1
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. Targeting γc family cytokines with biologics: current status and future prospects. MAbs 2025; 17:2468312. [PMID: 39967341 PMCID: PMC11845063 DOI: 10.1080/19420862.2025.2468312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Over the recent decades the market potential of biologics has substantially expanded, and many of the top-selling drugs worldwide are now monoclonal antibodies or antibody-like molecules. The common gamma chain (γc) cytokines, Interleukin (IL-)2, IL-4, IL-7, IL-9, IL-15, and IL-21, play pivotal roles in regulating immune responses, from innate to adaptive immunity. Dysregulation of cell signaling by these cytokines is strongly associated with a range of immunological disorders, which includes cancer as well as autoimmune and inflammatory diseases. Given the essential role of γc cytokines in maintaining immune homeostasis, the development of therapeutic interventions targeting these molecules poses unique challenges. Here, we provide an overview of current biologics targeting either single or multiple γc cytokines or their respective receptor subunits across a spectrum of diseases, primarily focusing on antibodies, antibody-like constructs, and antibody-cytokine fusions. We summarize therapeutic biologics currently in clinical trials, highlighting how they may offer advantages over existing therapies and standard of care, and discuss recent advances in this field. Finally, we explore future directions and the potential of novel therapeutic intervention strategies targeting this cytokine family.
Collapse
Affiliation(s)
- Fabian Bick
- Argenx BV, Zwijnaarde, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J. Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
2
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. A reappraisal of IL-9 in inflammation and cancer. Mucosal Immunol 2025; 18:1-15. [PMID: 39389468 DOI: 10.1016/j.mucimm.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
While much is known about the functional effects of type 2 cytokines interleukin (IL)-4, IL-5 and IL-13 in homeostasis and disease, we still poorly understand the functions of IL-9. Chronic inflammation seen in allergic diseases, autoimmunity and cancer is however frequently accompanied by overproduction of this elusive type 2 cytokine. Initially identified as a T cell and mast cell growth factor, and later as the hallmark cytokine defining TH9 cells, we now know that IL-9 is produced by multiple innate and adaptive immune cells. Recent evidence suggests that IL-9 controls discrete aspects of the allergic cascade, cellular responses of immune and stromal cells, cancer progression, tolerance and immune escape. Despite functioning as a pleiotropic cytokine in mucosal environments, like the lungs, the direct and indirect cellular targets of IL-9 are still not well characterized. Here, we discuss IL-9's cellular senders and receivers, focusing on asthma and cancer. Moreover, we review current research directions and the outlook of targeted therapy centered around the biology of IL-9.
Collapse
Affiliation(s)
- Fabian Bick
- argenx BV, 9052 Zwijnaarde, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
3
|
Bick F, Brenis Gómez CM, Lammens I, Van Moorleghem J, De Wolf C, Dupont S, Dumoutier L, Smith NP, Villani AC, Browaeys R, Alladina J, Haring AM, Medoff BD, Cho JL, Bigirimana R, Vieira J, Hammad H, Blanchetot C, Schuijs MJ, Lambrecht BN. IL-2 family cytokines IL-9 and IL-21 differentially regulate innate and adaptive type 2 immunity in asthma. J Allergy Clin Immunol 2024; 154:1129-1145. [PMID: 39147327 DOI: 10.1016/j.jaci.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/07/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Asthma is often accompanied by type 2 immunity rich in IL-4, IL-5, and IL-13 cytokines produced by TH2 lymphocytes or type 2 innate lymphoid cells (ILC2s). IL-2 family cytokines play a key role in the differentiation, homeostasis, and effector function of innate and adaptive lymphocytes. OBJECTIVE IL-9 and IL-21 boost activation and proliferation of TH2 and ILC2s, but the relative importance and potential synergism between these γ common chain cytokines are currently unknown. METHODS Using newly generated antibodies, we inhibited IL-9 and IL-21 alone or in combination in various murine models of asthma. In a translational approach using segmental allergen challenge, we recently described elevated IL-9 levels in human subjects with allergic asthma compared with nonasthmatic controls. Here, we also measured IL-21 in both groups. RESULTS IL-9 played a central role in controlling innate IL-33-induced lung inflammation by promoting proliferation and activation of ILC2s in an IL-21-independent manner. Conversely, chronic house dust mite-induced airway inflammation, mainly driven by adaptive immunity, was solely dependent on IL-21, which controlled TH2 activation, eosinophilia, total serum IgE, and formation of tertiary lymphoid structures. In a model of innate on adaptive immunity driven by papain allergen, a clear synergy was found between both pathways, as combined anti-IL-9 or anti-IL-21 blockade was superior in reducing key asthma features. In human bronchoalveolar lavage samples we measured elevated IL-21 protein within the allergic asthmatic group compared with the allergic control group. We also found increased IL21R transcripts and predicted IL-21 ligand activity in various disease-associated cell subsets. CONCLUSIONS IL-9 and IL-21 play important and nonredundant roles in allergic asthma by boosting ILC2s and TH2 cells, revealing a dual IL-9 and IL-21 targeting strategy as a new and testable approach.
Collapse
Affiliation(s)
- Fabian Bick
- argenx BV, Zwijnaarde, Belgium; Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Claudia M Brenis Gómez
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Inés Lammens
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Justine Van Moorleghem
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Caroline De Wolf
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sam Dupont
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Laure Dumoutier
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Neal P Smith
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass; Massachusetts General Hospital Cancer Center, Boston, Mass
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass; Massachusetts General Hospital Cancer Center, Boston, Mass
| | - Robin Browaeys
- Bioinformatics Expertise Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Jehan Alladina
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Alexis M Haring
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Benjamin D Medoff
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Josalyn L Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | | | | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | | | - Martijn J Schuijs
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium.
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Ji Y, Wang E, Mohammed MT, Hameed N, Christodoulou MI, Liu X, Zhou W, Fang Z, Jia N, Yu H, Zhou Z, Sun Y, Huang SK, McSharry C, Zhong NS, Xiao X, Li J, Xu D. Selective production of IL-33-neutralizing autoantibody ameliorates asthma responses and severity. Clin Immunol 2024; 264:110234. [PMID: 38740111 DOI: 10.1016/j.clim.2024.110234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Natural anti-cytokine autoantibodies can regulate homeostasis of infectious and inflammatory diseases. The anti-cytokine autoantibody profile and relevance to the pathogenesis of asthma are unknown. We aim to identify key anti-cytokine autoantibodies in asthma patients, and reveal their immunological function and clinical significance. METHODS A Luciferase Immunoprecipitation System was used to screen serum autoantibodies against 11 key cytokines in patients with allergic asthma and healthy donors. The antigen-specificity, immunomodulatory functions and clinical significance of anti-cytokine autoantibodies were determined by ELISA, qPCR, neutralization assays and statistical analysis, respectively. Potential conditions for autoantibody induction were revealed by in vitro immunization. RESULTS Of 11 cytokines tested, only anti-IL-33 autoantibody was significantly increased in asthma, compare to healthy controls, and the proportion positive was higher in patients with mild-to-moderate than severe allergic asthma. In allergic asthma patients, the anti-IL-33 autoantibody level correlated negatively with serum concentration of pathogenic cytokines (e.g., IL-4, IL-13, IL-25 and IL-33), IgE, and blood eosinophil count, but positively with mid-expiratory flow FEF25-75%. The autoantibodies were predominantly IgG isotype, polyclonal and could neutralize IL-33-induced pathogenic responses in vitro and in vivo. The induction of the anti-IL-33 autoantibody in blood B-cells in vitro required peptide IL-33 antigen along with a stimulation cocktail of TLR9 agonist and cytokines IL-2, IL-4 or IL-21. CONCLUSIONS Serum natural anti-IL-33 autoantibodies are selectively induced in some asthma patients. They ameliorate key asthma inflammatory responses, and may improve lung function of allergic asthma.
