim Cho
- T&J TECH Inc., 212 Gasan digital 1-ro, Geumcheon-gu, Seoul, 08502, South Korea
| | - Seyoung Son
- Azcuris, Co., Ltd., 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Sang-Hyun Son
- Azcuris, Co., Ltd., 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Ki Yong Lee
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Kiho Lee
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Yong Woo Jung
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Young Ho Jeon
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| |
Collapse
|
53
|
Hassoun D, Malard O, Barbarot S, Magnan A, Colas L. Type 2 immunity-driven diseases: Towards a multidisciplinary approach. Clin Exp Allergy 2021; 51:1538-1552. [PMID: 34617355 PMCID: PMC9292742 DOI: 10.1111/cea.14029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Asthma, atopic dermatitis and chronic rhinoconjunctivitis are highly heterogeneous. However, epidemiologic associations exist between phenotypic groups of patients. Atopic march is one such association but is not the only common point. Indeed, beyond such phenotypes, hallmarks of type 2 immunity have been found in these diseases involving immune dysregulation as well as environmental triggers and epithelial dysfunction. From the canonical Th2 cytokines (IL-4, IL-5, IL-13), new cellular and molecular actors arise, from the epithelium's alarmins to new innate immune cells. Their interactions are now better understood across the different environmental barriers, and slight differences appeared. In parallel, the development of type 2-targeting biotherapies not only raised hope to treat those diseases but also raised new questions regarding their true pathophysiological involvement. Here, we review the place of type 2 immunity in the different phenotypes of asthma, chronic rhinitis, chronic rhinosinusitis and atopic dermatitis, highlighting nuances between them. New hypotheses rising from the use of biotherapies will be discussed along with the uncertainties and unmet needs of this field.
Collapse
Affiliation(s)
- Dorian Hassoun
- CHU Nantes, CNRS, INSERM, l'institut du Thorax, Université de Nantes, Nantes, France
| | - Olivier Malard
- Department of Otorhinolaryngology and Head and Neck Surgery, Nantes University Hospital, Nantes, France
| | - Sébastien Barbarot
- Department of Dermatology, CHU Nantes, UMR 1280 PhAN, INRA, Nantes Université, Nantes, France
| | - Antoine Magnan
- INRAe UMR_S 0892, Hôpital Foch, Université de Versailles Saint-Quentin, Paris Saclay, France
| | - Luc Colas
- Plateforme Transversale d'Allergologie et d'Immunologie Clinique, Institut du Thorax, CHU de Nantes, Nantes, France.,INSERM, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, ITUN, Nantes, France
| |
Collapse
|
54
|
Duan Z, Liu M, Yuan L, Du X, Wu M, Yang Y, Wang L, Zhou K, Yang M, Zou Y, Xiang Y, Qu X, Liu H, Qin X, Liu C. Innate lymphoid cells are double-edged swords under the mucosal barrier. J Cell Mol Med 2021; 25:8579-8587. [PMID: 34378306 PMCID: PMC8435454 DOI: 10.1111/jcmm.16856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022] Open
Abstract
As the direct contacting site for pathogens and allergens, the mucosal barrier plays a vital role in the lungs and intestines. Innate lymphoid cells (ILCs) are particularly resident in the mucosal barrier and participate in several pathophysiological processes, such as maintaining or disrupting barrier integrity, preventing various pathogenic invasions. In the pulmonary mucosae, ILCs sometimes aggravate inflammation and mucus hypersecretion but restore airway epithelial integrity and maintain lung tissue homeostasis at other times. In the intestinal mucosae, ILCs can increase epithelial permeability, leading to severe intestinal inflammation on the one hand, and assist mucosal barrier in resisting bacterial invasion on the other hand. In this review, we will illustrate the positive and negative roles of ILCs in mucosal barrier immunity.
Collapse
Affiliation(s)
- Zhen Duan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Mandie Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,China-Africa Infectious Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
55
|
CD52-targeted depletion by Alemtuzumab ameliorates allergic airway hyperreactivity and lung inflammation. Mucosal Immunol 2021; 14:899-911. [PMID: 33731828 PMCID: PMC8225558 DOI: 10.1038/s41385-021-00388-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 02/04/2023]
Abstract
Allergic asthma is a chronic inflammatory disorder associated with airway hyperreactivity (AHR) whose global prevalence is increasing at an alarming rate. Group 2 innate lymphoid cells (ILC2s) and T helper 2 (TH2) cells are producers of type 2 cytokines, which may contribute to development of AHR. In this study, we explore the potential of CD52-targeted depletion of type 2 immune cells for treating allergic AHR. Here we show that anti-CD52 therapy can prevent and remarkably reverse established IL-33-induced AHR by reducing airway resistance and alleviating lung inflammation. We further show that CD52 depletion prevents and treats allergic AHR induced by clinically relevant allergens such as Alternaria alternata and house dust mite. Importantly, we leverage various humanized mice models of AHR to show new therapeutic applications for Alemtuzumab, an anti-CD52 depleting antibody that is currently FDA approved for treatment of multiple sclerosis. Our results demonstrate that CD52 depletion is a viable therapeutic option for reduction of pulmonary inflammation, abrogation of eosinophilia, improvement of lung function, and thus treatment of allergic AHR. Taken together, our data suggest that anti-CD52 depleting monoclonal antibodies, such as Alemtuzumab, can serve as viable therapeutic drugs for amelioration of TH2- and ILC2-dependent AHR.
Collapse
|
56
|
Kotas ME, Dion J, Van Dyken S, Ricardo-Gonzalez RR, Danel CJ, Taillé C, Mouthon L, Locksley RM, Terrier B. A role for IL-33-activated ILC2s in eosinophilic vasculitis. JCI Insight 2021; 6:143366. [PMID: 33974563 PMCID: PMC8262498 DOI: 10.1172/jci.insight.143366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/05/2021] [Indexed: 01/21/2023] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare but serious disease with poorly understood mechanisms. Here, we report that patients with EGPA have elevated levels of TSLP, IL-25, and soluble ST2, which are well-characterized cytokine “alarmins” that activate or modulate type 2 innate lymphoid cells (ILC2s). Patients with active EGPA have a concurrent reduction in circulating ILC2s, suggesting a role for ILC2s in the pathogenesis of this disease. To explore the mechanism of these findings in patients, we established a model of EGPA in which active vasculitis and pulmonary hemorrhage were induced by IL-33 administration in predisposed, hypereosinophilic mice. In this model, induction of pulmonary hemorrhage and vasculitis was dependent on ILC2s and signaling through IL4Rα. In the absence of IL4Rα or STAT6, IL-33–treated mice had less vascular leak and pulmonary edema, less endothelial activation, and reduced eotaxin production, cumulatively leading to a reduction of pathologic eosinophil migration into the lung parenchyma. These results offer a mouse model for use in future mechanistic studies of EGPA, and they suggest that IL-33, ILC2s, and IL4Rα signaling may be potential targets for further study and therapeutic targeting in patients with EGPA.
Collapse
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy & Sleep Medicine, University of California, San Francisco, California, USA
| | - Jérémie Dion
- Department of Internal Medicine, National Referral Center for Rare and Systemic Autoimmune Diseases, Cochin Hospital, AP-HP, Paris, France
| | - Steven Van Dyken
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, Missouri, USA
| | | | | | - Camille Taillé
- Department of Pulmonology, Bichat Hospital, AP-HP, Paris, France
| | - Luc Mouthon
- Department of Internal Medicine, National Referral Center for Rare and Systemic Autoimmune Diseases, Cochin Hospital, AP-HP, Paris, France
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, California, USA.,Department of Medicine, University of California, San Francisco, California, USA
| | - Benjamin Terrier
- Department of Internal Medicine, National Referral Center for Rare and Systemic Autoimmune Diseases, Cochin Hospital, AP-HP, Paris, France
| |
Collapse
|
57
|
Yu MY, Kwon S, Moon JJ, Kim YC, Song EY, Lee H, Moon KC, Ha J, Kim DK, Han SW, Kim GH, Kim YS, Yang SH. Role of the IL-33/ST2 pathway in renal allograft rejection. Exp Cell Res 2021; 405:112705. [PMID: 34166678 DOI: 10.1016/j.yexcr.2021.112705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 11/25/2022]
Abstract
The interleukin-33 (IL-33)/suppression of tumorigenicity 2 (ST2) pathway modulates immune response and inflammation, associated with allograft dysfunction and rejection. We hypothesized that IL-33/ST2 is a marker of renal allograft rejection and IL-33/ST2 expression may differ according to rejection type. IL-33/ST2 expression was measured in sera and kidney tissues from recipients with acute antibody-mediated rejection (AAMR), acute cell-mediated rejection (ACMR), chronic antibody-mediated rejection (CAMR), and healthy controls. The soluble ST2 and IL-33/ST2 expression levels were higher in participants with all three rejection types than in controls. Although the expression levels in recipients with AAMR and ACMR were significantly higher than those with CAMR, there was no significant difference between the expression levels in AAMR and ACMR. Although IL-33, IL-8, and fibronectin expression were significantly increased after the addition of the recipients' serum in primary cultured human renal proximal tubular epithelial cells, the levels decreased after treatment with an anti-ST2 antibody. Furthermore, the anti-ST2 antibody specifically suppressed the upregulation of the mixed lymphocyte reaction. Boyden chamber assays demonstrated that anti-ST2 antibody abrogated chemotaxis induced by recombinant IL-33. Thus, IL-33 and ST2 are potent mediators of rejection. Treatment with an anti-ST2 antibody ameliorates rejection and could be a potential therapeutic strategy for renal allograft rejection.
Collapse
Affiliation(s)
- Mi-Yeon Yu
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea; Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Soie Kwon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yong-Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Chul Moon
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang-Woong Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea; Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Gheun-Ho Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
58
|
Shen C, Liu C, Zhang Z, Ping Y, Shao J, Tian Y, Yu W, Qin G, Liu S, Wang L, Zhang Y. PD-1 Affects the Immunosuppressive Function of Group 2 Innate Lymphoid Cells in Human Non-Small Cell Lung Cancer. Front Immunol 2021; 12:680055. [PMID: 34194433 PMCID: PMC8237944 DOI: 10.3389/fimmu.2021.680055] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/31/2021] [Indexed: 01/22/2023] Open
Abstract
Background There is increasing evidence that group 2 innate lymphoid cells (ILC2s) play an essential role in allergy and parasitic infection. However, the role of ILC2s in human lung cancer remains unclear. Methods ILC2s from peripheral blood mononuclear cells (PBMCs) obtained from healthy donors (HDs) and non-small cell lung cancer (NSCLC) patients, and NSCLC tumor tissues were analyzed via multicolor flow cytometry. ILC2s or CD14+ cells were sorted by fluorescence-activated cell sorting. qPCR and flow cytometry were performed to assess the gene and protein expression of the indicated molecules. M1-like and M2-like macrophages were induced from CD14+ monocytes in vitro. Results ILC2s were significantly more enriched in PBMCs and tumor tissues from NSCLC patients than in HDs. After screening for the main immune checkpoint molecules, we found that PD-1 was upregulated in ILC2s in NSCLC patients. Functionally, PD-1high ILC2s from tumor tissues expressed higher levels of IL-4 and IL-13 regarding both mRNA and protein levels than PD-1low ILC2s. Furthermore, PD-1high ILC2s robustly boosted M2-like macrophage polarization in vitro, by secreting IL-4 and IL-13, while neutralization of IL-4 and IL-13 by antibodies abrogated M2-like macrophage polarization. Conclusion ILC2s are enriched in NSCLC patients and upregulate PD-1 expression. Upregulation of PD-1 facilitates the immunosuppressive function of ILC2s. PD-1high ILC2s enhance M2-like macrophage polarization by secreting IL-4 and IL-13. PD-1 acts as a positive regulator of the immunosuppressive function of ILC2s in human NSCLC.
