1
|
Perez-Garcia J, Cardenas A, Lorenzo-Diaz F, Pino-Yanes M. Precision medicine for asthma treatment: Unlocking the potential of the epigenome and microbiome. J Allergy Clin Immunol 2025; 155:298-315. [PMID: 38906272 PMCID: PMC12002393 DOI: 10.1016/j.jaci.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Asthma is a leading worldwide biomedical concern. Patients can experience life-threatening worsening episodes (exacerbations) usually controlled by anti-inflammatory and bronchodilator drugs. However, substantial heterogeneity in treatment response exists, and a subset of patients with unresolved asthma carry the major burden of this disease. The study of the epigenome and microbiome might bridge the gap between human genetics and environmental exposure to partially explain the heterogeneity in drug response. This review aims to provide a critical examination of the existing literature on the microbiome and epigenetic studies examining associations with asthma treatments and drug response, highlight convergent pathways, address current challenges, and offer future perspectives. Current epigenetic and microbiome studies have shown the bilateral relationship between asthma pharmacologic interventions and the human epigenome and microbiome. These studies, focusing on corticosteroids and to a lesser extent on bronchodilators, azithromycin, immunotherapy, and mepolizumab, have improved the understanding of the molecular basis of treatment response and identified promising biomarkers for drug response prediction. Immune and inflammatory pathways (eg, IL-2, TNF-α, NF-κB, and C/EBPs) underlie microbiome-epigenetic associations with asthma treatment, representing potential therapeutic pathways to be targeted. A comprehensive evaluation of these omics biomarkers could significantly contribute to precision medicine and new therapeutic target discovery.
Collapse
Affiliation(s)
- Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain.
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, Calif
| | - Fabian Lorenzo-Diaz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Potaczek DP, Bazan-Socha S, Wypasek E, Wygrecka M, Garn H. Recent Developments in the Role of Histone Acetylation in Asthma. Int Arch Allergy Immunol 2024; 185:641-651. [PMID: 38522416 DOI: 10.1159/000536460] [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: 12/14/2023] [Accepted: 01/22/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND Epigenetic modifications are known to mediate both beneficial and unfavorable effects of environmental exposures on the development and clinical course of asthma. On the molecular level, epigenetic mechanisms participate in multiple aspects of the emerging and ongoing asthma pathology. SUMMARY Studies performed in the last several years expand our knowledge on the role of histone acetylation, a classical epigenetic mark, in the regulation of (patho)physiological processes of diverse cells playing a central role in asthma, including those belonging to the immune system (e.g., CD4+ T cells, macrophages) and lung structure (e.g., airway epithelial cells, pulmonary fibroblasts). Those studies demonstrate a number of specific histone acetylation-associated mechanisms and pathways underlying pathological processes characteristic for asthma, as well as report their modification modalities. KEY MESSAGES Dietary modulation of histone acetylation levels in the immune system might protect against the development of asthma and other allergies. Interfering with the enzymes controlling the histone acetylation status of structural lung and (local) immune cells might provide future therapeutic options for asthmatics. Despite some methodological obstacles, analysis of the histone acetylation levels might improve asthma diagnostics.
Collapse
Affiliation(s)
- Daniel P Potaczek
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Marburg, Germany
- Center for Infection and Genomics of the Lung (CIGL), Member of the Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Bioscientia MVZ Labor Mittelhessen GmbH, Giessen, Germany
| | - Stanisława Bazan-Socha
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Ewa Wypasek
- Krakow Center for Medical Research and Technology, John Paul II Hospital, Krakow, Poland
- Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Krakow, Poland
| | - Małgorzata Wygrecka
- Center for Infection and Genomics of the Lung (CIGL), Member of the Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Institute of Lung Health, Member of the German Center for Lung Research (DZL), Giessen, Germany
- CSL Behring Innovation GmbH, Marburg, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Marburg, Germany
| |
Collapse
|
3
|
Cha J, Choi S. Gene-Smoking Interaction Analysis for the Identification of Novel Asthma-Associated Genetic Factors. Int J Mol Sci 2023; 24:12266. [PMID: 37569643 PMCID: PMC10419280 DOI: 10.3390/ijms241512266] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/26/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Asthma is a complex heterogeneous disease caused by gene-environment interactions. Although numerous genome-wide association studies have been conducted, these interactions have not been systemically investigated. We sought to identify genetic factors associated with the asthma phenotype in 66,857 subjects from the Health Examination Study, Cardiovascular Disease Association Study, and Korea Association Resource Study cohorts. We investigated asthma-associated gene-environment (smoking status) interactions at the level of single nucleotide polymorphisms, genes, and gene sets. We identified two potentially novel (SETDB1 and ZNF8) and five previously reported (DM4C, DOCK8, MMP20, MYL7, and ADCY9) genes associated with increased asthma risk. Numerous gene ontology processes, including regulation of T cell differentiation in the thymus (GO:0033081), were significantly enriched for asthma risk. Functional annotation analysis confirmed the causal relationship between five genes (two potentially novel and three previously reported genes) and asthma through genome-wide functional prediction scores (combined annotation-dependent depletion, deleterious annotation of genetic variants using neural networks, and RegulomeDB). Our findings elucidate the genetic architecture of asthma and improve the understanding of its biological mechanisms. However, further studies are necessary for developing preventive treatments based on environmental factors and understanding the immune system mechanisms that contribute to the etiology of asthma.