Collapse
Affiliation(s)
- Yuan Ji
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen 518060, China
| | - Eryi Wang
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mohammed T Mohammed
- Faculty of Veterinary medicine, University of Kufa, Kufa, Iraq; School of Infection and Immunity, 120 University Place, University of Glasgow, Glasgow, UK
| | - Najwa Hameed
- School of Infection and Immunity, 120 University Place, University of Glasgow, Glasgow, UK
| | - Maria-Ioanna Christodoulou
- School of Infection and Immunity, 120 University Place, University of Glasgow, Glasgow, UK; Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Xiaoyu Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China
| | - Wei Zhou
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China
| | - Zhangfu Fang
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China
| | - Nan Jia
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangdong, China
| | - Haiqiong Yu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China; Department of Respiratory and Critical Care Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhenwen Zhou
- Clinical Laboratory, Longgang District Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Shau-Ku Huang
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; National Institute of Environmental Health Sciences, National Health Research Institutes, Taipei, Taiwan, China; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles McSharry
- School of Infection and Immunity, 120 University Place, University of Glasgow, Glasgow, UK
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangdong, China
| | - Xiaojun Xiao
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China.
| | - Jing Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangdong, China.
| | - Damo Xu
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China; State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University, Shenzhen, China.
| |
Collapse
|
5
|
Saadh MJ, Alfattah MA, Ismail AH, Saeed BA, Abbas HH, Elashmawy NF, Hashim GA, Ismail KS, Abo-Zaid MA, Waggiallah HA. The role of Interleukin-21 (IL-21) in allergic disorders: Biological insights and regulatory mechanisms. Int Immunopharmacol 2024; 134:111825. [PMID: 38723368 DOI: 10.1016/j.intimp.2024.111825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 06/03/2024]
Abstract
In recent decades, allergic diseases subsequent from an IgE-mediated response to specific allergens have become a progressively public chronic disease worldwide. They have shaped an important medical and socio-economic burden. A significant proportion of allergic disorders are branded via a form 2 immune response relating Th2 cells, type 2 natural lymphoid cells, mast cells and eosinophils. Interleukin-21 (IL-21) is a participant of the type-I cytokine family manufactured through numerous subsets of stimulated CD4+ T cells and uses controlling properties on a diversity of immune cells. Increasingly, experimental sign suggests a character for IL-21 in the pathogenesis of numerous allergic disorders. The purpose of this review is to discuss the biological properties of IL-21 and to summaries current developments in its role in the regulation of allergic disorders.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Mohammed A Alfattah
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Bashar Abdullah Saeed
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | | | - Nabila F Elashmawy
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ghassan A Hashim
- Department of Nursing, Al-Zahrawi University College, Karbala, Iraq
| | - Khatib Sayeed Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
6
|
Elkoshi Z. TGF-β, IL-1β, IL-6 levels and TGF-β/Smad pathway reactivity regulate the link between allergic diseases, cancer risk, and metabolic dysregulations. Front Immunol 2024; 15:1371753. [PMID: 38629073 PMCID: PMC11019030 DOI: 10.3389/fimmu.2024.1371753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
The risk of cancer is higher in patients with asthma compared to those with allergic rhinitis for many types of cancer, except for certain cancers where a contrasting pattern is observed. This study offers a potential explanation for these observations, proposing that the premalignant levels of circulating transforming growth factor-β (TGF-β), IL-1β, and IL-6 as well as the reactivity of the TGF-β/Smad signaling pathway at the specific cancer site, are crucial factors contributing to the observed disparities. Circulating TGF-β, IL- β and IL-6 levels also help clarify why asthma is positively associated with obesity, Type 2 diabetes, hypertension, and insulin resistance, whereas allergic rhinitis is negatively linked to these conditions. Furthermore, TGF-β/Smad pathway reactivity explains the dual impact of obesity, increasing the risk of certain types of cancer while offering protection against other types of cancer. It is suggested that the association of asthma with cancer and metabolic dysregulations is primarily linked to the subtype of neutrophilic asthma. A binary classification of TGF-β activity as either high (in the presence of IL-1β and IL-6) or low (in the presence or absence of IL-1β and IL-6) is proposed to differentiate between allergy patients prone to cancer and metabolic dysregulations and those less prone. Glycolysis and oxidative phosphorylation, the two major metabolic pathways utilized by cells for energy exploitation, potentially underlie this dichotomous classification by reprogramming metabolic pathways in immune cells.
Collapse
Affiliation(s)
- Zeev Elkoshi
- Research and Development Department, Taro Pharmaceutical Industries Ltd, Haifa, Israel
| |
Collapse
|
7
|
Xie X, Li Y, Yan B, Peng Q, Yao R, Deng Q, Li J, Wu Y, Chen S, Yang X, Ma P. Mediation of the JNC/ILC2 pathway in DBP-exacerbated allergic asthma: A molecular toxicological study on neuroimmune positive feedback mechanism. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133360. [PMID: 38157815 DOI: 10.1016/j.jhazmat.2023.133360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Dibutyl phthalate (DBP), a commonly used plasticizer, has been found to be strongly linked to a consistently high prevalence of allergic diseases, particularly allergic asthma. Previous animal experiments have demonstrated that exposure to DBP can worsen asthma by triggering the production of calcitonin gene-related peptide (CGRP), a neuropeptide in the lung tissue. However, the precise neuroimmune mechanism and pathophysiology of DBP-exacerbated allergic asthma with the assistance of CGRP remain unclear. OBJECTIVE The present study was to investigate the potential pathophysiological mechanism in DBP-exacerbated asthma from the perspective of neural-immune interactions. METHODS AND RESULTS C57BL/6 mice were orally exposed to different concentrations (0.4, 4, 40 mg/kg) of DBP for 28 days. They were then sensitized with OVA and nebulized with OVA for 7 consecutive excitations. To investigate whether DBP exacerbates allergic asthma in OVA induced mice, we analyzed airway hyperresponsiveness and lung histopathology. To investigate the activation of JNC and TRPV1 neurons and the release of CGRP by JNC cells, we measured the levels of TRPV1 channels, calcium inward flow, and downstream neuropeptide CGRP. Results showed that TRPV1 expression, inward calcium flux, and CGRP levels were significantly elevated in the lung tissues of the 40DBP + OVA group, suggesting the release of CGRP by JNC cells. To counteract the detrimental effects of DBP mediated by CGRP, we employed olcegepant (also known as BIBN-4096), a CGRP receptor specific antagonist. Results revealed that 40DBP + OVA + olcegepant led to notable decreases in TRPV1, calcium inward flow, and CGRP expression in lung tissues compare with 40DBP + OVA, further supporting the efficacy of olcegepant. Additionally, we also conducted ILC2 flow sorting and observed that neuropeptide CGRP-activated ILC2 cells have a crucial role as key effector cells in DBP-induced neuroimmune positive feedback regulation. Finally, we examined the protein expression of CGRP, GATA3 and P-GATA3, and found that significant upregulations of CGRP and P-GATA3 in the 40DBP + OVA group, suggest that GATA3 acted as a key regulator of CGRP-activated ILC2. CONCLUSION The aforementioned studies indicate that exposure to DBP can exacerbate allergic asthma, leading to airway inflammation. This exacerbation occurs through the activation of TRPV1 in JNC, resulting in the release of CGRP. The excessive release of CGRP further promotes the release of Th2 cytokines by inducing the activation of ILC2 through GATA phosphorylation. Consequently, this process contributes to the development of airway inflammation and allergic asthma. The increased production of Th2 cytokines also triggers the production of IgE, which interacts with FcεRI on JNC neurons, thereby mediating neuro-immune positive feedback regulation.
Collapse
Affiliation(s)
- Xiaomin Xie
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Yan Li
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Department of Pharmacy, Ezhou Central Hospital, Ezhou 436000, China
| | - Biao Yan
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Qi Peng
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Runming Yao
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Qihong Deng
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jinquan Li
- Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yang Wu
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Shaohui Chen
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xu Yang
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Ping Ma
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China.
| |
Collapse
|
8
|
Song G, Sun M, Zhang Y, Zhang B, Peng M, Bao B. Anti-inflammation of LZTFL1 knockdown in OVA-induced asthmatic mice: Through ERK/GATA3 signaling pathway. Mol Immunol 2024; 167:16-24. [PMID: 38310669 DOI: 10.1016/j.molimm.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/29/2023] [Accepted: 01/18/2024] [Indexed: 02/06/2024]
Abstract
Asthma is a common chronic respiratory disease characterized by Th2-type inflammation in the airways. Leucine zip transcription factor-like 1 (LZTFL1) has been implicated in the regulation of Th2-related factors. The knockdown of LZTFL1 resulted in decreased levels of IL-4, IL-5, and IL-13. We hypothesize that LZTFL1 may have an effect on asthma. We established an acute asthmatic mouse model using the Ovalbumin (OVA) sensitization, and we found that LZTFL1 expression was upregulated in OVA-induced CD4 + T cells. Mice challenged with OVA were administered 5 × 107 TU of lentivirus via tail vein injection. LZTFL1 knockdown reversed the frequency of sneezing and nose rubbing in OVA mice. LZTFL1 knockdown reduced inflammatory cell infiltration, reduced goblet cell numbers, and mitigated collagen deposition in lung tissue. LZTFL1 knockdown decreased the levels of OVA-specific IgE, IL-4, IL-5, and IL-13 in alveolar lavage fluid of asthmatic mice. Furthermore, LZTFL1 knockdown inhibited the aberrant activation of MEK/ERK signaling pathway in asthmatic mice. GATA binding protein 3 (GATA3) is an essential transcription factor in Th2 differentiation. Flow cytometry results revealed that LZTFL1 knockdown reduced the number of GATA3 + CD4 + Th2 cells, while it did not affect the stability of GATA3 mRNA. This may be attributed to ERK signaling which stabilized GATA3 by preventing its ubiquitination and subsequent degradation. In conclusion, LZTFL1 knockdown attenuates inflammation and pathological changes in OVA-induced asthmatic mice through ERK/GATA3 signaling pathway.