Collapse
Affiliation(s)
- Chunyi Shen
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaojun Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Yu Ping
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingwen Shao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonggui Tian
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weina Yu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guohui Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Shasha Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| |
Collapse
|
59
|
Zaini A, Fulford TS, Grumont RJ, Runting J, Rodrigues G, Ng J, Gerondakis S, Zaph C, Scheer S. c-Rel Is Required for IL-33-Dependent Activation of ILC2s. Front Immunol 2021; 12:667922. [PMID: 34194431 PMCID: PMC8236704 DOI: 10.3389/fimmu.2021.667922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are emerging as important cellular regulators of homeostatic and disease-associated immune processes. The cytokine interleukin-33 (IL-33) promotes ILC2-dependent inflammation and immunity, with IL-33 having been shown to activate NF-κB in a wide variety of cell types. However, it is currently unclear which NF-κB members play an important role in IL-33-dependent ILC2 biology. Here, we identify the NF-κB family member c-Rel as a critical component of the IL-33-dependent activation of ILC2s. Although c-Rel is dispensable for ILC2 development, it is critical for ILC2 function in the lung, with c-Rel-deficient (c-Rel-/- ) mice present a significantly reduced response to papain- and IL-33-induced lung inflammation. We also show that the absence of c-Rel reduces the IL-33-dependent expansion of ILC2 precursors and lower levels of IL-5 and IL-13 cytokine production by mature ILC2s in the lung. Together, these results identify the IL-33-c-Rel axis as a central control point of ILC2 activation and function.
Collapse
Affiliation(s)
- Aidil Zaini
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Thomas S. Fulford
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Raelene J. Grumont
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jessica Runting
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Grace Rodrigues
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Judy Ng
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Steve Gerondakis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Colby Zaph
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Sebastian Scheer
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
60
|
Zheng H, Zhang Y, Pan J, Liu N, Qin Y, Qiu L, Liu M, Wang T. The Role of Type 2 Innate Lymphoid Cells in Allergic Diseases. Front Immunol 2021; 12:586078. [PMID: 34177881 PMCID: PMC8220221 DOI: 10.3389/fimmu.2021.586078] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Allergic diseases are significant diseases that affect many patients worldwide. In the past few decades, the incidence of allergic diseases has increased significantly due to environmental changes and social development, which has posed a substantial public health burden and even led to premature death. The understanding of the mechanism underlying allergic diseases has been substantially advanced, and the occurrence of allergic diseases and changes in the immune system state are known to be correlated. With the identification and in-depth understanding of innate lymphoid cells, researchers have gradually revealed that type 2 innate lymphoid cells (ILC2s) play important roles in many allergic diseases. However, our current studies of ILC2s are limited, and their status in allergic diseases remains unclear. This article provides an overview of the common phenotypes and activation pathways of ILC2s in different allergic diseases as well as potential research directions to improve the understanding of their roles in different allergic diseases and ultimately find new treatments for these diseases.
Collapse
Affiliation(s)
- Haocheng Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiachuang Pan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nannan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Qin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Qiu
- Journal Press of Global Traditional Chinese Medicine, Beijing, China
| | - Min Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
61
|
Transient IL-33 upregulation in neonatal mouse lung promotes acute but not chronic type 2 immune responses induced by allergen later in life. PLoS One 2021; 16:e0252199. [PMID: 34048460 PMCID: PMC8162637 DOI: 10.1371/journal.pone.0252199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/11/2021] [Indexed: 01/02/2023] Open
Abstract
Early life respiratory insults, such as viral infections or hyperoxia, often increase asthma susceptibility later in life. The mechanisms underlying this increased susceptibility are not fully understood. IL-33 has been shown to be critically involved in allergic airway diseases. IL-33 expression in the neonatal lung can be increased by various respiratory insults associated with asthma development. Therefore, we investigated whether and how early life increases in IL-33 impact allergic airway responses later in life. Using a novel IL-33 transgenic mouse model, in which full-length IL-33 was inducible overexpressed in lung epithelial cells, we transiently upregulated lung IL-33 expression in neonatal mice for one week. After resting for 4–6 weeks, mice were intranasally exposed to a single-dose of recombinant IL-33 or the airborne allergen Alternaria. Alternatively, mice were exposed to Alternaria and ovalbumin multiple times for one month. We found that a transient increase in IL-33 expression during the neonatal period promoted IL-5 and IL-13 production when mice were later exposed to a single-dose of IL-33 or Alternaria in adulthood. However, increased IL-33 expression during the neonatal period did not affect airway inflammation, type 2 cytokine production, lung mucus production, or antigen-specific antibody responses when adult mice were exposed to Alternaria and ovalbumin multiple times. These results suggest that transient increased IL-33 expression early in life may have differential effects on allergic airway responses in later life, preferentially affecting allergen-induced acute type 2 cytokine production.
Collapse
|
62
|
Hardman CS, Chen YL, Salimi M, Nahler J, Corridoni D, Jagielowicz M, Fonseka CL, Johnson D, Repapi E, Cousins DJ, Barlow JL, McKenzie ANJ, Simmons A, Ogg G. IL-6 effector function of group 2 innate lymphoid cells (ILC2) is NOD2 dependent. Sci Immunol 2021; 6:eabe5084. [PMID: 34021026 PMCID: PMC7611333 DOI: 10.1126/sciimmunol.abe5084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 02/26/2021] [Accepted: 04/21/2021] [Indexed: 01/24/2023]
Abstract
Cutaneous group 2 innate lymphoid cells (ILC2) are spatially and epigenetically poised to respond to barrier compromise and associated immunological threats. ILC2, lacking rearranged antigen-specific receptors, are primarily activated by damage-associated cytokines and respond with type 2 cytokine production. To investigate ILC2 potential for direct sensing of skin pathogens and allergens, we performed RNA sequencing of ILC2 derived from in vivo challenged human skin or blood. We detected expression of NOD2 and TLR2 by skin and blood ILC2. Stimulation of ILC2 with TLR2 agonist alone not only induced interleukin-5 (IL-5) and IL-13 expression but also elicited IL-6 expression in combination with Staphylococcus aureus muramyl dipeptide (MDP). Heat-killed skin-resident bacteria provoked an IL-6 profile in ILC2 in vitro that was notably impaired in ILC2 derived from patients with nucleotide-binding oligomerization domain-containing protein 2 (NOD2) mutations. In addition, we show that NOD2 signaling can stimulate autophagy in ILC2, which was also impaired in patients with NOD2 mutations. Here, we have identified a role for ILC2 NOD2 signaling in the differential regulation of ILC2-derived IL-6 and have reported a previously unrecognized pathway of direct ILC2 bacterial sensing.
Collapse
Affiliation(s)
- Clare S Hardman
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yi-Ling Chen
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Maryam Salimi
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Janina Nahler
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniele Corridoni
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Jagielowicz
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Chathuranga L Fonseka
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Emmanouela Repapi
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford, UK
| | - David J Cousins
- Department of Infection, Immunity and Inflammation, NIHR Leicester Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | | | | | - Alison Simmons
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
63
|
Miyata J, Yokokura Y, Moro K, Arai H, Fukunaga K, Arita M. 12/15-Lipoxygenase Regulates IL-33-Induced Eosinophilic Airway Inflammation in Mice. Front Immunol 2021; 12:687192. [PMID: 34093589 PMCID: PMC8170304 DOI: 10.3389/fimmu.2021.687192] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Dysregulated fatty acid metabolism is clinically associated with eosinophilic allergic diseases, including severe asthma and chronic rhinosinusitis. This study aimed to demonstrate the role of 12/15-lipoxygenase (12/15-LOX) in interleukin (IL)-33-induced eosinophilic airway inflammation; to this end, we used 12/15-LOX-deficient mice, which displayed augmented IL-33-induced lung inflammation, characterized by an increased number of infiltrated eosinophils and group 2 innate lymphoid cells (ILC2s) in the airway. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomics revealed that the levels of a series of 12/15-LOX-derived metabolites were significantly decreased, and application of 14(S)-hydroxy docosahexaenoic acid (HDoHE), a major 12/15-LOX-derived product, suppressed IL-33-mediated eosinophilic inflammation in 12/15-LOX-deficient mice. Using bioactive lipid screening, we found that 14(S)-HDoHE and 10(S),17(S)-diHDoHE markedly attenuated ILC2 proliferation and cytokine production at micromolar concentration in vitro. In addition, maresin 1 (MaR1) and resolvin D1 (RvD1), 12/15-LOX-derived specialized proresolving mediators (SPMs), inhibited cytokine production of ILC2s at nanomolar concentration. These findings demonstrate the protective role of endogenous 12/15-LOX-derived lipid mediators in controlling ILC2-mediated eosinophilic airway inflammation and related diseases. Thus, 12/15-LOX-derived lipid mediators may represent a potential therapeutic strategy for ameliorating airway inflammation-associated conditions.
Collapse
Affiliation(s)
- Jun Miyata
- Laboratory of Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yoshiyuki Yokokura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Laboratory for Innate Immune Systems, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroyuki Arai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Arita
- Laboratory of Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy, Tokyo, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
64
|
CD200-CD200R immune checkpoint engagement regulates ILC2 effector function and ameliorates lung inflammation in asthma. Nat Commun 2021; 12:2526. [PMID: 33953190 PMCID: PMC8100131 DOI: 10.1038/s41467-021-22832-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/24/2021] [Indexed: 12/25/2022] Open
Abstract
The prevalence of asthma and airway hyperreactivity (AHR) is increasing at an alarming rate. Group 2 innate lymphoid cells (ILC2s) are copious producers of type 2 cytokines, which leads to AHR and lung inflammation. Here, we show that mouse ILC2s express CD200 receptor (CD200R) and this expression is inducible. CD200R engagement inhibits activation, proliferation and type 2 cytokine production, indicating an immunoregulatory function for the CD200-CD200R axis on ILC2s. Furthermore, CD200R engagement inhibits both canonical and non-canonical NF-κB signaling pathways in activated ILC2s. Additionally, we demonstrate both preventative and therapeutic approaches utilizing CD200R engagement on ILC2s, which lead to improved airway resistance, dynamic compliance and eosinophilia. These results show CD200R is expressed on human ILC2s, and its engagement ameliorates AHR in humanized mouse models, emphasizing the translational applications for treatment of ILC2-related diseases such as allergic asthma.