Collapse
Affiliation(s)
- Junho Cha
- Department of Applied Artificial Intelligence, College of Computing, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
| | - Sungkyoung Choi
- Department of Applied Artificial Intelligence, College of Computing, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
- Department of Mathematical Data Science, College of Science and Convergence Technology, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| |
Collapse
|
4
|
McDaniel MM, Lara HI, von Moltke J. Initiation of type 2 immunity at barrier surfaces. Mucosal Immunol 2023; 16:86-97. [PMID: 36642383 DOI: 10.1016/j.mucimm.2022.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 01/15/2023]
Abstract
Although seemingly unrelated, parasitic worms, venoms, and allergens all induce a type 2 immune response. The effector functions and clinical features of type 2 immunity are well-defined, but fundamental questions about the initiation of type 2 immunity remain unresolved. How are these enormously diverse type 2 stimuli first detected? How are type 2 helper T cells primed and regulated? And how do mechanisms of type 2 initiation vary across tissues? Here, we review the common themes governing type 2 immune sensing and explore aspects of T cell priming and effector reactivation that make type 2 helper T cells a unique T helper lineage. Throughout the review, we emphasize the importance of non-hematopoietic cells and highlight how the unique anatomy and physiology of each barrier tissue shape mechanisms of type 2 immune initiation.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, USA.
| | - Heber I Lara
- Department of Immunology, University of Washington School of Medicine, Seattle, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, USA
| |
Collapse
|
5
|
Tissue adaptation and clonal segregation of human memory T cells in barrier sites. Nat Immunol 2023; 24:309-319. [PMID: 36658238 PMCID: PMC10063339 DOI: 10.1038/s41590-022-01395-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/22/2022] [Indexed: 01/21/2023]
Abstract
T lymphocytes migrate to barrier sites after exposure to pathogens, providing localized immunity and long-term protection. Here, we obtained blood and tissues from human organ donors to examine T cells across major barrier sites (skin, lung, jejunum), associated lymph nodes, lymphoid organs (spleen, bone marrow), and in circulation. By integrating single-cell protein and transcriptome profiling, we demonstrate that human barrier sites contain tissue-resident memory T (TRM) cells that exhibit site-adapted profiles for residency, homing and function distinct from circulating memory T cells. Incorporating T cell receptor and transcriptome analysis, we show that circulating memory T cells are highly expanded, display extensive overlap between sites and exhibit effector and cytolytic functional profiles, while TRM clones exhibit site-specific expansions and distinct functional capacities. Together, our findings indicate that circulating T cells are more disseminated and differentiated, while TRM cells exhibit tissue-specific adaptation and clonal segregation, suggesting that strategies to promote barrier immunity require tissue targeting.