Collapse
Affiliation(s)
- Guihua Song
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Mengmeng Sun
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Bingxue Zhang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Minghao Peng
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Beibei Bao
- Department of Pediatrics, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Tee JH, Vijayakumar U, Shanmugasundaram M, Lam TYW, Liao W, Yang Y, Wong WSF, Ge R. Isthmin-1 attenuates allergic Asthma by stimulating adiponectin expression and alveolar macrophage efferocytosis in mice. Respir Res 2023; 24:269. [PMID: 37932719 PMCID: PMC10626717 DOI: 10.1186/s12931-023-02569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Allergic asthma is a common respiratory disease that significantly impacts human health. Through in silico analysis of human lung RNASeq, we found that asthmatic lungs display lower levels of Isthmin-1 (ISM1) expression than healthy lungs. ISM1 is an endogenous anti-inflammatory protein that is highly expressed in mouse lungs and bronchial epithelial cells, playing a crucial role in maintaining lung homeostasis. However, how ISM1 influences asthma remains unclear. This study aims to investigate the potential involvement of ISM1 in allergic airway inflammation and uncover the underlying mechanisms. METHODS We investigated the pivotal role of ISM1 in airway inflammation using an ISM1 knockout mouse line (ISM1-/-) and challenged them with house dust mite (HDM) extract to induce allergic-like airway/lung inflammation. To examine the impact of ISM1 deficiency, we analyzed the infiltration of immune cells into the lungs and cytokine levels in bronchoalveolar lavage fluid (BALF) using flow cytometry and multiplex ELISA, respectively. Furthermore, we examined the therapeutic potential of ISM1 by administering recombinant ISM1 (rISM1) via the intratracheal route to rescue the effects of ISM1 reduction in HDM-challenged mice. RNA-Seq, western blot, and fluorescence microscopy techniques were subsequently used to elucidate the underlying mechanisms. RESULTS ISM1-/- mice showed a pronounced worsening of allergic airway inflammation and hyperresponsiveness upon HDM challenge. The heightened inflammation in ISM1-/- mice correlated with enhanced lung cell necroptosis, as indicated by higher pMLKL expression. Intratracheal delivery of rISM1 significantly reduced the number of eosinophils in BALF and goblet cell hyperplasia. Mechanistically, ISM1 stimulates adiponectin secretion by type 2 alveolar epithelial cells partially through the GRP78 receptor and enhances adiponectin-facilitated apoptotic cell clearance via alveolar macrophage efferocytosis. Reduced adiponectin expression under ISM1 deficiency also contributed to intensified necroptosis, prolonged inflammation, and heightened severity of airway hyperresponsiveness. CONCLUSIONS This study revealed for the first time that ISM1 functions to restrain airway hyperresponsiveness to HDM-triggered allergic-like airway/lung inflammation in mice, consistent with its persistent downregulation in human asthma. Direct administration of rISM1 into the airway alleviates airway inflammation and promotes immune cell clearance, likely by stimulating airway adiponectin production. These findings suggest that ISM1 has therapeutic potential for allergic asthma.
Collapse
Affiliation(s)
- Jong Huat Tee
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Udhaya Vijayakumar
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Mahalakshmi Shanmugasundaram
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Terence Y W Lam
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, A*STAR, Singapore, 138668, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore, 138602, Singapore.
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117600, Singapore.
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
10
|
Mary R, Chalmin F, Accogli T, Bruchard M, Hibos C, Melin J, Truntzer C, Limagne E, Derangère V, Thibaudin M, Humblin E, Boidot R, Chevrier S, Arnould L, Richard C, Klopfenstein Q, Bernard A, Urade Y, Harker JA, Apetoh L, Ghiringhelli F, Végran F. Hematopoietic Prostaglandin D2 Synthase Controls Tfh/Th2 Communication and Limits Tfh Antitumor Effects. Cancer Immunol Res 2022; 10:900-916. [PMID: 35612500 DOI: 10.1158/2326-6066.cir-21-0568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/06/2021] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
T follicular helper (Tfh) cells are a subset of CD4+ T cells essential in immunity and have a role in helping B cells produce antibodies against pathogens. However, their role during cancer progression remains unknown. The mechanism of action of Tfh cells remains elusive because contradictory data have been reported on their protumor or antitumor responses in human and murine tumors. Like Tfh cells, Th2 cells are also involved in humoral immunity and are regularly associated with tumor progression and poor prognosis, mainly through their secretion of IL4. Here, we showed that Tfh cells expressed hematopoietic prostaglandin D2 (PGD2) synthase in a pSTAT1/pSTAT3-dependent manner. Tfh cells produced PGD2, which led to recruitment of Th2 cells via the PGD2 receptor chemoattractant receptor homologous molecule expressed on Th type 2 cells (CRTH2) and increased their effector functions. This cross-talk between Tfh and Th2 cells promoted IL4-dependent tumor growth. Correlation between Th2 cells, Tfh cells, and hematopoietic PGD2 synthase was observed in different human cancers and associated with outcome. This study provides evidence that Tfh/Th2 cross-talk through PGD2 limits the antitumor effects of Tfh cells and, therefore, could serve as a therapeutic target.
Collapse
Affiliation(s)
- Romain Mary
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Fanny Chalmin
- CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Théo Accogli
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Mélanie Bruchard
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Christophe Hibos
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Joséphine Melin
- LipSTIC LabEx, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | | | | | - Valentin Derangère
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | | | - Etienne Humblin
- CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,Precision Immunology Institute, New York, New York
| | - Romain Boidot
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | | | | | - Corentin Richard
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | | | - Antoine Bernard
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Yoshihiro Urade
- Intemational Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - James A Harker
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Lionel Apetoh
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France
| | - François Ghiringhelli
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Frédérique Végran
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France.,CRI INSERM UMR1231 "Lipids, Nutrition and Cancer", Dijon, France.,LipSTIC LabEx, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
11
|
Li Y, Chen Q, Ji W, Fan Y, Huang L, Chu C, Zhou W. TLR2 deficiency promotes IgE and inhibits IgG1 class-switching following ovalbumin sensitization. Ital J Pediatr 2021; 47:162. [PMID: 34315511 PMCID: PMC8314519 DOI: 10.1186/s13052-021-01088-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To explore the roles of Toll-like receptor (TLR)2 in Th2 cytokine production and immunoglobulin (Ig) class switching following ovalbumin (OVA) sensitization. METHODS TLR2-/- and wild-type C57BL/6 mice were sensitized by intraperitoneal injection with OVA. Lung pathology was assessed by hematoxylin and eosin staining. Abundance of interleukin (IL)4, IL5, IL13, and IL21 transcripts in the lungs was quantified by RT-PCR. OVA-specific IgG1, IgG2a, IgG2b, IgE and IgM were quantified by enzyme-linked immunosorbent assay. Phosphorylated signal transducer and activator of transcription (STAT)3 in lung tissue was detected by immunohistochemistry staining and nuclear factor (NF) κB activation was measured by immunofluorescence staining. STAT3 activation was inhibited using cryptotanshinone (CPT) treatment. Germline transcripts (Iμ-Cμ, Iγ-Cγ, Iα-Cα or Iε-Cε), post-recombination transcripts (Iμ-Cγ, Iμ-Cα or Iμ- Cε) and mature transcripts (VHDJH-Cγ, VHDJH-Cα or VHDJH-Cε) were analyzed from splenic B cells of OVA-sensitized wild-type mice (with or without CPT treatment) and TLR2-/- mice (with or without IL21 treatment). RESULTS The lungs of TLR2-/- mice showed a lesser degree of inflammation than wild-type mice after OVA sensitization. Following OVA sensitization, levels of IL4, IL13, and IL21, but not IL5, were significantly lower in TLR2-/- compared with wild-type mice. Moreover, OVA-specific IgG1 and IgE titers were markedly lower and higher, respectively, in TLR2-/- mice. TLR2 deficiency inhibited STAT3 activation but not NF-κB p65 activation. CPT treatment reduced IgG1 titers via inhibition of Stat3 phosphorylation. Both TLR2 knockout and CPT treatment reduced the frequencies of Iγ1-Cγ1, Iγ3-Cγ3 and Iα-Cα transcripts, but IL21 treatment compensated for the effects of TLR2 deficiency. CONCLUSION These results suggest a role of TLR2 in restricting OVA-sensitized lung inflammation via promotion of IgG1 and inhibition of IgE class switching regulated by IL21 and STAT3.