Collapse
|
65
|
Rodriguez-Rodriguez N, Gogoi M, McKenzie AN. Group 2 Innate Lymphoid Cells: Team Players in Regulating Asthma. Annu Rev Immunol 2021; 39:167-198. [PMID: 33534604 PMCID: PMC7614118 DOI: 10.1146/annurev-immunol-110119-091711] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Type 2 immunity helps protect the host from infection, but it also plays key roles in tissue homeostasis, metabolism, and repair. Unfortunately, inappropriate type 2 immune reactions may lead to allergy and asthma. Group 2 innate lymphoid cells (ILC2s) in the lungs respond rapidly to local environmental cues, such as the release of epithelium-derived type 2 initiator cytokines/alarmins, producing type 2 effector cytokines such as IL-4, IL-5, and IL-13 in response to tissue damage and infection. ILC2s are associated with the severity of allergic asthma, and experimental models of lung inflammation have shown how they act as playmakers, receiving signals variously from stromal and immune cells as well as the nervous system and then distributing cytokine cues to elicit type 2 immune effector functions and potentiate CD4+ T helper cell activation, both of which characterize the pathology of allergic asthma. Recent breakthroughs identifying stromal- and neuronal-derived microenvironmental cues that regulate ILC2s, along with studies recognizing the potential plasticity of ILC2s, have improved our understanding of the immunoregulation of asthma and opened new avenues for drug discovery.
Collapse
Affiliation(s)
- Noe Rodriguez-Rodriguez
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Mayuri Gogoi
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Andrew N.J. McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK,Corresponding author:
| |
Collapse
|
66
|
Rajput C, Han M, Ishikawa T, Lei J, Goldsmith AM, Jazaeri S, Stroupe CC, Bentley JK, Hershenson MB. Rhinovirus C Infection Induces Type 2 Innate Lymphoid Cell Expansion and Eosinophilic Airway Inflammation. Front Immunol 2021; 12:649520. [PMID: 33968043 PMCID: PMC8100319 DOI: 10.3389/fimmu.2021.649520] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Rhinovirus C (RV-C) infection is associated with severe asthma exacerbations. Since type 2 inflammation is an important disease mechanism in asthma, we hypothesized that RV-C infection, in contrast to RV-A, preferentially stimulates type 2 inflammation, leading to exacerbated eosinophilic inflammation. To test this, we developed a mouse model of RV-C15 airways disease. RV-C15 was generated from the full-length cDNA clone and grown in HeLa-E8 cells expressing human CDHR3. BALB/c mice were inoculated intranasally with 5 x 106 ePFU RV-C15, RV-A1B or sham. Mice inoculated with RV-C15 showed lung viral titers of 1 x 105 TCID50 units 24 h after infection, with levels declining thereafter. IFN-α, β, γ and λ2 mRNAs peaked 24-72 hrs post-infection. Immunofluorescence verified colocalization of RV-C15, CDHR3 and acetyl-α-tubulin in mouse ciliated airway epithelial cells. Compared to RV-A1B, mice infected with RV-C15 demonstrated higher bronchoalveolar eosinophils, mRNA expression of IL-5, IL-13, IL-25, Muc5ac and Gob5/Clca, protein production of IL-5, IL-13, IL-25, IL-33 and TSLP, and expansion of type 2 innate lymphoid cells. Analogous results were found in mice treated with house dust mite before infection, including increased airway responsiveness. In contrast to Rorafl/fl littermates, RV-C-infected Rorafl/flIl7rcre mice deficient in ILC2s failed to show eosinophilic inflammation or mRNA expression of IL-13, Muc5ac and Muc5b. We conclude that, compared to RV-A1B, RV-C15 infection induces ILC2-dependent type 2 airway inflammation, providing insight into the mechanism of RV-C-induced asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marc B. Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
67
|
Mathä L, Martinez-Gonzalez I, Steer CA, Takei F. The Fate of Activated Group 2 Innate Lymphoid Cells. Front Immunol 2021; 12:671966. [PMID: 33968080 PMCID: PMC8100346 DOI: 10.3389/fimmu.2021.671966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in both mucosal and non-mucosal tissues and play critical roles in the first line of defense against parasites and irritants such as allergens. Upon activation by cytokines released from epithelial and stromal cells during tissue damage or stimulation, ILC2s produce copious amounts of IL-5 and IL-13, leading to type 2 inflammation. Over the past 10 years, ILC2 involvement in a variety of human diseases has been unveiled. However, questions remain as to the fate of ILC2s after activation and how that might impact their role in chronic inflammatory diseases such as asthma and fibrosis. Here, we review studies that have revealed novel properties of post-activation ILC2s including the generation of immunological memory, exhausted-like phenotype, transdifferentiation and activation-induced migration.
Collapse
Affiliation(s)
- Laura Mathä
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | | | - Catherine A Steer
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
68
|
Saikumar Jayalatha AK, Hesse L, Ketelaar ME, Koppelman GH, Nawijn MC. The central role of IL-33/IL-1RL1 pathway in asthma: From pathogenesis to intervention. Pharmacol Ther 2021; 225:107847. [PMID: 33819560 DOI: 10.1016/j.pharmthera.2021.107847] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-33 (IL-33), a member of the IL-1 family, and its cognate receptor, Interleukin-1 receptor like-1 (IL-1RL1 or ST2), are susceptibility genes for childhood asthma. In response to cellular damage, IL-33 is released from barrier tissues as an 'alarmin' to activate the innate immune response. IL-33 drives type 2 responses by inducing signalling through its receptor IL-1RL1 in several immune and structural cells, thereby leading to type 2 cytokine and chemokine production. IL-1RL1 gene transcript encodes different isoforms generated through alternative splicing. Its soluble isoform, IL-1RL1-a or sST2, acts as a decoy receptor by sequestering IL-33, thereby inhibiting IL1RL1-b/IL-33 signalling. IL-33 and its receptor IL-1RL1 are therefore considered as putative biomarkers or targets for pharmacological intervention in asthma. This review will provide an overview of the genetics and biology of the IL-33/IL-1RL1 pathway in the context of asthma pathogenesis. It will discuss the potential and complexities of targeting the cytokine or its receptor, how genetics or biomarkers may inform precision medicine for asthma targeting this pathway, and the possible positioning of therapeutics targeting IL-33 or its receptor in the expanding landscape of novel biologicals applied in asthma management.
Collapse
Affiliation(s)
- A K Saikumar Jayalatha
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - L Hesse
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - M E Ketelaar
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - G H Koppelman
- University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - M C Nawijn
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands.
| |
Collapse
|
69
|
de Lucía Finkel P, Xia W, Jefferies WA. Beyond Unconventional: What Do We Really Know about Group 2 Innate Lymphoid Cells? THE JOURNAL OF IMMUNOLOGY 2021; 206:1409-1417. [PMID: 33753565 DOI: 10.4049/jimmunol.2000812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/24/2020] [Indexed: 01/20/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a set of effectors that mediate the expulsion of helminthic parasites but also drive allergic lung inflammation. As innate agents, they do not recognize Ag, instead, they are sensitive to alarmin engagement, upon which they produce type 2 cytokines that amplify adaptive immunity. Their lymphoid identity appoints them as an intriguing group of unconventional cells; however, increasing evidence is unraveling a series of unprecedented functions that <5 years ago were unthinkable for ILC2s, such as acquiring a proinflammatory identity that enables them to support TH1 immune responses. Their plastic nature has allowed the characterization of ILC2s in more detail than ever; however, the novelty of ILC2 biology requires constant updates and recapitulations. This review provides an overview of ILC2s and describes memory ILC2, regulatory ILC2, inflammatory ILC2, and type 1 ILC2 subsets based on activation status, tissue environments, and function.
Collapse
Affiliation(s)
- Pablo de Lucía Finkel
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia V6H 3Z6, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Wenjing Xia
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia V6H 3Z6, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; .,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia V6H 3Z6, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; and.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| |
Collapse
|
70
|
Howard E, Lewis G, Galle-Treger L, Hurrell BP, Helou DG, Shafiei-Jahani P, Painter JD, Muench GA, Soroosh P, Akbari O. IL-10 production by ILC2s requires Blimp-1 and cMaf, modulates cellular metabolism, and ameliorates airway hyperreactivity. J Allergy Clin Immunol 2021; 147:1281-1295.e5. [PMID: 32905799 DOI: 10.1016/j.jaci.2020.08.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/21/2020] [Accepted: 08/21/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are the dominant innate lymphoid cell population in the lungs at steady state, and their release of type 2 cytokines is a central driver in responding eosinophil infiltration and increased airway hyperreactivity. Our laboratory has identified a unique subset of ILC2s in the lungs that actively produce IL-10 (ILC210s). OBJECTIVE Our aim was to characterize the effector functions of ILC210s in the development and pathology of allergic asthma. METHODS IL-4-stimulated ILC210s were isolated to evaluate cytokine secretion, transcription factor signaling, metabolic dependence, and effector functions in vitro. ILC210s were also adoptively transferred into Rag2-/-γc-/- mice, which were then challenged with IL-33 and assessed for airway hyperreactivity and lung inflammation. RESULTS We have determined that the transcription factors cMaf and Blimp-1 regulate IL-10 expression in ILC210s. Strikingly, our results demonstrate that ILC210s can utilize both autocrine and paracrine signaling to suppress proinflammatory ILC2 effector functions in vitro. Further, this subset dampens airway hyperreactivity and significantly reduces lung inflammation in vivo. Interestingly, ILC210s demonstrated a metabolic dependency on the glycolytic pathway for IL-10 production, shifting from the fatty acid oxidation pathway conventionally utilized for proinflammatory effector functions. CONCLUSION These findings provide an important and previously unrecognized role of ILC210s in diseases associated with ILC2s such as allergic lung inflammation and asthma. They also provide new insights into the metabolism dependency of proinflammatory and anti-inflammatory ILC2 phenotypes.