Collapse
|
6
|
Li H, Wang H, Sokulsky L, Liu S, Yang R, Liu X, Zhou L, Li J, Huang C, Li F, Lei X, Jia H, Cheng J, Li F, Yang M, Zhang G. Single-cell transcriptomic analysis reveals key immune cell phenotypes in the lungs of patients with asthma exacerbation. J Allergy Clin Immunol 2021; 147:941-954. [PMID: 33039479 DOI: 10.1016/j.jaci.2020.09.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Asthma exacerbations are associated with heightened asthma symptoms, which can result in hospitalization in severe cases. However, the molecular immunologic processes that determine the course of an exacerbation remain poorly understood, impeding the progression of development of effective therapies. OBJECTIVE Our aim was to identify candidate genes that are strongly associated with asthma exacerbation at a cellular level. METHODS Subjects with asthma exacerbation and healthy control subjects were recruited, and bronchoalveolar lavage fluid was isolated from these subjects via bronchoscopy. Cells were isolated through fluorescence-activated cell sorting, and single-cell RNA sequencing was performed on enriched cell populations. RESULTS We showed that the levels of monocytes, CD8+ T cells, and macrophages are significantly elevated in the bronchoalveolar lavage fluid of patients. A set of cytokines and intracellular transduction regulators are associated with asthma exacerbations and are shared across multiple cell clusters, forming a complicated molecular framework. An additional group of core exacerbation-associated modules is activated, including eukaryotic initiation factor 2 signaling, ephrin receptor signaling, and C-X-C chemokine receptor type 4 signaling in the subpopulations of CD8+ T cells (C1-a) and monocyte clusters (C7 clusters), which are associated with infection. CONCLUSION Our study identified a significant number of severe asthma-associated genes that are differentially expressed by multiple cell clusters.
Collapse
Affiliation(s)
- Hui Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huaqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leon Sokulsky
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Shaoxia Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Liu
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lujia Zhou
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Juan Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxia Jia
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiuling Cheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuguang Li
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia; Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
7
|
Wang CM, Chang CB, Lee SP, W-Y Chan M, Wu SF. Differential DNA methylation profiles of peripheral blood mononuclear cells in allergic asthmatic children following dust mite immunotherapy. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:986-995. [PMID: 32684341 DOI: 10.1016/j.jmii.2020.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND/PURPOSE Allergen-specific immunotherapy (SIT) is now considered curative to allergic diseases such as asthma. Mechanistically, our previous work showed DNA hypermethylation of cytokine genes, in T-helper cells, in allergic asthmatic children treated with allergen-SIT. In this study, we extended to work to assess possible changes in the DNA methylomes of peripheral blood mononuclear cells (PBMCs), isolated from mite allergen-SIT asthmatic children, to explore further the underlying methylation changes. METHODS Thirteen allergic asthmatic children who received Der p-SIT, 12 non-SIT allergic asthmatic controls, and 12 healthy controls were enrolled. Bisulfite-converted DNA from Der p-stimulated PBMCs was analyzed using Human Methylation 450 k BeadChip. Pyrosequencing and quantitative real-time PCR were used to validate the DNA methylation levels and the gene expression of individual samples. RESULTS We identified 108 significantly differentially methylated regions (DMRs) unique to Der p-treated PBMCs, with 53 probes linked to demethylated DMRs, and 55 probes linked to methylated DMRs. Three associated genes (BCL6, HSPG2, and HSP90AA1), of selected DMRs, were subjected to bisulfite pyrosequencing. Of these, BCL6 showed significant hypomethylation, while HSPG2 and HSP90AA1 were hypermethylated in SIT group, compared to the AA group. Furthermore, SIT group had significantly higher gene expression of BCL6 and lower gene expression of HSPG2. KEGG pathway analysis further revealed DMR genes involved in ECM-receptor interactions, asthma, and antigen processing and presentation pathways. CONCLUSIONS Several DNA regions showed DNA methylation altered by Der p specific immunotherapy, indicating desensitization-associated methylomes. Genes belonging to these SIT-altered pathways may represent therapeutic targets for better clinical management of asthma.
Collapse
Affiliation(s)
- Chuang-Ming Wang
- Department of Pediatrics, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan; Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
| | - Chia-Bin Chang
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, Chia-Yi, Taiwan
| | - Shiao-Pieng Lee
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Michael W-Y Chan
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, Chia-Yi, Taiwan
| | - Shu-Fen Wu
- Center for Innovative Research on Aging Society, National Chung Cheng University, Chia-Yi, Taiwan.