Collapse
Affiliation(s)
- Yuqin Li
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| | - Qiu Chen
- School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
| | - Wei Ji
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| | - Yujie Fan
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| | - Li Huang
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| | - Chu Chu
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| | - Weifang Zhou
- Children’s Hospital of Soochow University, Suzhou, 215003 People’s Republic of China
| |
Collapse
|
12
|
Korb E, Drinić M, Wagner A, Geissler N, Inic-Kanada A, Peschke R, Joachim A, Wiedermann U, Schabussova I. Reduction of Allergic Lung Disease by Mucosal Application of Toxoplasma gondii-Derived Molecules: Possible Role of Carbohydrates. Front Immunol 2021; 11:612766. [PMID: 33776987 PMCID: PMC7988086 DOI: 10.3389/fimmu.2020.612766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/31/2020] [Indexed: 11/25/2022] Open
Abstract
Background The hygiene hypothesis suggests a link between parasitic infections and immune disorders, such as allergic diseases. We previously showed that infection with Toxoplasma gondii or systemic application of T. gondii tachyzoites lysate antigen (TLA) in a prophylactic, but not therapeutic protocol, prevented allergic airway inflammation in mice. Here we tested the effect of prophylactic and therapeutic application of TLA via the mucosal route. Methods Mice were intranasally treated with TLA either i) prior to sensitization, ii) during sensitization and challenge, or iii) after sensitization with ovalbumin (OVA). Recruitment of inflammatory cells to the lung, cytokine levels in restimulated lung and spleen cell cultures as well as levels of OVA-specific antibodies in serum were measured. In parallel, the effect of native TLA, heat-inactivated (hiTLA) or deglycosylated TLA (dgTLA) on sensitized splenocytes was evaluated ex vivo. Results When applied together with OVA i) during systemic sensitization and local challenge or ii) exclusively during local challenge, TLA reduced infiltration of eosinophils into the lung, OVA-specific type 2 cytokines in restimulated lung cell cultures, and partially, type 2 cytokines in restimulated spleen cell cultures in comparison to allergic controls. No beneficial effect was observed when TLA was applied prior to the start of sensitization. Analysis of epitope sugars on TLA indicated a high abundance of mannose, fucose, N-acetylglucosamine, and N-acetylgalactosamine. Deglycosylation of TLA, but not heat-inactivation, abolished the potential of TLA to reduce type 2 responses ex vivo, suggesting a significant role of carbohydrates in immunomodulation. Conclusion We showed that mucosal application of TLA reduced the development of experimental allergy in mice. The beneficial effects depended on the timing of the application in relation to the time point of sensitization. Not only co-application, but also therapy in sensitized/allergic animals with native TLA reduced local allergic responses. Furthermore, we show that TLA is highly glycosylated and glycoconjugates seem to play a role in anti-allergic effects. In summary, given the powerful modulatory effect that TLA exhibits, understanding its exact mechanisms of action may lead to the development of novel immunomodulators in clinical application.
Collapse
Affiliation(s)
- Elke Korb
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Mirjana Drinić
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Nora Geissler
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Aleksandra Inic-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Roman Peschke
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Anja Joachim
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Korb E, Bağcıoğlu M, Garner-Spitzer E, Wiedermann U, Ehling-Schulz M, Schabussova I. Machine Learning-Empowered FTIR Spectroscopy Serum Analysis Stratifies Healthy, Allergic, and SIT-Treated Mice and Humans. Biomolecules 2020; 10:biom10071058. [PMID: 32708591 PMCID: PMC7408032 DOI: 10.3390/biom10071058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
The unabated global increase of allergic patients leads to an unmet need for rapid and inexpensive tools for the diagnosis of allergies and for monitoring the outcome of allergen-specific immunotherapy (SIT). In this proof-of-concept study, we investigated the potential of Fourier-Transform Infrared (FTIR) spectroscopy, a high-resolution and cost-efficient biophotonic method with high throughput capacities, to detect characteristic alterations in serum samples of healthy, allergic, and SIT-treated mice and humans. To this end, we used experimental models of ovalbumin (OVA)-induced allergic airway inflammation and allergen-specific tolerance induction in BALB/c mice. Serum collected before and at the end of the experiment was subjected to FTIR spectroscopy. As shown by our study, FTIR spectroscopy, combined with deep learning, can discriminate serum from healthy, allergic, and tolerized mice, which correlated with immunological data. Furthermore, to test the suitability of this biophotonic method for clinical diagnostics, serum samples from human patients were analyzed by FTIR spectroscopy. In line with the results from the mouse models, machine learning-assisted FTIR spectroscopy allowed to discriminate sera obtained from healthy, allergic, and SIT-treated humans, thereby demonstrating its potential for rapid diagnosis of allergy and clinical therapeutic monitoring of allergic patients.
Collapse
Affiliation(s)
- Elke Korb
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (E.G.-S.); (U.W.)
| | - Murat Bağcıoğlu
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Erika Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (E.G.-S.); (U.W.)
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (E.G.-S.); (U.W.)
| | - Monika Ehling-Schulz
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: (M.E.-S.); (I.S.); Tel.: +43-1-25077-2460 (M.E.-S.); +43-1-40160-38250 (I.S.)
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.K.); (E.G.-S.); (U.W.)
- Correspondence: (M.E.-S.); (I.S.); Tel.: +43-1-25077-2460 (M.E.-S.); +43-1-40160-38250 (I.S.)
| |
Collapse
|
14
|
Serum amyloid A is a soluble pattern recognition receptor that drives type 2 immunity. Nat Immunol 2020; 21:756-765. [PMID: 32572240 DOI: 10.1038/s41590-020-0698-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
The molecular basis for the propensity of a small number of environmental proteins to provoke allergic responses is largely unknown. Herein, we report that mite group 13 allergens of the fatty acid-binding protein (FABP) family are sensed by an evolutionarily conserved acute-phase protein, serum amyloid A1 (SAA1), that promotes pulmonary type 2 immunity. Mechanistically, SAA1 interacted directly with allergenic mite FABPs (Der p 13 and Blo t 13). The interaction between mite FABPs and SAA1 activated the SAA1-binding receptor, formyl peptide receptor 2 (FPR2), which drove the epithelial release of the type-2-promoting cytokine interleukin (IL)-33 in a SAA1-dependent manner. Importantly, the SAA1-FPR2-IL-33 axis was upregulated in nasal epithelial cells from patients with chronic rhinosinusitis. These findings identify an unrecognized role for SAA1 as a soluble pattern recognition receptor for conserved FABPs found in common mite allergens that initiate type 2 immunity at mucosal surfaces.
Collapse
|
15
|
Gong F, Zheng T, Zhou P. T Follicular Helper Cell Subsets and the Associated Cytokine IL-21 in the Pathogenesis and Therapy of Asthma. Front Immunol 2019; 10:2918. [PMID: 31921177 PMCID: PMC6923700 DOI: 10.3389/fimmu.2019.02918] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
For many decades, T helper 2 (TH2) cells have been considered to predominantly regulate the pathogenic manifestations of allergic asthma, such as IgE-mediated sensitization, airway hyperresponsiveness, and eosinophil infiltration. However, recent discoveries have significantly shifted our understanding of asthma from a simple TH2 cell-dependent disease to a heterogeneous disease regulated by multiple T cell subsets, including T follicular helper (TFH) cells. TFH cells, which are a specialized cell population that provides help to B cells, have attracted intensive attention in the past decade because of their crucial role in regulating antibody response in a broad range of diseases. In particular, TFH cells are essential for IgE antibody class-switching. In this review, we summarize the recent progress regarding the role of TFH cells and their signature cytokine interleukin (IL)-21 in asthma from mouse studies and clinical reports. We further discuss future therapeutic strategies to treat asthma by targeting TFH cells and IL-21.