Collapse
Affiliation(s)
- Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Gavin Lewis
- Janssen Research and Development, San Diego, Calif
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | | | | | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
| |
Collapse
|
71
|
Hennessy C, Lewik G, Cross A, Hester J, Issa F. Recent advances in our understanding of the allograft response. Fac Rev 2021; 10:21. [PMID: 33718938 PMCID: PMC7946390 DOI: 10.12703/r/10-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Organ transplantation is a life-saving treatment for end-stage organ failure. However, despite advances in immunosuppression, donor matching, tissue typing, and organ preservation, many organs are still lost each year to rejection. Ultimately, tolerance in the absence of immunosuppression is the goal, and although this seldom occurs spontaneously, a deeper understanding of alloimmunity may provide avenues for future therapies which aid in its establishment. Here, we highlight the recent key advances in our understanding of the allograft response. On the innate side, recent work has highlighted the previously unrecognised role of innate lymphoid cells as well as natural killer cells in promoting the alloresponse. The two major routes of allorecognition have recently been joined by a third newly identified pathway, semi-direct allorecognition, which is proving to be a key active pathway in transplantation. Through this review, we detail these newly defined areas in the allograft response and highlight areas for potential future therapeutic intervention.
Collapse
Affiliation(s)
- Conor Hennessy
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Guido Lewik
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Amy Cross
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
72
|
Scadding GK, Scadding GW. Innate and Adaptive Immunity: ILC2 and Th2 Cells in Upper and Lower Airway Allergic Diseases. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:1851-1857. [PMID: 33618052 DOI: 10.1016/j.jaip.2021.02.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
Advances in our understanding of the immune system, with the recent discovery of a parallel set of innate T lymphocytes, the innate lymphocytes (ILCs), have led to a reassessment of the pathogenesis of allergic and eosinophilic airway disorders, including allergic rhinitis (AR), asthma, and chronic rhinosinusitis with nasal polyps. We review current understanding of both elements of type-2 inflammatory responses and their relative influence in these common conditions and consider possible impacts of this on treatment selection.
Collapse
|
73
|
Pelaia C, Pelaia G, Crimi C, Longhini F, Lombardo N, Savino R, Sciacqua A, Vatrella A. Biologics in severe asthma. Minerva Med 2021; 113:51-62. [PMID: 33555158 DOI: 10.23736/s0026-4806.21.07296-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Asthma is a chronic airway disease consisting of usually variable airflow limitation and bronchial hyperresponsiveness. Many different phenotypes characterize the clinical expression of asthma, determined by heterogeneous inflammatory patterns driven by distinct cellular and molecular mechanisms known as endotypes. Inside the complex framework of asthma pathobiology, several molecules such as immunoglobulins E (IgE), pro-inflammatory cytokines and their receptors can be targeted by present and future biological treatments of severe asthma. Within this context, already registered monoclonal antibodies including omalizumab, mepolizumab, reslizumab, benralizumab and dupilumab may interfere at various levels with the pathogenic pathways responsible for type-2 airway inflammation. In particular, these drugs target IgE (omalizumab), IL-5 (mepolizumab and reslizumab), IL-5 receptor (benralizumab) and IL-4/IL-13 receptors (dupilumab), respectively. Moreover, other biological therapies are under evaluation in pre-marketing trials, mainly aimed to assess the efficacy and safety of monoclonal antibodies directed against innate cytokines such as IL-33 and thymic stromal lymphopoietin (TSLP). Among current and perspective therapeutic approaches, clinicians can choose phenotype/endotype-driven tailored treatments, able to pursue an effective control of difficult to treat type-2 asthma.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy -
| | - Giulia Pelaia
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Claudia Crimi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Federico Longhini
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Nicola Lombardo
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Rocco Savino
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| |
Collapse
|
74
|
Mukendi JPK, Nakamura R, Uematsu S, Hamano S. Interleukin (IL)-33 is dispensable for Schistosoma mansoni worm maturation and the maintenance of egg-induced pathology in intestines of infected mice. Parasit Vectors 2021; 14:70. [PMID: 33482904 PMCID: PMC7821721 DOI: 10.1186/s13071-020-04561-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Schistosomes are trematode worms that dwell in their definitive host's blood vessels, where females lay eggs that need to be discharged into the environment with host excreta to maintain their life-cycle. Both worms and eggs require type 2 immunity for their maturation and excretion, respectively. However, the immune molecules that orchestrate such immunity remain unclear. Interleukin (IL)-33 is one of the epithelium-derived cytokines that induce type 2 immunity in tissues. The aim of this study was to determine the role of IL-33 in the maturation, reproduction and excretion of Schistosoma mansoni eggs, and in the maintenance of egg-induced pathology in the intestines of mice. METHODS The morphology of S. mansoni worms and the number of eggs in intestinal tissues were studied at different time points post-infection in S. mansoni-infected IL-33-deficient (IL-33-/-) and wild-type (WT) mice. IL-5 and IL-13 production in the spleens and mesenteric lymph nodes were measured. Tissue histology was performed on the terminal ilea of both infected and non-infected mice. RESULTS Worms from IL-33-/- and WT mice did not differ morphologically at 4 and 6 weeks post-infection (wpi). The number of eggs in intestinal tissues of IL-33-/- and WT mice differed only slightly. At 6 wpi, IL-33-/- mice presented impaired type 2 immunity in the intestines, characterized by a decreased production of IL-5 and IL-13 in mesenteric lymph nodes and fewer inflammatory infiltrates with fewer eosinophils in the ilea. There was no difference between IL-33-/- and WT mice in the levels of IL-25 and thymic stromal lymphopoietin (TSLP) in intestinal tissues. CONCLUSIONS Despite its ability to initiate type 2 immunity in tissues, IL-33 alone seems dispensable for S. mansoni maturation and its absence may not affect much the accumulation of eggs in intestinal tissues. The transient impairment of type 2 immunity observed in the intestines, but not spleens, highlights the importance of IL-33 over IL-25 and TSLP in initiating, but not maintaining, locally-induced type 2 immunity in intestinal tissues during schistosome infection. Further studies are needed to decipher the role of each of these molecules in schistosomiasis and clarify the possible interactions that might exist between them.
Collapse
Affiliation(s)
- Jean Pierre Kambala Mukendi
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Risa Nakamura
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinjiro Hamano
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| |
Collapse
|
75
|
Cephus J, Gandhi VD, Shah R, Brooke Davis J, Fuseini H, Yung JA, Zhang J, Kita H, Polosukhin VV, Zhou W, Newcomb DC. Estrogen receptor-α signaling increases allergen-induced IL-33 release and airway inflammation. Allergy 2021; 76:255-268. [PMID: 32648964 DOI: 10.1111/all.14491] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/04/2020] [Accepted: 06/20/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2) are stimulated by IL-33 to increase IL-5 and IL-13 production and airway inflammation. While sex hormones regulate airway inflammation, it remained unclear whether estrogen signaling through estrogen receptor-α (ER-α, Esr1) or ER-β (Esr2) increased ILC2-mediated airway inflammation. We hypothesize that estrogen signaling increases allergen-induced IL-33 release, ILC2 cytokine production, and airway inflammation. METHODS Female Esr1-/- , Esr2-/- , wild-type (WT), and IL33fl/fl eGFP mice were challenged with Alternaria extract (Alt Ext) or vehicle for 4 days. In select experiments, mice were administered tamoxifen or vehicle pellets for 21 days prior to challenge. Lung ILC2, IL-5 and IL-13 production, and BAL inflammatory cells were measured on day 5 of Alt Ext challenge model. Bone marrow from WT and Esr1-/- female mice was transferred (1:1 ratio) into WT female recipients for 6 weeks followed by Alt Ext challenge. hBE33 cells and normal human bronchial epithelial cells (NHBE) were pretreated with 17β-estradiol (E2), propyl-pyrazole-triol (PPT, ER-α agonist), or diarylpropionitrile (DPN, ER-β agonist) before allergen challenge to determine IL-33 gene expression and release, extracellular ATP release, DUOX-1 production, and necrosis. RESULTS Alt Ext challenged Esr1-/- , but not Esr2-/- , mice had decreased IL-5 and IL-13 production, BAL eosinophils, and IL-33 release compared to WT mice. Tamoxifen decreased IL-5 and IL-13 production and BAL eosinophils. IL-33eGFP + epithelial cells were decreased in Alt Ext challenged Esr1-/- mice compared to WT mice. 17β-E2 or PPT, but not DPN, increased IL-33 gene expression, release, and DUOX-1 production in hBE33 or NHBE cells. CONCLUSION Estrogen receptor -α signaling increased IL-33 release and ILC2-mediated airway inflammation.
Collapse
Affiliation(s)
- Jacqueline‐Yvonne Cephus
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Vivek D. Gandhi
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Ruchi Shah
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Jordan Brooke Davis
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Hubaida Fuseini
- Department of Pathology, Microbiology, and Immunology Vanderbilt University Nashville Tennessee USA
| | - Jeffrey A. Yung
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Jian Zhang
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Hirohito Kita
- Allergic Diseases Research Laboratory Mayo Clinic Phoenix Arizona USA
| | - Vasiliy V. Polosukhin
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Weisong Zhou
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Dawn C. Newcomb
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
- Department of Pathology, Microbiology, and Immunology Vanderbilt University Nashville Tennessee USA
| |
Collapse
|
76
|
Panova V, Gogoi M, Rodriguez-Rodriguez N, Sivasubramaniam M, Jolin HE, Heycock MWD, Walker JA, Rana BMJ, Drynan LF, Hodskinson M, Pannell R, King G, Wing M, Easton AJ, Oedekoven CA, Kent DG, Fallon PG, Barlow JL, McKenzie ANJ. Group-2 innate lymphoid cell-dependent regulation of tissue neutrophil migration by alternatively activated macrophage-secreted Ear11. Mucosal Immunol 2021; 14:26-37. [PMID: 32457448 PMCID: PMC7790759 DOI: 10.1038/s41385-020-0298-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 02/04/2023]
Abstract
Type-2 immunity is characterised by interleukin (IL)-4, IL-5 and IL-13, eosinophilia, mucus production, IgE, and alternatively activated macrophages (AAM). However, despite the lack of neutrophil chemoattractants such as CXCL1, neutrophils, a feature of type-1 immunity, are observed in type-2 responses. Consequently, alternative mechanisms must exist to ensure that neutrophils can contribute to type-2 immune reactions without escalation of deleterious inflammation. We now demonstrate that type-2 immune-associated neutrophil infiltration is regulated by the mouse RNase A homologue, eosinophil-associated ribonuclease 11 (Ear11), which is secreted by AAM downstream of IL-25-stimulated ILC2. Transgenic overexpression of Ear11 resulted in tissue neutrophilia, whereas Ear11-deficient mice have fewer resting tissue neutrophils, whilst other type-2 immune responses are not impaired. Notably, administration of recombinant mouse Ear11 increases neutrophil motility and recruitment. Thus, Ear11 helps maintain tissue neutrophils at homoeostasis and during type-2 reactions when chemokine-producing classically activated macrophages are infrequently elicited.