| |
Collapse
|
8
|
Zwiers A, van Wanrooij RL, Dieckman T, Nijeboer P, Kraal G, Bouma G. Celiac disease associated SNP rs17810546 is located in a gene silencing region. Gene 2020; 726:144165. [DOI: 10.1016/j.gene.2019.144165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022]
|
9
|
Farahnak S, Simon L, McGovern TK, Chen M, Khazaei N, Martin JG. HB-EGF Synthesized by CD4 T Cells Modulates Allergic Airway Eosinophilia by Regulating IL-5 Synthesis. THE JOURNAL OF IMMUNOLOGY 2019; 203:39-47. [PMID: 31127030 DOI: 10.4049/jimmunol.1801686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022]
Abstract
CD4 T cells express the epidermal growth factor (EGF) receptor ligand, heparin-binding EGF (HB-EGF), with no defined immuno-pathophysiological function. Therefore, we wished to elucidate the function of HB-EGF synthesized by CD4 T cells in the context of allergic pulmonary inflammation and the asthma surrogate, airway hyperresponsiveness, in a murine acute model of asthma. In this study, we show how knocking out HB-EGF expression in CD4 T cells in vivo attenuates IL-5 synthesis in the lung that is accompanied by diminished eosinophilic inflammation and airway hyperresponsiveness. HB-EGF coimmunoprecipitates with the transcriptional repressor B cell lymphoma 6 (Bcl-6) in CD4 T cells. Knocking out HB-EGF in CD4 T cells resulted in increased Bcl-6 binding to the IL-5 gene and decreased IL-5 mRNA expression. Thus, these findings suggest an immunoregulatory function for intrinsic HB-EGF expressed by CD4 T cells in TH2 inflammation and airway dysfunction by modulating IL-5 expression via binding to and inhibiting the repressive function of Bcl-6.
Collapse
Affiliation(s)
- Soroor Farahnak
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Leora Simon
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Toby K McGovern
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Michael Chen
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Niusha Khazaei
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
10
|
Ogasawara T, Kohashi Y, Ikari J, Taniguchi T, Tsuruoka N, Watanabe-Takano H, Fujimura L, Sakamoto A, Hatano M, Hirata H, Fukushima Y, Fukuda T, Kurasawa K, Tatsumi K, Tokuhisa T, Arima M. Allergic T H2 Response Governed by B-Cell Lymphoma 6 Function in Naturally Occurring Memory Phenotype CD4 + T Cells. Front Immunol 2018; 9:750. [PMID: 29696026 PMCID: PMC5904433 DOI: 10.3389/fimmu.2018.00750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 03/26/2018] [Indexed: 02/05/2023] Open
Abstract
Transcriptional repressor B-cell lymphoma 6 (Bcl6) appears to regulate TH2 immune responses in allergies, but its precise role is unclear. We previously reported that Bcl6 suppressed IL-4 production in naïve CD4+ T cell-derived memory TH2 cells. To investigate Bcl6 function in allergic responses in naturally occurring memory phenotype CD4+ T (MPT) cells and their derived TH2 (MPTH2) cells, Bcl6-manipulated mice, highly conserved intron enhancer (hcIE)-deficient mice, and reporter mice for conserved noncoding sequence 2 (CNS2) 3′ distal enhancer region were used to elucidate Bcl6 function in MPT cells. The molecular mechanisms of Bcl6-mediated TH2 cytokine gene regulation were elucidated using cellular and molecular approaches. Bcl6 function in MPT cells was determined using adoptive transfer to naïve mice, which were assessed for allergic airway inflammation. Bcl6 suppressed IL-4 production in MPT and MPTH2 cells by suppressing CNS2 enhancer activity. Bcl6 downregulated Il4 expression in MPTH2 cells, but not MPT cells, by suppressing hcIE activity. The inhibitory functions of Bcl6 in MPT and MPTH2 cells attenuated allergic responses. Bcl6 is a critical regulator of IL-4 production by MPT and MPTH2 cells in TH2 immune responses related to the pathogenesis of allergies.