Collapse
Affiliation(s)
- Fang Gong
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ting Zheng
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Pengcheng Zhou
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
16
|
Li Y, Wang Y, Sun N, Yang H, Zhang Q, Zhang X, Huang R, Jia X. Activation-induced cytidine deaminase plays crucial role in ovalbumin-induced food allergy and promoted by IL-21. Mol Immunol 2019; 114:369-377. [PMID: 31450182 DOI: 10.1016/j.molimm.2019.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/08/2019] [Accepted: 08/18/2019] [Indexed: 10/26/2022]
Abstract
The prevalence of IgE-mediated food allergy is increasing in the whole wide world which often causes skin and gastrointestinal tract symptoms, or even fatal anaphylactic shock. However, the evaluation of food allergens remains difficult, and the mechanism of food allergy is still not fully clear. To study the gene expression profile in food allergy animal models and identify the regulatory mechanism of the crucial genes, two administration routes were used to build animal models in our study. OVA-specific IgE and IL-4 levels were tested by ELISA, transcriptome profiling was carried out by microarray, and the regulatory mechanism of the highest expressed gene was studied in the primary spleen cells. We found that activation-induced cytidine deaminase (Aicda) is the highest expressed gene in the allergic mice, IL-21 can dramatically enhance the expression of Aicda in the lymph node microenvironment, and IL-17A can promote this effect significantly though it has only limited influence by itself. At last, we illuminated that the promotion of IL-21 on Aicda is partially through STAT3. In summary, our results suggest that IL-21 and IL-17A may play important role in the expression of Aicda as well as food allergy.
Collapse
Affiliation(s)
- Yongning Li
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China
| | - Yongjun Wang
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan 250014, China
| | - Nanan Sun
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China
| | - Hui Yang
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China
| | - Qiannan Zhang
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China
| | - Xiaopeng Zhang
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China
| | - Run Huang
- Food Science and Engineering College, Beijing University Of Agriculture, Beijing 102206, China
| | - Xudong Jia
- NHC Key Laboratory of Food Safety Risk Assessment (China National Center for Food Safety Risk Assessment), Beijing, China.
| |
Collapse
|
17
|
Wang Z, Ji N, Chen Z, Wu C, Sun Z, Yu W, Hu F, Huang M, Zhang M. Next Generation Sequencing for Long Non-coding RNAs Profile for CD4 + T Cells in the Mouse Model of Acute Asthma. Front Genet 2019; 10:545. [PMID: 31231429 PMCID: PMC6565949 DOI: 10.3389/fgene.2019.00545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/22/2019] [Indexed: 01/13/2023] Open
Abstract
Background and Aims Although long non-coding RNAs (lncRNAs) have been linked to many diseases including asthma, little is known about lncRNA transcriptomes of CD4+ T cells in asthma. The present study aimed to explore the lncRNAs profile in the CD4+T cells from the mouse model of acute asthma. Methods Next generation sequencing for lncRNAs and mRNAs was performed on CD4+ T cells from asthma and control mice. Gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) pathway analyses were performed to predict the functions and signal pathways for the aberrant lncRNAs. The selected lncRNAs were further measured using quantitative real-time PCR (polymerase chain reaction) and observed in the fluorescence in situ hybridization (FISH). The lncRNA–mRNA co-expression network was constructed via Pearson’s correlation coefficient and Cytoscape 3.6. Results Next generation sequencing revealed 36 up-regulated lncRNAs and 98 down-regulated lncRNAs in acute asthma compared with controls. KEGG pathway analysis showed that cytokine-cytokine receptor interaction had the highest enrichment scores. A co-expression network was constructed in which 23 lncRNAs and 301 mRNAs altered formed a total of 12424 lncRNA and mRNA pairs. To validate the RNA sequencing results, we measured the 4 different lncRNAs using qPCR. The lncRNA fantom3_9230106C11 was significantly reduced in CD4+ T cells of asthma. Bioinformatics analysis showed that lncRNA fantom3_9230106C11 had the potential to interact with many miRNAs and transcription factors related to Th2 differentiation. Conclusion This study provided the first evidence for different expression of lncRNAs of CD4+T cells in asthma and may serve as a template for further, larger functional in-depth analyses regarding asthma molecular lncRNAs.
Collapse
Affiliation(s)
- Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chaojie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixiao Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenqin Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, China
| | - Fan Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China.,Department of Immunology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Gour N, Lajoie S, Smole U, White M, Hu D, Goddard P, Huntsman S, Eng C, Mak A, Oh S, Kim JH, Sharma A, Plante S, Salem IH, Resch Y, Xiao X, Yao N, Singh A, Vrtala S, Chakir J, Burchard EG, Lane AP, Wills-Karp M. Dysregulated invertebrate tropomyosin-dectin-1 interaction confers susceptibility to allergic diseases. Sci Immunol 2018; 3:3/20/eaam9841. [PMID: 29475849 DOI: 10.1126/sciimmunol.aam9841] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/25/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022]
Abstract
The key factors underlying the development of allergic diseases-the propensity for a minority of individuals to develop dysfunctional responses to harmless environmental molecules-remain undefined. We report a pathway of immune counter-regulation that suppresses the development of aeroallergy and shrimp-induced anaphylaxis. In mice, signaling through epithelially expressed dectin-1 suppresses the development of type 2 immune responses through inhibition of interleukin-33 (IL-33) secretion and the subsequent recruitment of IL-13-producing innate lymphoid cells. Although this homeostatic pathway is functional in respiratory epithelial cells from healthy humans, it is dramatically impaired in epithelial cells from asthmatic and chronic rhinosinusitis patients, resulting in elevated IL-33 production. Moreover, we identify an association between a single-nucleotide polymorphism (SNP) in the dectin-1 gene loci and reduced pulmonary function in two cohorts of asthmatics. This intronic SNP is a predicted eQTL (expression quantitative trait locus) that is associated with reduced dectin-1 expression in human tissue. We identify invertebrate tropomyosin, a ubiquitous arthropod-derived molecule, as an immunobiologically relevant dectin-1 ligand that normally serves to restrain IL-33 release and dampen type 2 immunity in healthy individuals. However, invertebrate tropomyosin presented in the context of impaired dectin-1 function, as observed in allergic individuals, leads to unrestrained IL-33 secretion and skewing of immune responses toward type 2 immunity. Collectively, we uncover a previously unrecognized mechanism of protection against allergy to a conserved recognition element omnipresent in our environment.
Collapse
Affiliation(s)
- Naina Gour
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stephane Lajoie
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Ursula Smole
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Marquitta White
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Donglei Hu
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pagé Goddard
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Scott Huntsman
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Celeste Eng
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Angel Mak
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sam Oh
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jung-Hyun Kim
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Annu Sharma
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Sophie Plante
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Ikhlass Haj Salem
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Yvonne Resch
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Xiao Xiao
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Nu Yao
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Anju Singh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jamila Chakir
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Esteban G Burchard
- Department of Medicine and Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andrew P Lane
- Division of Rhinology and Sinus Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Zwicker C, Sarate P, Drinić M, Ambroz K, Korb E, Smole U, Köhler C, Wilson MS, Kozakova H, Sebo P, Kverka M, Wiedermann U, Schabussova I. Prophylactic and therapeutic inhibition of allergic airway inflammation by probiotic Escherichia coli O83. J Allergy Clin Immunol 2018; 142:1987-1990.e7. [DOI: 10.1016/j.jaci.2018.07.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 07/15/2018] [Accepted: 07/31/2018] [Indexed: 11/26/2022]
|
20
|
Gour N, Smole U, Yong HM, Lewkowich IP, Yao N, Singh A, Gabrielson E, Wills-Karp M, Lajoie S. C3a is required for ILC2 function in allergic airway inflammation. Mucosal Immunol 2018; 11:1653-1662. [PMID: 30104625 PMCID: PMC6279480 DOI: 10.1038/s41385-018-0064-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/23/2018] [Indexed: 02/04/2023]
Abstract
Aberrant type 2 responses underlie the pathologies in allergic diseases like asthma, yet, our understanding of the mechanisms that drive them remains limited. Recent evidence suggests that dysregulated innate immune factors can perpetuate asthma pathogenesis. In susceptible individuals, allergen exposure triggers the activation of complement, a major arm of innate immunity, leading to the aberrant generation of the C3a anaphylatoxin. C3 and C3a have been shown to be important for the development of Th2 responses, yet remarkably, the mechanisms by which C3a regulates type 2 immunity are relatively unknown. We demonstrate a central role for C3a in driving type 2 innate lymphoid cells (ILC2)-mediated inflammation in response to allergen and IL-33. Our data suggests that ILC2 recruitment is C3a-dependent. Further, we show that ILC2s directly respond to C3a, promoting type 2 responses by specifically: (1) inducing IL-13 and granulocyte-macrophage colony-stimulating factor, whereas inhibiting IL-10 production from ILC2; and (2) enhancing their antigen-presenting capability during ILC-T-cell cross-talk. In summary, we identify a novel mechanism by which C3a can mediate aberrant type 2 responses to aeroallergen exposure, which involves a yet unrecognized cross-talk between two major innate immune components-complement and group 2 innate lymphoid cells.