Collapse
Affiliation(s)
- Veera Panova
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK ,grid.451388.30000 0004 1795 1830Present Address: The Francis Crick Institute, London, NW1 1AT UK
| | - Mayuri Gogoi
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Noe Rodriguez-Rodriguez
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Meera Sivasubramaniam
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Helen E. Jolin
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Morgan W. D. Heycock
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Jennifer A. Walker
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Batika M. J. Rana
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Lesley F. Drynan
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Michael Hodskinson
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Richard Pannell
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Gareth King
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Mark Wing
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| | - Andrew J. Easton
- grid.7372.10000 0000 8809 1613School of Life Sciences, University of Warwick, Coventry, CV4 7AL UK
| | | | - David G. Kent
- Stem Cell Institute, Clifford-Allbutt Building, Hills Road, Cambridge, CB2 0AH UK ,grid.5685.e0000 0004 1936 9668Present Address: Department of Biology, University of York, Wentworth Way, York, YO10 5DD UK
| | - Padraic G. Fallon
- grid.8217.c0000 0004 1936 9705Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Jillian L. Barlow
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK ,grid.5685.e0000 0004 1936 9668Present Address: Department of Biology, University of York, Wentworth Way, York, YO10 5DD UK
| | - Andrew N. J. McKenzie
- grid.42475.300000 0004 0605 769XMedical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH UK
| |
Collapse
|
77
|
Wu Y, Shi W, Wang H, Yue J, Mao Y, Zhou W, Kong X, Guo Q, Zhang L, Xu P, Wang Y. Anti-ST2 Nanoparticle Alleviates Lung Inflammation by Targeting ILC2s-CD4 +T Response. Int J Nanomedicine 2020; 15:9745-9758. [PMID: 33299314 PMCID: PMC7721292 DOI: 10.2147/ijn.s268282] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/04/2020] [Indexed: 11/23/2022] Open
Abstract
Background Asthma has been regarded as an inflammatory disease, and group 2 innate lymphoid cells (ILC2s) are implicated in asthma pathogenesis. However, no strategy is available to block ILC2s function. Efficiency is also limited due to the use of systemic or subcutaneous routes of administration. The purpose of this study was to investigate the effects of nanoparticles targeting suppression of tumorigenicity 2 (ST2), which is the ILC2 receptor, to alleviate lung inflammation in the murine model of asthma. Methods The ultra-small SPIO nanoparticles were firstly synthesized, OVA-induced mice were administered by anti-ST2-conjugated nanoparticles. The inflammatory degree of the lung was investigated by H&E. The percentages of ILC2s and CD4+T cells in bronchoalveolar lavage fluid (BALF) and lung tissue were determined by FACS. Th2-cytokine and OVA-IgE levels were detected by real-time PCR and ELISA, respectively. Results Treatment with anti-ST2-conjugated nanoparticles significantly alleviated airway inflammation, IL-33 and IL-13 levels and the percentage of CD4+T cells. The percentage of ILC2s was increased, whereas the levels of IL-13 and IL-5 expressed by ILC2s were reduced. Conclusion In the present study, we demonstrated that anti-ST2-conjugated nanoparticles can efficiently control lung inflammation in OVA-induced mice by reducing the ability of ILC2s to produce IL-5 and IL-13, thereby reducing CD4+T cells. Our study also demonstrated that the nanoparticle delivery system could improve the performance of anti-ST2, which may be used as a strategic tool to expand the current drug market.
Collapse
Affiliation(s)
- Yumin Wu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Weifeng Shi
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Honghai Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Jiawei Yue
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Yijie Mao
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Wei Zhou
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Xinagmin Kong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Qiqiong Guo
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Lirong Zhang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Pengxiao Xu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Yuyue Wang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| |
Collapse
|
78
|
Pelaia C, Crimi C, Vatrella A, Tinello C, Terracciano R, Pelaia G. Molecular Targets for Biological Therapies of Severe Asthma. Front Immunol 2020; 11:603312. [PMID: 33329598 PMCID: PMC7734054 DOI: 10.3389/fimmu.2020.603312] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Asthma is a heterogeneous respiratory disease characterized by usually reversible bronchial obstruction, which is clinically expressed by different phenotypes driven by complex pathobiological mechanisms (endotypes). Within this context, during the last years several molecular effectors and signalling pathways have emerged as suitable targets for biological therapies of severe asthma, refractory to standard treatments. Indeed, various therapeutic antibodies currently allow to intercept at different levels the chain of pathogenic events leading to type 2 (T2) airway inflammation. In addition to pro-allergic immunoglobulin E (IgE), that chronologically represents the first molecule against which an anti-asthma monoclonal antibody (omalizumab) was developed, today other targets are successfully exploited by biological treatments of severe asthma. In particular, pro-eosinophilic interleukin 5 (IL-5) can be targeted by mepolizumab or reslizumab, whereas benralizumab is a selective blocker of IL-5 receptor. Moreover, dupilumab behaves as a dual receptor antagonist of pleiotropic interleukins 4 (IL-4) and 13 (IL-13). Besides these drugs that are already available in medical practice, other biologics are under clinical development such as those targeting innate cytokines, also including the alarmin thymic stromal lymphopoietin (TSLP), which plays a key role in the pathogenesis of type 2 asthma. Therefore, ongoing and future biological therapies are significantly changing the global scenario of severe asthma management. These new therapeutic options make it possible to implement phenotype/endotype-specific treatments, that are delineating personalized approaches precisely addressing the individual traits of asthma pathobiology. Such tailored strategies are thus allowing to successfully target the immune-inflammatory responses underlying uncontrolled T2-high asthma.
Collapse
Affiliation(s)
- Corrado Pelaia
- Respiratory Medicine Unit, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Claudia Crimi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Caterina Tinello
- Pediatrics Unit, Provincial Outpatient Center of Catanzaro, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
79
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
80
|
Porsbjerg CM, Sverrild A, Lloyd CM, Menzies-Gow AN, Bel EH. Anti-alarmins in asthma: targeting the airway epithelium with next-generation biologics. Eur Respir J 2020; 56:2000260. [PMID: 32586879 PMCID: PMC7676874 DOI: 10.1183/13993003.00260-2020] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
Abstract
Monoclonal antibody therapies have significantly improved treatment outcomes for patients with severe asthma; however, a significant disease burden remains. Available biologic treatments, including anti-immunoglobulin (Ig)E, anti-interleukin (IL)-5, anti-IL-5Rα and anti-IL-4Rα, reduce exacerbation rates in study populations by approximately 50% only. Furthermore, there are currently no effective treatments for patients with severe, type 2-low asthma. Existing biologics target immunological pathways that are downstream in the type 2 inflammatory cascade, which may explain why exacerbations are only partly abrogated. For example, type 2 airway inflammation results from several inflammatory signals in addition to IL-5. Clinically, this can be observed in how fractional exhaled nitric oxide (F eNO), which is driven by IL-13, may remain unchanged during anti-IL-5 treatment despite reduction in eosinophils, and how eosinophils may remain unchanged during anti-IL-4Rα treatment despite reduction in F eNO The broad inflammatory response involving cytokines including IL-4, IL-5 and IL-13 that ultimately results in the classic features of exacerbations (eosinophilic inflammation, mucus production and bronchospasm) is initiated by release of "alarmins" thymic stromal lymphopoietin (TSLP), IL-33 and IL-25 from the airway epithelium in response to triggers. The central, upstream role of these epithelial cytokines has identified them as strong potential therapeutic targets to prevent exacerbations and improve lung function in patients with type 2-high and type 2-low asthma. This article describes the effects of alarmins and discusses the potential role of anti-alarmins in the context of existing biologics. Clinical phenotypes of patients who may benefit from these treatments are also discussed, including how biomarkers may help identify potential responders.
Collapse
Affiliation(s)
| | - Asger Sverrild
- Dept of Respiratory Medicine, Bispebjerg Hospital, Copenhagen, Denmark
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Elisabeth H Bel
- Dept of Respiratory Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
81
|
Hurrell BP, Howard E, Galle-Treger L, Helou DG, Shafiei-Jahani P, Painter JD, Akbari O. Distinct Roles of LFA-1 and ICAM-1 on ILC2s Control Lung Infiltration, Effector Functions, and Development of Airway Hyperreactivity. Front Immunol 2020; 11:542818. [PMID: 33193309 PMCID: PMC7662114 DOI: 10.3389/fimmu.2020.542818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022] Open
Abstract
Asthma is a heterogeneous airway inflammatory disease characterized by increased airway hyperreactivity (AHR) to specific and unspecific stimuli. Group 2 innate lymphoid cells (ILC2)s are type-2 cytokine secreting cells capable of inducing eosinophilic lung inflammation and AHR independent of adaptive immunity. Remarkably, reports show that ILC2s are increased in the blood of human asthmatics as compared to healthy donors. Nevertheless, whether ILC2 expression of adhesion molecules regulates ILC2 trafficking remains unknown. Our results show that IL-33-activated ILC2s not only express LFA-1 but also strikingly LFA-1 ligand ICAM-1. Both LFA-1-/- and ICAM-1-/- mice developed attenuated AHR in response to IL-33 intranasal challenge, associated with a lower airway inflammation and less lung ILC2 accumulation compared to controls. Our mixed bone marrow chimera studies however revealed that ILC2 expression of LFA-1 - but not ICAM-1 - was required for their accumulation in the inflamed lungs. Importantly, we found that LFA-1 remarkably controlled ILC2 homing to the lungs, suggesting that LFA-1 is involved in ILC2 trafficking to the lungs. Our exploratory transcriptomic analysis further revealed that ICAM-1 deficiency on ILC2s significantly affects their effector functions. While it downregulated pro-inflammatory cytokines such as Il5, Il9, Il13, and Csf2, it however notably also upregulated cytokines including Il10 both at the transcriptomic and protein levels. These findings provide novel avenues for future investigations, as modulation of LFA-1 and/or ICAM-1 represents an unappreciated regulatory mechanism for ILC2 trafficking and cytokine production respectively, potentially serving as therapeutic target for ILC2-dependent diseases such as allergic asthma.
Collapse
Affiliation(s)
- Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
82
|
He J, Yang Q, Xiao Q, Lei A, Li X, Zhou P, Liu T, Zhang L, Shi K, Yang Q, Dong J, Zhou J. IRF-7 Is a Critical Regulator of Type 2 Innate Lymphoid Cells in Allergic Airway Inflammation. Cell Rep 2020; 29:2718-2730.e6. [PMID: 31775040 DOI: 10.1016/j.celrep.2019.10.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/16/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Allergic asthma is a highly prevalent airway disease triggered by hyperresponsiveness to inhaled allergens. Interferon regulatory factor 7 (IRF7) has been shown to be highly expressed in nasal aspirates from children with asthma. Type 2 innate lymphoid cells (ILC2s) represent the major player in allergic airway inflammation. The role of IRF7 in ILC2-driven asthma remains to be explored. Here, we report that IRF7 expression in murine lung ILC2s is dramatically induced upon papain or interleukin-33 (IL-33) stimulation. ILC2s from asthma patients display a much higher level of IRF7 than those from healthy donors. Deficiency of IRF7 in mice significantly impairs the expansion and function of lung ILC2s in multiple models of allergic asthma. Furthermore, the regulation of ILC2s by IRF7 is cell intrinsic and mediated by the transcription factor Bcl11b. These observations identify IRF7 as a regulator of lung ILC2s, which may have immunotherapeutic value in allergic asthma.