Collapse
Affiliation(s)
- Takashi Ogasawara
- Department of Respirology (B2), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuko Kohashi
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jun Ikari
- Department of Respirology (B2), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toshibumi Taniguchi
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nobuhide Tsuruoka
- Department of Reproductive Medicine (G4), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Haruko Watanabe-Takano
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Lisa Fujimura
- Biomedical Research Center, Chiba University, Chiba, Japan
| | - Akemi Sakamoto
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masahiko Hatano
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hirokuni Hirata
- Department of Respiratory Medicine and Clinical Immunology, Dokkyo Medical University Koshigaya Hospital, Koshigaya, Japan
| | - Yasutsugu Fukushima
- Department of Respiratory Medicine and Clinical Immunology, Dokkyo Medical University Koshigaya Hospital, Koshigaya, Japan
| | - Takeshi Fukuda
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Kazuhiro Kurasawa
- Department of Rheumatology, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Koichiro Tatsumi
- Department of Respirology (B2), Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takeshi Tokuhisa
- Department of Developmental Genetics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masafumi Arima
- Department of Biomedical Science (M14), Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Rheumatology, Dokkyo Medical University School of Medicine, Mibu, Japan
| |
Collapse
|
11
|
Huang C, Zhang Z, Wang L, Liu J, Gong X, Zhang C. ML-7 attenuates airway inflammation and remodeling via inhibiting the secretion of Th2 cytokines in mice model of asthma. Mol Med Rep 2018; 17:6293-6300. [PMID: 29512725 PMCID: PMC5928606 DOI: 10.3892/mmr.2018.8683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/01/2017] [Indexed: 01/05/2023] Open
Abstract
Previous studies have indicated that smooth muscle myosin light chain kinase (MLCK) has a prominent role in the regulation of smooth muscle contraction, which tends to be upregulated in asthma. In recent years, numerous studies have reported that MLCK is intimately connected with the immunoregulatory mechanism of T cells. The imbalance of T helper type 1 cells (Th1)/Th2 constitutes the immune-associated pathological basis of chronic asthma. Th2-associated cytokines, including interleukin-4, −5, −13, −25 and −33, are involved in airway inflammation, hyperresponsiveness and remodeling, which leads to a progressive decline in lung function. The purpose of the present study was to verify whether inhibition of bronchial MLCK attenuated the expression Th2-associated cytokines in asthmatic mice, including the above-mentioned ones. Female BALB/c mice were used to establish an ovalbumin (OVA)-induced model of asthma, of which one group was treated with the MLCK inhibitor (5-iodonaphthalene-1-sulfonyl) homopiperazine (ML-7). The inhibitor of MLCK, ML-7 attenuated airway inflammation and remodeling by reducing inflammatory cell infiltration and the secretion of Th2 cytokines in mice model of asthma, which may represent a promising therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Chuanjun Huang
- Department of Respiratory Diseases, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Zewen Zhang
- Department of Medical Imaging and Nuclear Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Liuxin Wang
- Department of Respiratory Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Ju Liu
- Department of Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Xiaodan Gong
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Caiqing Zhang
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
12
|
Abstract
T helper 2 (Th2) cells are defined by their ability to produce the hallmark cytokine IL-4. However, to mediate allergic inflammation in tissues, Th2 cells must secrete additional cytokines including IL-13 and IL-5. We used IL-4 and IL-13 dual-reporter mice to show that naive CD4+ T cells cultured in the presence of IL-4 and thymic stromal lymphopoietin (TSLP) generate a population of IL-4negIL-13pos Th2 cells that develop from IL-4neg precursors and express the Th2 effector cytokines IL-5 and IL-9. In vivo, high TSLP levels promote the development of a similar population of IL-4negIL-13pos T cells that also express Gata3, Il5, and Il3 transcripts. Thus, TSLP drives the early differentiation of a distinct population of effector Th2 cells with pro-inflammatory properties. T helper 2 (Th2) cells are pivotal in the development of allergy. Allergen exposure primes IL-4+ Th2 cells in lymph node, but production of effector cytokines including IL-5 and IL-13 is thought to require additional signals from antigen and the environment. Here we report that a substantial proportion of naive CD4+ T cells in spleen and lymph node express receptors for the epithelium-derived inflammatory cytokine thymic stromal lymphopoietin (TSLP). Culture of naive CD4+ T cells in anti-(a)CD3, aCD28, and TSLP-supplemented Th2 conditions enabled the development of a unique population of IL-13-single positive (IL-13-SP) CD4+ T cells; TSLP and Th2 conditions were both required for their development. Sorting experiments revealed that IL-13-SP Th2 cells originated from IL-4-negative precursors and coexpressed transcripts for the Th2 cytokines IL-5 and IL-9. In vivo, high TSLP levels acted directly on CD4+ T cells to induce the development of IL-13-SP and IL-4+IL-13+ double-positive populations in lymph node. These cells were phenotypically similar to Th2 effector cells and were CXCR5lowPD1low and expressed low levels of Bcl6 and Il21 transcripts and high levels of Gata3, Il3, and Il5. Our findings suggest a role of TSLP in directly promoting Th2 cell effector function and support the notion of TSLP as a key driver of Th2 inflammation.
Collapse
|