Collapse
Affiliation(s)
- Naina Gour
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD,Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD
| | - Ursula Smole
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD,Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hwan-Mee Yong
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Ian P. Lewkowich
- Department of Immunobiology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH
| | - Nu Yao
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Anju Singh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Stephane Lajoie
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
21
|
Zhang X, Zhao X, Sun H, Yan Y, Huang L, Gu W, Jiang W, Wang Y, Zhu C, Ji W, Hao C, Chen Z. The role of miR-29c/B7-H3 axis in children with allergic asthma. J Transl Med 2018; 16:218. [PMID: 30075787 PMCID: PMC6076420 DOI: 10.1186/s12967-018-1590-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/25/2018] [Indexed: 12/26/2022] Open
Abstract
Background MicroRNAs play roles in the pathogenesis of bronchial asthma. However, the mechanism of miR-29c in allergic asthma remains unclear. This study is to elucidate the regulation of Th cell differentiation by miR-29c in mononuclear macrophages. Methods A total of 52 children with asthma exacerbation and 26 children as controls were enrolled in the study. CD14+ monocytes were isolated from the peripheral blood. Differential expressions of microRNAs were evaluated using microarray analysis and miR-29c expression in monocytes was determined by qRT-PCR. The plasma B7-H3 was determined by ELISA. Transfection studies and luciferase reporter assay were performed to confirm target gene of miR-29c and its function. Results Compared to controls, 88 miRNAs in blood monocytes were up-regulated and 41 miRNAs down-regulated including miR-29c in asthma children. Children with asthma exacerbation had significantly lower level of miR-29c and higher level of plasma B7-H3 compared to controls (both P < 0.05). Functional studies based on luciferase reporter assay and immunofluorescence staining suggest that B7-H3 is the direct target of miR-29c and transfection anti-miR-29c into macrophages could enhance ROR-γt and GATA-3 expression in co-cultured CD4+ T cells and increase levels of IL-4 and IL-17 in supernatants. Conclusion The axis of miR-29c/B7-H3 plays an important role in children with asthma through regulating Th2/Th17 cell differentiation and may provide new targets for treatment of asthma.
Collapse
Affiliation(s)
- Xinxing Zhang
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Huiming Sun
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yongdong Yan
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Li Huang
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wenjin Gu
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wujun Jiang
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yuqing Wang
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Canhong Zhu
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wei Ji
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China
| | - Chuangli Hao
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China.
| | - Zhengrong Chen
- Department of Respiratory Disease, Children's Hospital of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
22
|
Singh A, Liu C, Putman B, Zeig-Owens R, Hall CB, Schwartz T, Webber MP, Cohen HW, Berger KI, Nolan A, Prezant DJ, Weiden MD. Predictors of Asthma/COPD Overlap in FDNY Firefighters With World Trade Center Dust Exposure: A Longitudinal Study. Chest 2018; 154:1301-1310. [PMID: 30028968 DOI: 10.1016/j.chest.2018.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/31/2018] [Accepted: 07/02/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Previously healthy firefighters with World Trade Center (WTC) dust exposure developed airway disease. Risk factors for irritant-associated asthma/COPD overlap are poorly defined. METHODS This study included 2,137 WTC-exposed firefighters who underwent a clinically indicated bronchodilator pulmonary function test (BD-PFT) between 9/11/2001 and 9/10/2017. A post-BD FEV1 increase of > 12% and 200 mL from baseline defined asthma, and a post-BD FEV1/FVC ratio < 0.7 identified COPD cases. Participants who met both criteria had asthma/COPD overlap. Eosinophil levels were measured on screening blood tests performed shortly after 9/11/2001 and prior to BD-PFT; a subgroup of participants also had serum IgE and 21 cytokines measured (n = 215). Marginal Cox regression models for multiple events assessed the associations of eosinophil levels or serum biomarkers with subsequent diagnosis, with age, race, smoking, WTC exposure, first post-9/11 FEV1/FVC ratio, and BMI included as covariates. RESULTS BD-PFT diagnosed asthma/COPD overlap in 99 subjects (4.6%), isolated-asthma in 202 (9.5%), and isolated-COPD in 215 (10.1%). Eosinophil concentration ≥ 300 cells/μL was associated with increased risk of asthma/COPD overlap (hazard ratio [HR], 1.85; 95% CI, 1.16-2.95) but not with isolated-asthma or isolated-COPD. Serum IL-4 also predicted asthma/COPD overlap (HR, 1.51 per doubling of cytokine concentration; 95% CI, 1.17-1.95). Greater IL-21 concentration was associated with both isolated-asthma and isolated-COPD (HRs of 1.73 [95% CI, 1.27-2.35] and 2.06 [95% CI, 1.31-3.23], respectively). CONCLUSIONS In WTC-exposed firefighters, elevated blood eosinophil and IL-4 levels are associated with subsequent asthma/COPD overlap. Disease-specific T-helper cell type 2 biomarkers present years before diagnosis suggest patient-intrinsic predisposition to irritant-associated asthma/COPD overlap.
Collapse
Affiliation(s)
- Ankura Singh
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary Medicine Division, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Charles Liu
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Barbara Putman
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary, Critical Care and Sleep Medicine Division, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Rachel Zeig-Owens
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary Medicine Division, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY; Division of Epidemiology, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Charles B Hall
- Division of Biostatistics, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Theresa Schwartz
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary Medicine Division, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Mayris P Webber
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Division of Epidemiology, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Hillel W Cohen
- Division of Epidemiology, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Kenneth I Berger
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Anna Nolan
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary, Critical Care and Sleep Medicine Division, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - David J Prezant
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary Medicine Division, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Michael D Weiden
- Bureau of Health Services and the FDNY World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY; Pulmonary, Critical Care and Sleep Medicine Division, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York, NY.
| |
Collapse
|
23
|
O'Konek JJ, Landers JJ, Janczak KW, Goel RR, Mondrusov AM, Wong PT, Baker JR. Nanoemulsion adjuvant-driven redirection of T H2 immunity inhibits allergic reactions in murine models of peanut allergy. J Allergy Clin Immunol 2018; 141:2121-2131. [PMID: 29655584 DOI: 10.1016/j.jaci.2018.01.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 01/17/2018] [Accepted: 01/27/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immunotherapy for food allergies involves progressive increased exposures to food that result in desensitization to food allergens in some subjects but not tolerance to the food. Therefore new approaches to suppress allergic immunity to food are necessary. Previously, we demonstrated that intranasal immunization with a nanoemulsion (NE) adjuvant induces robust mucosal antibody and TH17-polarized immunity, as well as systemic TH1-biased cellular immunity with suppression of pre-existing TH2-biased immunity. OBJECTIVE We hypothesized that immunization with food in conjunction with the nanoemulsion adjuvant could lead to modulation of allergic reactions in food allergy by altering pre-existing allergic immunity and enhancing mucosal immunity. METHODS Mice were sensitized to peanut with aluminum hydroxide or cholera toxin. The animals were then administered 3 monthly intranasal immunizations with peanut in the nanoemulsion adjuvant or saline. Mice were then challenged with peanut to examine allergen reactivity. RESULTS The NE intranasal immunizations resulted in marked decreases in TH2 cytokine, IgG1, and IgE levels, whereas TH1 and mucosal TH17 immune responses were increased. After allergen challenge, these mice showed significant reductions in allergic hypersensitivity. Additionally, the NE immunizations significantly increased antigen-specific IL-10 production and regulatory T-cell counts, and the protection induced by NE was dependent in part on IL-10. Control animals immunized with intranasal peanut in saline had no modulation of their allergic response. CONCLUSIONS NE adjuvant-mediated induction of mucosal TH17 and systemic TH1-biased immunity can suppress TH2-mediated allergy through multiple mechanisms and protect against anaphylaxis. These results suggest the potential therapeutic utility of this approach in the setting of food allergy.