Collapse
Affiliation(s)
- Juan He
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiong Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xiao
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Aihua Lei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pan Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Zhang
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Kun Shi
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Junchao Dong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
83
|
Frech M, Knipfer L, Wirtz S, Zaiss MM. An in vivo gene delivery approach for the isolation of reasonable numbers of type 2 innate lymphoid cells. MethodsX 2020; 7:101054. [PMID: 33005569 PMCID: PMC7509459 DOI: 10.1016/j.mex.2020.101054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/02/2020] [Indexed: 11/30/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are a recently recognized subset of innate lymphocytes with crucial role in mucosal immunity and tissue homeostasis. Over the past decade, substantial advances in our understanding of ILC2 biology have established them as an essential element in innate and adaptive immunity. However, their relatively low abundance and laborious purification from mucosal tissues make their study difficult. Moreover, due to a lack of an ILC2-specific Cre mouse-line, adoptive transfer of ILC2s into ILC-deficient hosts is inevitable. Herein we describe an in-depth protocol for the induction, isolation, and expansion of murine ILC2s. By combining an in vivo gene delivery approach to boost ILC2 numbers and a cell culture strategy to expand isolated cells, large quantities of highly pure ILC2s can be obtained. The isolated cells maintain their phenotype and can be used for subsequent cell transfer or in vitro studies. In comparison to previous protocols, this approach is cost-effective and efficient with potential yield of more than 20 million ILC2s isolated per mouse. • Group 2 innate lymphoid cells (ILC2s) are extensively studied in mouse models and humans in recent years. • Low abundance of ILC2s and current lack of specific ILC2 knockout mice makes in vivo research challenging. • This method allows high and pure ILC2 numbers for in vitro or adoptive in vivo transfer experiments.
Collapse
Affiliation(s)
- Michael Frech
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Germany
| | - Lisa Knipfer
- Department of Internal Medicine 1, University of Erlangen-Nuremberg, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Germany
| | - Stefan Wirtz
- Department of Internal Medicine 1, University of Erlangen-Nuremberg, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 1, University of Erlangen-Nuremberg, Germany
| |
Collapse
|
84
|
Ding F, Liu B, Niu C, Wang T, Wang Y, Geng G, Tian D, Dai J, Fu Z. Low-Dose LPS Induces Tolerogenic Treg Skewing in Asthma. Front Immunol 2020; 11:2150. [PMID: 33072079 PMCID: PMC7538595 DOI: 10.3389/fimmu.2020.02150] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
The mechanism(s) underlying endotoxin tolerance in asthma remain elusive. As the endotoxin lipopolysaccharide (LPS) affects the expression of the regulatory T-cell (Treg)-suppressive glucocorticoid-induced tumor necrosis factor receptor ligand (GITRL) on antigen-presenting dendritic cells (DCs), we hypothesized that LPS-induced changes in DC GITRL expression may impact Treg-mediated T-helper (Th) cell suppression and the induction of endotoxin tolerance. Here, we propose a novel mechanism by which low-dose LPS inhalation in neonatal mice induces endotoxin tolerance, thereby offering protection from later asthma development. Three-day old wild-type and Toll-like receptor 4 (TLR4)-deficient neonatal mice were exposed to low-dose LPS (1 μg) intranasally for 10 consecutive days prior to ovalbumin (OVA)-induced asthma to better understand the tolerogenic mechanism(s) of low-dose LPS pre-exposure. In vivo findings were validated using in vitro co-culturing studies of primary CD11c+ DCs and CD4+ T-cells with or without low-dose LPS pre-exposure before OVA stimulation. Low-dose LPS pre-exposure upregulated the Treg response and downregulated pathogenic Th2 and Th17 responses through promoting apoptosis of Th2 and Th17 cells. Low-dose LPS pre-exposure downregulated DC GITRL expression and T-cell GITR expression. Artificial DC GITRL expression abrogated the tolerogenic Treg-skewing effect of low-dose LPS pre-exposure. Low-dose LPS pre-exposure inhibited TRIF/IRF3/IFNβ signaling and upregulated expression of tolerogenic TRIF/IRF3/IFNβ negative regulators in a TLR4-dependent manner. This tolerogenic DC GITRL downregulation was attributable to TRIF/IRF3/IFNβ signaling inhibition. Low-dose LPS pre-exposure produces tolerogenic Treg skewing in neonatal asthmatic mice, a phenomenon attributable to TLR4-dependent TRIF/IRF3/IFNβ-mediated DC GITRL downregulation.
Collapse
Affiliation(s)
- Fengxia Ding
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Bo Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Niu
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Ting Wang
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Yaping Wang
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Gang Geng
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Daiyin Tian
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Jihong Dai
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Zhou Fu
- Department of Pediatric Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| |
Collapse
|
85
|
IL-33-ST2 axis regulates myeloid cell differentiation and activation enabling effective club cell regeneration. Nat Commun 2020; 11:4786. [PMID: 32963227 PMCID: PMC7508874 DOI: 10.1038/s41467-020-18466-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Evidence points to an indispensable function of macrophages in tissue regeneration, yet the underlying molecular mechanisms remain elusive. Here we demonstrate a protective function for the IL-33-ST2 axis in bronchial epithelial repair, and implicate ST2 in myeloid cell differentiation. ST2 deficiency in mice leads to reduced lung myeloid cell infiltration, abnormal alternatively activated macrophage (AAM) function, and impaired epithelial repair post naphthalene-induced injury. Reconstitution of wild type (WT) AAMs to ST2-deficient mice completely restores bronchial re-epithelialization. Central to this mechanism is the direct effect of IL-33-ST2 signaling on monocyte/macrophage differentiation, self-renewal and repairing ability, as evidenced by the downregulation of key pathways regulating myeloid cell cycle, maturation and regenerative function of the epithelial niche in ST2−/− mice. Thus, the IL-33-ST2 axis controls epithelial niche regeneration by activating a large multi-cellular circuit, including monocyte differentiation into competent repairing AAMs, as well as group-2 innate lymphoid cell (ILC2)-mediated AAM activation. Signaling of IL-33 via its receptor, ST2, has been implicated in macrophage function in tissue repair. Here the authors show, using genetic mouse models and single-cell transcriptomic data, that the IL-33/ST2 axis regulates both ILC2-derived IL-13 and macrophage differentiation/reparative function required for club cell regeneration.
Collapse
|
86
|
Caminati M, Bagnasco D, Rosenwasser LJ, Vianello A, Senna G. Biologics for the Treatments of Allergic Conditions: Severe Asthma. Immunol Allergy Clin North Am 2020; 40:549-564. [PMID: 33012319 DOI: 10.1016/j.iac.2020.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
By selectively targeting specific steps of the immune inflammation cascade, biologic drugs for severe asthma have substantially contributed to increase the standard of care, to reduce drug-related morbidity. and most importantly to ameliorate patients' quality of life. Upcoming molecules are going to provide a chance for severe phenotypes besides Th2 high through the interaction with epithelial and innate immunity. Some practical aspects including optimal treatment duration, the possibility of a dose treatment modulation, the place and relevance of ICS in best responders are still under debate. Long-term safety, especially when interacting with innate immunity needs to be further investigated.
Collapse
Affiliation(s)
- Marco Caminati
- Department of Medicine, Allergy and Clinical Immunology Section, University of Verona and Verona University Hospital, Piazzale Scuro 10, Verona 37134, Italy.
| | - Diego Bagnasco
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa, Largo Rosanna Benzi, 10, Genoa 16132, Italy
| | | | - Andrea Vianello
- Respiratory Pathophysiology Division, University of Padua, Padua, Italy; Dipartimento di Scienze CardioToraco Vascolari e Sanità Pubblica, Via Nicolo` Giustiniani, 2, Padua 35128, Italy
| | - Gianenrico Senna
- Department of Medicine, Allergy and Clinical Immunology School, University of Verona & Asthma Center and Allergy Unit, Verona University Hospital, Piazzale Scuro 10, Verona 37134, Italy
| |
Collapse
|
87
|
Pointner L, Bethanis A, Thaler M, Traidl-Hoffmann C, Gilles S, Ferreira F, Aglas L. Initiating pollen sensitization - complex source, complex mechanisms. Clin Transl Allergy 2020; 10:36. [PMID: 32884636 PMCID: PMC7461309 DOI: 10.1186/s13601-020-00341-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanisms involved in the induction of allergic sensitization by pollen are not fully understood. Within the last few decades, findings from epidemiological and experimental studies support the notion that allergic sensitization is not only dependent on the genetics of the host and environmental factors, but also on intrinsic features of the allergenic source itself. In this review, we summarize the current concepts and newest advances in research focusing on the initial mechanisms inducing pollen sensitization. Pollen allergens are embedded in a complex and heterogeneous matrix composed of a myriad of bioactive molecules that are co-delivered during the allergic sensitization. Surprisingly, several purified allergens were shown to lack inherent sensitizing potential. Thus, growing evidence supports an essential role of pollen-derived components co-delivered with the allergens in the initiation of allergic sensitization. The pollen matrix, which is composed by intrinsic molecules (e.g. proteins, metabolites, lipids, carbohydrates) and extrinsic compounds (e.g. viruses, particles from air pollutants, pollen-linked microbiome), provide a specific context for the allergen and has been proposed as a determinant of Th2 polarization. In addition, the involvement of various pattern recognition receptors (PRRs), secreted alarmins, innate immune cells, and the dependency of DCs in driving pollen-induced Th2 inflammatory processes suggest that allergic sensitization to pollen most likely results from particular combinations of pollen-specific signals rather than from a common determinant of allergenicity. The exact identification and characterization of such pollen-derived Th2-polarizing molecules should provide mechanistic insights into Th2 polarization and pave the way for novel preventive and therapeutic strategies against pollen allergies.
Collapse
Affiliation(s)
- Lisa Pointner
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Athanasios Bethanis
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Michael Thaler
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Claudia Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
- Christine-Kühne-Center for Allergy Research and Education (CK-Care), Davos, Switzerland
| | - Stefanie Gilles
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| |
Collapse
|
88
|
Corren J. New Targeted Therapies for Uncontrolled Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 7:1394-1403. [PMID: 31076057 DOI: 10.1016/j.jaip.2019.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
Mechanistic studies have improved our understanding of molecular and cellular components involved in asthma and our ability to treat severe patients. An mAb directed against IgE (omalizumab) has become an established add-on therapy for patients with uncontrolled allergic asthma and mAbs specific for IL-5 (reslizumab, mepolizumab), IL-5R (benralizumab), and IL-4R (dupilumab) have been approved as add-on treatments for uncontrolled eosinophilic (type 2) asthma. While these medications have proven highly effective, some patients with severe allergic and/or eosinophilic asthma, as well as most patients with severe non-type-2 disease, have poorly controlled disease. Agents that have recently been evaluated in clinical trials include an antibody directed against thymic stromal lymphopoietin, small molecule antagonists to the chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2) and the receptor for stem cell factor on mast cells (KIT), and a DNA enzyme directed at GATA3. Antibodies to IL-33 and its receptor, ST2, are being evaluated in ongoing clinical studies. In addition, a number of antagonists directed against other potential targets are under consideration for future trials, including IL-25, IL-6, TNF-like ligand 1A, CD6, and activated cell adhesion molecule (ALCAM). Clinical data from ongoing and future trials will be important in determining whether these new medications will offer benefits in place of or in addition to existing therapies for asthma.