Collapse
Affiliation(s)
- Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| | - Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | | | - Rishi R Goel
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Anna M Mondrusov
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Pamela T Wong
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
24
|
Miljkovic D, Psaltis AJ, Wormald PJ, Vreugde S. Chronic Rhinosinusitis with Polyps Is Characterized by Increased Mucosal and Blood Th17 Effector Cytokine Producing Cells. Front Physiol 2017; 8:898. [PMID: 29311948 PMCID: PMC5742278 DOI: 10.3389/fphys.2017.00898] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
Background: Recent studies have implied a role for Th17 cells in CRS with nasal polyps (CRSwNP) patients. However, the capacity of these cells to produce Th17 cytokines is still unknown. Here we sought to quantify IL-17A, IL-17F, IL-21, and IL-22 cytokines produced by Th17 cells in mucosal tissue and peripheral blood of CRSwNP, CRS without nasal polyps (CRSsNP) and control patients. Methods: Samples were prospectively collected from CRS patients and non-CRS controls. We used flow cytometry to characterize the Th17 cells and their cytokines in sinonasal tissue and peripheral blood. Results: A total of 36 patients were recruited to the study. CRSwNP patients had significantly more tissue IL-17A (9.53 ± 2.71 vs. 1.11 ± 0.43 vs. 0.77 ± 0.07), IL-17F (4.96 ± 1.48 vs. 0.88 ± 0.31 vs. 0.56 ± 0.04), IL-21 (5.55 ± 2.01 vs. 1.60 ± 0.71 vs. 1.53 ± 0.55) and IL-22 (4.73 ± 1.58 vs. 0.70 ± 0.28 vs. 0.88 ± 0.26) producing Th17 cells compared to CRSsNP and control mucosa per mg of tissue, respectively. Allergic CRSwNP patients had decreased numbers of IL-21 producing Th17 cells compared to non-Allergic CRSwNP. (1.69 ± 0.57 vs. 9.41 ± 3.23) per mg of tissue, respectively (Kruskal-Wallis p < 0.05). Conclusion: In summary our study identified increased numbers of IL-17A, IL-17F, IL-21 and IL-22 positive Th17 cells in CRSwNP patient polyps and peripheral blood suggesting an altered activation state of those cells both locally and systemically. Atopic CRSwNP had decreased amounts of tissue Th17 cell derived IL-21 implying a potential protective role for IL-21 in CRSwNP allergic inflammation.
Collapse
Affiliation(s)
- Dijana Miljkovic
- Department of Surgery-Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Alkis J Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Peter J Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
25
|
Toxoplasma gondii tachyzoite-extract acts as a potent immunomodulator against allergic sensitization and airway inflammation. Sci Rep 2017; 7:15211. [PMID: 29123241 PMCID: PMC5680314 DOI: 10.1038/s41598-017-15663-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
Epidemiological and experimental studies have shown an inverse relationship between infections with certain parasites and a reduced incidence of allergic diseases. We and others have shown that infection with Toxoplasma gondii prevents the development of allergy in mice. To establish whether this beneficial effect could be recapitulated by soluble products of this parasite, we tested an extract derived from T. gondii tachyzoites. Immunization of BALB/c mice with tachyzoites lysate antigen (TLA) elicited mixed Th1/Th2 responses. When TLA was applied together with the sensitizing ovalbumin (OVA), the development of allergic airway inflammation was reduced, with decreased airway hyperresponsiveness associated with reduced peribronchial and perivascular cellular infiltration, reduced production of OVA-specific Th2 cytokines in lungs and spleens and reduced levels of serum OVA-specific IgG1 as well as IgE-dependent basophil degranulation. Of note, TLA retained its immunomodulatory properties, inducing high levels of IL-6, TNFα, IL-10 and IL-12p70 in bone marrow-derived dendritic cells after heat-inactivation or proteinase K-treatment for disruption of proteins, but not after sodium metaperiodate-treatment that degrades carbohydrate structures, suggesting that carbohydrates may play a role in immunomodulatory properties of TLA. Here we show that extracts derived from parasites may replicate the benefits of parasitic infection, offering new therapies for immune-mediated disorders.
Collapse
|
26
|
Abstract
Inflammation is a hallmark of many airway diseases. Improved understanding of the cellular and molecular mechanisms of airway disease will facilitate the transition in our understanding from phenotypes to endotypes, thereby improving our ability to target treatments based on pathophysiologic characteristics. For example, allergic asthma has long been considered to be driven by an allergen-specific T helper 2 response. However, clinical and mechanistic studies have begun to shed light on the role of other cell subsets in the pathogenesis and regulation of lung inflammation. In this review, we discuss the importance of different lymphocyte subsets to asthma and other airway diseases, while highlighting the growing evidence that asthma is a syndrome that incorporates many immune phenotypes.
Collapse
Affiliation(s)
- Daniel M Moldaver
- Firestone Institute for Respiratory Health, St. Joseph's Hospital Healthcare, Divisions of Clinical Immunology & Allergy and Respirology, Department of Medicine, McMaster University, Hamilton, ON
| | - Mark Larché
- Firestone Institute for Respiratory Health, St. Joseph's Hospital Healthcare, Divisions of Clinical Immunology & Allergy and Respirology, Department of Medicine, McMaster University, Hamilton, ON.
| | - Christopher D Rudulier
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
27
|
Halwani R, Sultana A, Vazquez-Tello A, Jamhawi A, Al-Masri AA, Al-Muhsen S. Th-17 regulatory cytokines IL-21, IL-23, and IL-6 enhance neutrophil production of IL-17 cytokines during asthma. J Asthma 2017. [PMID: 28635548 DOI: 10.1080/02770903.2017.1283696] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND In a subset of severe asthma patients, chronic airway inflammation is associated with infiltration of neutrophils, Th-17 cells and elevated expression of Th-17-derived cytokines (e.g., interleukin [IL]-17, IL-21, IL-22). Peripheral neutrophils from allergic asthmatics are known to express higher IL-17 cytokine levels than those from healthy subjects, but the regulatory mechanisms involved are not well understood. We hypothesize that Th-17 regulatory cytokines could modulate IL-17 expression in neutrophils. METHODS Peripheral blood neutrophils isolated from asthmatics were stimulated with IL-21, IL-23, and IL-6 cytokines and their ability to produce IL-17A and IL-17F was determined relative to healthy controls. Signal transducer and activator of transcription 3 (STAT3) phosphorylation levels were measured in stimulated neutrophil using flow cytometry. The requirement for STAT3 phosphorylation was determined by blocking its activation using a specific chemical inhibitor. RESULTS Stimulating asthmatic neutrophils with IL-21, 23, and 6 enhanced the production of IL-17A and IL-17F at significantly higher levels comparatively to healthy controls. Stimulating neutrophils with IL-21, IL-23, and IL-6 cytokines enhanced STAT3 phosphorylation, in all cases. Interestingly, inhibiting STAT3 phosphorylation using a specific chemical inhibitor dramatically blocked the ability of neutrophils to produce IL-17, demonstrating that STAT3 activation is the major factor mediating IL-17 gene expression. CONCLUSIONS These findings suggest that neutrophil infiltration in lungs of severe asthmatics may represent an important source of pro-inflammatory IL-17A and -F cytokines, a production enhanced by Th-17 regulatory cytokines, and thus providing a feedback mechanism that sustains inflammation. Our results suggest that STAT3 pathway could be a potential target for regulating neutrophilic inflammation during severe asthma.
Collapse
Affiliation(s)
- Rabih Halwani
- a Department of Pediatrics, Prince Naif Center for Immunology Research, Asthma Research Chair , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Asma Sultana
- a Department of Pediatrics, Prince Naif Center for Immunology Research, Asthma Research Chair , College of Medicine, King Saud University , Riyadh , Saudi Arabia.,b Prince Naif Health Research Center , King Saud University , Riyadh , Saudi Arabia
| | - Alejandro Vazquez-Tello
- a Department of Pediatrics, Prince Naif Center for Immunology Research, Asthma Research Chair , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Amer Jamhawi
- a Department of Pediatrics, Prince Naif Center for Immunology Research, Asthma Research Chair , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Abeer A Al-Masri
- c Department of Physiology , Faculty of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Saleh Al-Muhsen
- a Department of Pediatrics, Prince Naif Center for Immunology Research, Asthma Research Chair , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|
28
|
Hales BJ, Hizawa N, Jenmalm M, Sverremark-Ekström E, Wardlaw AJ. Developments in the field of allergy in 2014 through the eyes of Clinical and Experimental Allergy. Clin Exp Allergy 2016; 45:1723-45. [PMID: 26492197 DOI: 10.1111/cea.12663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of asthma continues to be a major topic of interest to our authors with reviews and original papers on the role of viruses, mechanisms of inflammation, biomarkers, and phenotypes of asthma being major topics. A number of papers described new treatments for asthma focusing on blocking the Th2 response reflecting the fact that two decades of work in this area is finally bearing fruit. The pathogenesis of chronic rhinosinusitis is a growing area of interest, but there has been less on the genetics of airways disease than in previous years possibly reflecting the degree of rigour (and therefore a smaller body of work), with which these sorts of studies are now being undertaken. There continues to be a wide range of papers dealing with mechanisms of allergic disease ranging from clinical-based studies to basic research and the use of in vivo animal models especially mice. As before, mechanisms and new approaches to immunotherapy are common themes. Several were published in the allergens section investigating modification of allergens to increase their effectiveness and reduce the risk of adverse events. Risk factors for allergic disease was a common theme in the epidemiology section and food allergy a common theme in clinical allergy with papers on the development of protocols to induce tolerance and attempts to find biomarkers to distinguish sensitization from allergic disease. This was another exciting year for the editors, and we hope the readers of the journal.
Collapse
Affiliation(s)
- B J Hales
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - N Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - M Jenmalm
- Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - E Sverremark-Ekström
- M.C., Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - A J Wardlaw
- Department of Infection Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK.,Department of Respiratory Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
29
|
Tian Y, Zajac AJ. IL-21 and T Cell Differentiation: Consider the Context. Trends Immunol 2016; 37:557-568. [PMID: 27389961 DOI: 10.1016/j.it.2016.06.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/27/2016] [Accepted: 06/01/2016] [Indexed: 12/18/2022]
Abstract
Accumulating studies demonstrate that IL-21 modulates the differentiation of various CD4 and CD8 T cell subsets and provide insights into the underlying cellular and molecular processes that are influenced by this cytokine. Intriguingly, the effects of IL-21 on T cells can be complex and vary depending on the experimental system used. We review our current understanding of the roles of IL-21 in the generation of phenotypically distinct CD4 and CD8 T cell populations and discuss the potential environmental cues, cellular factors, and molecular mediators that impact the actions of IL-21. We propose that IL-21 acts in a context-dependent manner to accentuate T cell subset development.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA
| | - Allan J Zajac
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA.
| |
Collapse
|
30
|
T-cell-intrinsic Tif1α/Trim24 regulates IL-1R expression on TH2 cells and TH2 cell-mediated airway allergy. Proc Natl Acad Sci U S A 2016; 113:E568-76. [PMID: 26787865 DOI: 10.1073/pnas.1522287113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αβ(+)CD4(+) T-helper 2 (TH2) cells orchestrate the type-2-driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081-E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4(gfp+)αβ(+)CD4(+) TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell-intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24(-/-) T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1-regulated signaling. Following this prediction, we found that Trim24(-/-) T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1β-mediated activation in vitro and in vivo, and fail to respond to IL-1β-exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell-intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.
Collapse
|
31
|
Halwani R, Sultana A, Al-Kufaidy R, Jamhawi A, Vazquez-Tello A, Al-Muhsen S. Th-17 regulatory cytokines inhibit corticosteroid induced airway structural cells apoptosis. Respir Res 2016; 17:6. [PMID: 26772733 PMCID: PMC4715361 DOI: 10.1186/s12931-015-0307-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022] Open
Abstract
Background Although corticosteroid is a powerful anti-inflammatory drug that is used widely to control asthma, still severe asthmatics can develop steroid resistance. Airway fibroblasts are quite resistant to steroids during Idiopathic pulmonary fibrosis (IPF) and fibrosis in asthmatic lungs is not always controlled. Th-17 regulatory cytokine which are elevated in lung tissues of asthmatics were shown to enhance the survival of various types of cells. STAT factors are central to this anti-apoptotic function. However, it is not yet clear whether these cytokines contribute to steroid hypo-responsiveness in asthma. Therefore, in this study, we investigated the ability of Th-17 regulatory cytokines, specifically IL-21, IL22 and IL23, to protect structural airway cells against dexamethasone-induced apoptosis. Methods Primary human fibroblasts, ASM cells, and lung endothelial cells line were treated with IL-21, IL-22, and IL-23 cytokines before incubation with dexamethasone and the level of apoptosis was determined by measuring cellular Annexin-V using Flow cytometry. Results Our data indicated that treatment with Th-17 regulatory cytokines was effective in inhibiting induced apoptosis for both fibroblasts and endothelial cells but not ASM cells. STAT3 phosphorylation levels were also upregulated in fibroblasts and endothelial upon treatment with these cytokines. Interestingly, inhibiting STAT3 phosphorylation abrogated IL-21, IL-22, and IL-23 anti-apoptotic effect on fibroblasts and endothelial cells. Conclusions This data suggest that Th-17 regulatory cytokines may play a critical role in regulating the survival of fibroblasts during asthma, IPF as well as other chronic lung inflammatory diseases leading to enhanced fibrosis. Accordingly, findings of this paper may pave the way for more extensive research on the role of these regulatory cytokines in fibrosis development in various chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rabih Halwani
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia.
| | - Asma Sultana
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia.,Prince Naif Health Research Center, King Saud University, Riyadh, Saudi Arabia
| | - Roua Al-Kufaidy
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Amer Jamhawi
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Alejandro Vazquez-Tello
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Saleh Al-Muhsen
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Cheng S, Chen H, Wang A, Bunjhoo H, Cao Y, Xie J, Xu Y, Xiong W. Exclusion of IL-21 in the pathogenesis of OVA-induced asthma in mice. Int J Clin Exp Med 2014; 7:3202-3208. [PMID: 25419350 PMCID: PMC4238472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/15/2014] [Indexed: 06/04/2023]
Abstract
Asthma is characterized by airway inflammation, mucus overproduction, and airway hyperreactivity. Cytokines, especially T helper 2-derived cytokines interleukin (IL)-4, IL-5, and IL-13, are involved in the pathogenesis of asthma. IL-21 has a variety of effects on the immune system. However, the contribution of IL-21 to the development of allergic diseases is currently controversial. The aim of this study was to investigate the effect of IL-21 on asthma airway inflammation in vivo. A murine ovalbumin (OVA)-induced allergic asthma model was used. The concentration of IL-21 in the bronchoalveolar lavage fluid (BALF) of mice was evaluated by enzyme-linked immunosorbent assay. BALF cellularity, lung histopathology, and sera IgE levels were compared between the normal control group, OVA sensitization/challenge group, and OVA sensitization/challenge plus IL-21-administered group. An OVA-induced allergic rhinitis model with IL-21 was used as a positive control and the infiltration of eosinophils in the nasal mucosa was evaluated. The concentration of IL-21 in the BALF was lower in the asthmatic group compared with the normal control group. However, no significant differences in airway eosinophilia, lung histopathology, and sera IgE levels were observed between the OVA sensitization/challenge group and OVA sensitization/challenge plus IL-21-administered group. Decreased eosinophilic infiltration of nasal mucosa was observed in the positive control allergic rhinitis model administered IL-21 during the challenge period. Exogenous administration of IL-21 alone may not alleviate allergic lung inflammation. The role of IL-21 in allergic lung inflammation needs further research.
Collapse
Affiliation(s)
- Sheng Cheng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Huilong Chen
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Aili Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Hansvin Bunjhoo
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Yong Cao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology 1095 Jiefang Ave., Wuhan 430030, China
| |
Collapse
|