Collapse
MESH Headings
- Activated-Leukocyte Cell Adhesion Molecule/immunology
- Anti-Asthmatic Agents/therapeutic use
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Asthma/drug therapy
- Asthma/immunology
- Asthma/physiopathology
- Cytokines/antagonists & inhibitors
- Cytokines/immunology
- DNA, Catalytic/therapeutic use
- Eosinophils/immunology
- GATA3 Transcription Factor
- Humans
- Imatinib Mesylate/therapeutic use
- Indoleacetic Acids/therapeutic use
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/immunology
- Interleukin-6/immunology
- Lymphocytes/immunology
- Mast Cells/immunology
- Molecular Targeted Therapy
- Omalizumab/therapeutic use
- Proto-Oncogene Proteins c-kit/antagonists & inhibitors
- Proto-Oncogene Proteins c-kit/immunology
- Pyridines/therapeutic use
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/immunology
- Receptors, Interleukin-17/antagonists & inhibitors
- Receptors, Interleukin-17/immunology
- Receptors, Prostaglandin/antagonists & inhibitors
- Receptors, Prostaglandin/immunology
- Ribonucleases/therapeutic use
- Th2 Cells/immunology
- Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors
- Tumor Necrosis Factor Ligand Superfamily Member 15/immunology
Collapse
Affiliation(s)
- Jonathan Corren
- Departments of Medicine and Pediatrics, Division of Allergy and Clinical Immunology, David Geffen School of Medicine at UCLA, Los Angeles, Calif.
| |
Collapse
|
89
|
Drake LY, Prakash YS. Contributions of IL-33 in Non-hematopoietic Lung Cells to Obstructive Lung Disease. Front Immunol 2020; 11:1798. [PMID: 32903501 PMCID: PMC7438562 DOI: 10.3389/fimmu.2020.01798] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-33 plays important roles in pulmonary immune responses and lung diseases including asthma and chronic obstructive pulmonary disease (COPD). There is substantial interest in identifying and characterizing cellular sources vs. targets of IL-33, and downstream signaling pathways involved in disease pathophysiology. While epithelial and immune cells have largely been the focus, in this review, we summarize current knowledge of expression, induction, and function of IL-33 and its receptor ST2 in non-hematopoietic lung cells in the context of health and disease. Under basal conditions, epithelial cells and endothelial cells are thought to be the primary resident cell types that express high levels of IL-33 and serve as ligand sources compared to mesenchymal cells (smooth muscle cells and fibroblasts). Under inflammatory conditions, IL-33 expression is increased in most non-hematopoietic lung cells, including epithelial, endothelial, and mesenchymal cells. In comparison to its ligand, the receptor ST2 shows low expression levels at baseline but similar to IL-33, ST2 expression is upregulated by inflammation in these non-hematopoietic lung cells which may then participate in chronic inflammation both as sources and autocrine/paracrine targets of IL-33. Downstream effects of IL-33 may occur via direct receptor activation or indirect interactions with the immune system, overall contributing to lung inflammation, airway hyper-responsiveness and remodeling (proliferation and fibrosis). Accordingly from a therapeutic perspective, targeting IL-33 and/or its receptor in non-hematopoietic lung cells becomes relevant.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
90
|
Miller MM, Reinhardt RL. The Heterogeneity, Origins, and Impact of Migratory iILC2 Cells in Anti-helminth Immunity. Front Immunol 2020; 11:1594. [PMID: 32793230 PMCID: PMC7390839 DOI: 10.3389/fimmu.2020.01594] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Soil-transmitted helminths represent a major global health burden with infections and infection-related comorbidities causing significant reductions in the quality of life for individuals living in endemic areas. Repeated infections and chronic colonization by these large extracellular worms in mammals led to the evolution of type-2 immunity characterized by the production of the type-2 cytokines interleukin (IL)-4, IL-5, and IL-13. Although a number of adaptive and innate immune cells produce type-2 cytokines, a key cellular source in the context of helminth infection is group 2 innate lymphoid cells (ILC2s). ILC2s promote mucosal barrier homeostasis, integrity, and repair by rapidly responding to epithelial cues in mucosal tissues. Though tissue-resident ILC2s (nILC2s) have been studied in detail over the last decade, considerably less is known with regard to a subset of inflammatory ILC2s (iILC2s) that migrate to the lungs of mice early after Nippostrongylus brasiliensis infection and are potent early producers of type-2 cytokines. This review will discuss the relationship and differences between nILC2s and iILC2s that establish their unique roles in anti-helminth immunity. We have placed particular emphasis on studies investigating iILC2 origin, function, and their potential long-term contribution to tissue-resident ILC2 reservoirs in settings of helminth infection.
Collapse
Affiliation(s)
- Mindy M Miller
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - R Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States.,Department of Immunology and Microbiology, University of Colorado-Anschutz Medical, Aurora, CO, United States
| |
Collapse
|
91
|
Zhou W, Zhang J, Toki S, Goleniewska K, Norlander AE, Newcomb DC, Wu P, Boyd KL, Kita H, Peebles RS. COX Inhibition Increases Alternaria-Induced Pulmonary Group 2 Innate Lymphoid Cell Responses and IL-33 Release in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 205:1157-1166. [PMID: 32690653 DOI: 10.4049/jimmunol.1901544] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023]
Abstract
The cyclooxygenase (COX) metabolic pathway regulates immune responses and inflammation. The effect of the COX pathway on innate pulmonary inflammation induced by protease-containing fungal allergens, such as Alternaria alternata, is not fully defined. In this study, we tested the hypothesis that COX inhibition augments Alternaria-induced pulmonary group 2 innate lymphoid cell (ILC2) responses and IL-33 release. Mice were treated with the COX inhibitors indomethacin, flurbiprofen, or vehicle and challenged intranasally with Alternaria extract for four consecutive days to induce innate lung inflammation. We found that indomethacin and flurbiprofen significantly increased the numbers of ILC2 and IL-5 and IL-13 expression by ILC2 in the lung. Indomethacin also increased ILC2 proliferation, the percentages of eosinophils, and mucus production in the lung. Both indomethacin and flurbiprofen augmented the release of IL-33 in bronchoalveolar lavage fluid after Alternaria challenge, suggesting that more IL-33 was available for ILC2 activation and that a COX product(s) inhibited IL-33 release. This is supported by the in vitro finding that the COX product PGE2 and the PGI2 analogs cicaprost decreased Alternaria extract-induced IL-33 release by human bronchial epithelial cells. Although contrasting effects of PGD2, PGE2, and PGI2 on ILC2 responses have been previously reported, the overall effect of the COX pathway on ILC2 function is inhibitory in Alternaria-induced innate airway inflammation.
Collapse
Affiliation(s)
- Weisong Zhou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232;
| | - Jian Zhang
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Shinji Toki
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Allison E Norlander
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Dawn C Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Pingsheng Wu
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
| | - R Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232.,Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| |
Collapse
|
92
|
Abstract
Although, as the major organ of gas exchange, the lung is considered a nonlymphoid organ, an interconnected network of lung-resident innate cells, including epithelial cells, dendritic cells, macrophages, and natural killer cells is crucial for its protection. These cells provide defense against a daily assault by airborne bacteria, viruses, and fungi, as well as prevent the development of cancer, allergy, and the outgrowth of commensals. Our understanding of this innate immune environment has recently changed with the discovery of a family of innate lymphoid cells (ILCs): ILC1s, ILC2s, and ILC3s. All lack adaptive antigen receptors but can provide a substantial and rapid source of IFN-γ, IL-5 and IL-13, and IL-17A or IL-22, respectively. Their ability to afford immediate protection to the lung and to influence subsequent adaptive immune responses highlights the importance of understanding ILC-regulated immunity for the design of future therapeutic interventions.
Collapse
Affiliation(s)
- Jillian L Barlow
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| | - Andrew N J McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| |
Collapse
|
93
|
Cui J, Dong M, Yi L, Wei Y, Tang W, Zhu X, Dong J, Wang W. Acupuncture inhibited airway inflammation and group 2 innate lymphoid cells in the lung in an ovalbumin-induced murine asthma model. Acupunct Med 2020; 39:217-225. [PMID: 32539427 DOI: 10.1177/0964528420924033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are known to serve important functions in the pathogenesis of allergic airway inflammation. Studies have shown that acupuncture has an anti-inflammatory effect in the airways. However, how acupuncture treatment affects innate immunity, especially with regard to the function of ILC2s in ovalbumin (OVA)-induced allergic airway inflammation, is poorly understood. METHODS BALB/c mice were injected and subsequently challenged with OVA ± treated with manual acupuncture. At the end of the experimental course, lung function was assessed by measurement of airway resistance (RL) and lung dynamic compliance (Cdyn). Cytokine levels were detected by enzyme-linked immunosorbent assay (ELISA). ILC2 proportions in the lung were analyzed by flow cytometry. RESULTS The results showed that airway inflammation and mucus secretion were significantly suppressed by acupuncture treatment. RL decreased while Cdyn increased after acupuncture treatment. There was an apparent decrease in the bronchoalveolar lavage fluid (BALF) concentrations of interleukin (IL)-5, IL-13, IL-9, IL-25 and IL-33 and an increase in soluble IL-33 receptor (sST2) levels compared with untreated asthmatic mice. Acupuncture also reduced the lin-CD45+KLRG1+ST2+ cell proportion in the lung. CONCLUSION In conclusion, this study has demonstrated that acupuncture treatment alleviates allergic airway inflammation and inhibits pulmonary ILC2 influx and IL-5, IL-9 and IL-13 production. The inhibition of ILC2s by acupuncture may be associated with the IL-33/ST2-signaling pathway and IL-25 levels, thereby offering protection from the respiratory inflammation associated with asthma.
Collapse
Affiliation(s)
- Jie Cui
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ming Dong
- Gumei Community Health Center of Minhang District of Shanghai, Shanghai, China
| | - La Yi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wenqian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
94
|
Yasuda Y, Nagano T, Kobayashi K, Nishimura Y. Group 2 Innate Lymphoid Cells and the House Dust Mite-Induced Asthma Mouse Model. Cells 2020; 9:E1178. [PMID: 32397396 PMCID: PMC7290734 DOI: 10.3390/cells9051178] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/03/2020] [Accepted: 05/08/2020] [Indexed: 12/21/2022] Open
Abstract
Asthma is an important issue not only in health but also in economics worldwide. Therefore, asthma animal models have been frequently used to understand the pathogenesis of asthma. Recently, in addition to acquired immunity, innate immunity has also been thought to be involved in asthma. Among innate immune cells, group 2 innate lymphoid cells (ILC2s) have been considered to be crucial for eosinophilic airway inflammation by releasing T helper 2 cytokines. Moreover, house dust mites (HDMs) belonging to group 1 act on airway epithelial cells not only as allergens but also as cysteine proteases. The production of interleukin-25 (IL-25), IL-33, and thymic stromal lymphopoietin (TSLP) from airway epithelial cells was induced by the protease activity of HDMs. These cytokines activate ILC2s, and activated ILC2s produce IL-5, IL-9, IL-13, and amphiregulin. Hence, the HDM-induced asthma mouse model greatly contributes to understanding asthma pathogenesis. In this review, we highlight the relationship between ILC2s and the HDM in the asthma mouse model to help researchers and clinicians not only choose a proper asthma mouse model but also to understand the molecular mechanisms underlying HDM-induced asthma.
Collapse
Affiliation(s)
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Kobe, Hyogo 650-0017, Japan; (Y.Y.); (K.K.); (Y.N.)
| | | | | |
Collapse
|
95
|
Kasahara DI, Shore SA. IL-33, diet-induced obesity, and pulmonary responses to ozone. Respir Res 2020; 21:98. [PMID: 32326950 PMCID: PMC7181525 DOI: 10.1186/s12931-020-01361-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/14/2020] [Indexed: 12/21/2022] Open
Abstract
Background Obesity augments pulmonary responses to ozone. We have reported that IL-33 contributes to these effects of obesity in db/db mice. The purpose of this study was to determine whether IL-33 also contributes to obesity-related changes in the response to ozone in mice with diet-induced obesity. Methods Male wildtype C57BL/6 mice and mice deficient in ST2, the IL-33 receptor, were placed on chow or high fat diets for 12 weeks from weaning. Because the microbiome has been implicated in obesity-related changes in the pulmonary response to ozone, mice were either housed with other mice of the same genotype (same housed) or with mice of the opposite genotype (cohoused). Cohousing transfers the gut microbiome from one mouse to its cagemates. Results Diet-induced increases in body mass were not affected by ST2 deficiency or cohousing. In same housed mice, ST2 deficiency reduced ozone-induced airway hyperresponsiveness and neutrophil recruitment in chow-fed but not HFD-fed mice even though ST2 deficiency reduced bronchoalveolar lavage IL-5 in both diet groups. In chow-fed mice, cohousing abolished ST2-related reductions in ozone-induced airway hyperresponsiveness and neutrophil recruitment, but in HFD-fed mice, no effect of cohousing on these responses to ozone was observed. In chow-fed mice, ST2 deficiency and cohousing caused changes in the gut microbiome. High fat diet-feeding caused marked changes in the gut microbiome and overrode both ST2-related and cohousing-related differences in the gut microbiome observed in chow-fed mice. Conclusion Our data indicate a role for IL-33 in pulmonary responses to ozone in chow-fed but not high fat diet-fed mice and are consistent with the hypothesis that these diet-related differences in the role of IL-33 are the result of changes in the gut microbiome.
Collapse
Affiliation(s)
- David I Kasahara
- Department of Environmental Health, Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115-6021, USA
| | - Stephanie A Shore
- Department of Environmental Health, Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115-6021, USA.
| |
Collapse
|
96
|
Tay HL, Foster PS. Biologics or immunotherapeutics for asthma? Pharmacol Res 2020; 158:104782. [PMID: 32275962 DOI: 10.1016/j.phrs.2020.104782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 01/06/2023]
Abstract
Asthma is now recognised as a heterogenous inflammatory disease of the lung based on cellular infiltrates and transcriptional profiles of blood and airway cells. Four distinct subgroups have been defined, eosinophilic (T2), neutrophilic (T1), mixed eosinophilic/neutrophilic and paucigranulocytic. Patients can also be stratified at a molecular level into T2-high, T2-low and/or T1 based on their gene signatures. Current treatments for asthma have been centred on administration of steroids and/or bronchodilators for the relief of bronchoconstriction and inflammation. These treatments are not always effective and often have limited efficacy during exacerbations. Eosinophil expansion and homing to tissues, bronchoconstriction, IgE production and mucus hypersecretion (hallmark features of asthma) are regulated by the type 2 cytokines IL-4, IL-5 and IL-13, the latter of which can induce the expression of the eosinophil chemotactic factors CCL11 and CCL24. A number of new generation biologics (monoclonal antibodies) targeting pathways regulated by the T2 cytokines IL-5 and IL-4/13 (IL-4 receptor alpha) have yielded effective therapies for eosinophil induced exacerbations of severe asthma. Despite these advances, difficulties still remain in treating all exacerbations, and this may reflect the contribution of other inflammatory cells such as neutrophils to pathogenesis. This review describes the effectiveness of targeting T2 pathways, emerging approaches and identifies the potential next steps for therapeutic intervention.
Collapse
Affiliation(s)
- Hock L Tay
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Hunter Medical Research Institute, Australia.
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Hunter Medical Research Institute, Australia.
| |
Collapse
|
97
|
Zhou S, Li Q, Wu H, Lu Q. The pathogenic role of innate lymphoid cells in autoimmune-related and inflammatory skin diseases. Cell Mol Immunol 2020; 17:335-346. [PMID: 32203190 PMCID: PMC7109064 DOI: 10.1038/s41423-020-0399-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/27/2020] [Indexed: 12/31/2022] Open
Abstract
Innate lymphoid cells (ILCs), as an important component of the innate immune system, arise from a common lymphoid progenitor and are located in mucosal barriers and various tissues, including the intestine, skin, lung, and adipose tissue. ILCs are heterogeneous subsets of lymphocytes that have emerging roles in orchestrating immune response and contribute to maintain metabolic homeostasis and regulate tissue inflammation. Currently, more details about the pathways for the development and differentiation of ILCs have largely been elucidated, and cytokine secretion and downstream immune cell responses in disease pathogenesis have been reported. Recent research has identified that several distinct subsets of ILCs at skin barriers are involved in the complex regulatory network in local immunity, potentiating adaptive immunity and the inflammatory response. Of note, additional studies that assess the effects of ILCs are required to better define how ILCs regulate their development and functions and how they interact with other immune cells in autoimmune-related and inflammatory skin disorders. In this review, we will distill recent research progress in ILC biology, abnormal functions and potential pathogenic mechanisms in autoimmune-related skin diseases, including systemic lupus erythematosus (SLE), scleroderma and inflammatory diseases, as well as psoriasis and atopic dermatitis (AD), thereby giving a comprehensive review of the diversity and plasticity of ILCs and their unique functions in disease conditions with the aim to provide new insights into molecular diagnosis and suggest potential value in immunotherapy.
Collapse
Affiliation(s)
- Suqing Zhou
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Qianwen Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| |
Collapse
|
98
|
Magat JM, Thomas JL, Dumouchel JP, Murray F, Li WX, Li J. Endogenous IL-33 and Its Autoamplification of IL-33/ST2 Pathway Play an Important Role in Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1592-1597. [PMID: 31988179 PMCID: PMC7065953 DOI: 10.4049/jimmunol.1900690] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022]
Abstract
IL-33 and its receptor ST2 are contributing factors to airway inflammation and asthma exacerbation. The IL-33/ST2 signaling pathway is involved in both the onset and the acute exacerbations of asthma. In this study, we address the role of endogenous IL-33 and its autoamplification of the IL-33/ST2 pathway in Ag-dependent and Ag-independent asthma-like models. Wild-type, IL-33 knockout, ST2 knockout mice were either intratracheally administrated with 500 ng of rIL-33 per day for four consecutive days or were sensitized and challenged with OVA over 21 d. In wild-type mice, IL-33 or OVA induced similar airway hyperresponsiveness and eosinophilic airway inflammation. IL-33 induced its own mRNA and ST2L mRNA expression in the lung. IL-33 autoamplified itself and ST2 protein expression in airway epithelial cells. OVA also induced IL-33 and ST2 protein expression. In IL-33 knockout mice, the IL-33- and OVA-induced airway hyperresponsiveness and eosinophilic airway inflammation were both significantly attenuated, whereas IL-33-induced ST2L mRNA expression was preserved, although no autoamplification of IL-33/ST2 pathway was observed. In ST2 knockout mice, IL-33 and OVA induced airway hyperresponsiveness and eosinophilic airway inflammation were both completely diminished, and no IL-33/ST2 autoamplification was observed. These results suggest that endogenous IL-33 and its autoamplification of IL-33/ST2 pathway play an important role in the induction of asthma-like phenotype. Thus an intact IL-33/ST2 pathway is necessary for both Ag-dependent and Ag-independent asthma-like mouse models.
Collapse
Affiliation(s)
- Jenna M Magat
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Joanna L Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92093
| | - Justin P Dumouchel
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Fiona Murray
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Willis X Li
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Jinghong Li
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| |
Collapse
|
99
|
Tuft-Cell-Derived Leukotrienes Drive Rapid Anti-helminth Immunity in the Small Intestine but Are Dispensable for Anti-protist Immunity. Immunity 2020; 52:528-541.e7. [PMID: 32160525 DOI: 10.1016/j.immuni.2020.02.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/08/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Helminths, allergens, and certain protists induce type 2 immune responses, but the underlying mechanisms of immune activation remain poorly understood. In the small intestine, chemosensing by epithelial tuft cells results in the activation of group 2 innate lymphoid cells (ILC2s), which subsequently drive increased tuft cell frequency. This feedforward circuit is essential for intestinal remodeling and helminth clearance. ILC2 activation requires tuft-cell-derived interleukin-25 (IL-25), but whether additional signals regulate the circuit is unclear. Here, we show that tuft cells secrete cysteinyl leukotrienes (cysLTs) to rapidly activate type 2 immunity following chemosensing of helminth infection. CysLTs cooperate with IL-25 to activate ILC2s, and tuft-cell-specific ablation of leukotriene synthesis attenuates type 2 immunity and delays helminth clearance. Conversely, cysLTs are dispensable for the tuft cell response induced by intestinal protists. Our findings identify an additional tuft cell effector function and suggest context-specific regulation of tuft-ILC2 circuits within the small intestine.
Collapse
|
100
|
Mehrabi Nasaba E, Athari S, Motlagh B, Athari S. Effects of oral administration of Ocimum basilicum on goblet cell hyperplasia and upstream cytokine gene expression in allergic asthma. REVUE FRANCAISE D ALLERGOLOGIE 2020. [DOI: 10.1016/j.reval.2019.02.226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|