1
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Wisnewski AV, Liu J. Lung Gene Expression Suggests Roles for Interferon-Stimulated Genes and Adenosine Deaminase Acting against RNA-1 in Pathologic Responses to Diisocyanate. Chem Res Toxicol 2024; 37:476-485. [PMID: 38494904 PMCID: PMC11748834 DOI: 10.1021/acs.chemrestox.3c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Mechanisms underlying methylene diphenyl diisocyanate (MDI) and other low molecular weight chemical-induced asthma are unclear and appear distinct from those of high molecular weight (HMW) allergen-induced asthma. We sought to elucidate molecular pathways that differentiate asthma-like pathogenic vs nonpathogenic responses to respiratory tract MDI exposure in a murine model. Lung gene expression differences in MDI exposed immune-sensitized and nonsensitized mice vs unexposed controls were measured by microarrays, and associated molecular pathways were identified through bioinformatic analyses and further compared with published studies of a prototypic HMW asthmagen (ovalbumin). Respiratory tract MDI exposure significantly altered lung gene expression in both nonsensitized and immune-sensitized mice, vs controls. Fifty-three gene transcripts were altered in all MDI exposed lung tissue vs controls, with levels up to 10-fold higher in immune-sensitized vs nonsensitized mice. Gene transcripts selectively increased in MDI exposed immune-sensitized animals were dominated by chitinases and chemokines and showed substantial overlap with those increased in ovalbumin-induced asthma. In contrast, MDI exposure of nonsensitized mice increased type I interferon stimulated genes (ISGs) in a pattern reflecting deficiency in adenosine deaminase acting against RNA (ADAR-1), an important regulator of innate, as well as "sterile" or autoimmunity triggered by tissue damage. Thus, MDI-induced changes in lung gene expression were identified that differentiate nonpathogenic innate responses in nonsensitized hosts from pathologic adaptive responses in immune-sensitized hosts. The data suggest that MDI alters unique biological pathways involving ISGs and ADAR-1, potentially explaining its unique immunogenicity/allergenicity.
Collapse
Affiliation(s)
- Adam V Wisnewski
- Department of Internal Medicine, Yale University School of Medicine, New Haven, 06520, Connecticut United States
| | - Jian Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, 06520, Connecticut United States
| |
Collapse
|
3
|
Ge X, Xu T, Wang M, Gao L, Tang Y, Zhang N, Zheng R, Zeng W, Chen G, Zhang B, Dai Y, Zhang Y. Chalcone-derivative L6H21 attenuates the OVA-induced asthma by targeting MD2. Eur J Med Res 2024; 29:65. [PMID: 38245791 PMCID: PMC10799361 DOI: 10.1186/s40001-023-01630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/28/2023] [Indexed: 01/22/2024] Open
Abstract
Asthma represents a significant global challenge that affects individuals across all age groups and imposes substantial social and economic burden. Due to heterogeneity of the disease, not all patients obtain benefit with current treatments. The objective of this study was to explore the impact of MD2 on the progression of asthma using L6H21, a novel MD2 inhibitor, to identify potential targets and drug candidates for asthma treatment. To establish an asthma-related murine model and evaluate the effects of L6H21, ovalbumin (OVA) was used to sensitize and challenge mice. Pathological changes were examined with various staining techniques, such as H&E staining, glycogen staining, and Masson staining. Inflammatory cell infiltration and excessive cytokine secretion were evaluated by analyzing BALF cell count, RT-PCR, and ELISA. The TLR4/MD2 complex formation, as well as the activation of the MAPK and NF-кB pathways, was examined using western blot and co-IP. Treatment with L6H21 demonstrated alleviation of increased airway resistance, lung tissue injury, inflammatory cell infiltration and excessive cytokine secretion triggered by OVA. In addition, it also ameliorated mucus production and collagen deposition. In the L6H21 treatment group, inhibition of MAPK and NF-кB activation was observed, along with the disruption of TLR4/MD2 complex formation, in contrast to the model group. Thus, L6H21 effectively reduced the formation of the MD2 and TLR4 complex induced by OVA in a dose-dependent manner. This reduction resulted in the attenuation of MAPKs/NF-κB activation, enhanced suppression of inflammatory factor secretion, reduced excessive recruitment of inflammatory cells, and ultimately mitigated airway damage. MD2 emerges as a crucial target for asthma treatment, and L6H21, as an MD2 inhibitor, shows promise as a potential drug candidate for the treatment of asthma.
Collapse
Affiliation(s)
- Xiangting Ge
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Tingting Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Meiyan Wang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lijiao Gao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yue Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ningjie Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Weimin Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
| | - Yuanrong Dai
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yali Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
4
|
Lin Q, Wang T, Zuo X, Ni H, Zhong J, Zhan L, Cheng H, Huang Y, Ding X, Yu H, Nie H. Anti-CD1d treatment suppresses immunogenic maturation of lung dendritic cells dependent on lung invariant natural killer T cells in asthmatic mice. Int Immunopharmacol 2023; 124:110921. [PMID: 37725846 DOI: 10.1016/j.intimp.2023.110921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
Our previous findings show that invariant natural killer T (iNKT)cells can promote immunogenic maturation of lung dendritic cells (LDCs) to enhance Th2 cell responses in asthma. It has been accepted that recognition of glycolipid antigens presented by CD1d molecules by the T cell receptors of iNKT cells leads to iNKT cell activation. Therefore, we examine the immunoregulatory influences of anti-CD1d treatment on Th2 cell response and immunogenic maturation of LDCs and subsequently explored whether these influences were dependent on lung iNKT cells in asthmatic mice. We discoveredthat in wild-type mice sensitized and challenged with house dust mite or ovalbumin (OVA), anti-CD1d treatment inhibited Th2 cell response and immunogenic maturation of LDCs. LDCs from asthmatic mice with anti-CD1d treatment had a markedly decreased influence on Th2 cell responses in vivo and in vitro. Furthermore, anti-CD1d treatment reduced the abundance and activation of lung iNKT cells in asthmatic mice. Moreover, in asthmatic iNKT cell-deficient Jα18-/- mice, anti-CD1d treatment did not influence Th2 cell responses and immunogenic maturation of LDCs. Meanwhile, the quantity of CD40L+ iNKT cells in asthmatic mice was significant decreased by anti-CD1d treatment. Finally, the inhibition of anti-CD1d treatment on LDC immunogenic maturation and Th2 cell responses in asthmatic mice was reversed by anti-CD40 treatment. Our data suggest that anti-CD1d treatment can suppress Th2 cell responses through inhibiting immunogenic maturation of LDCs dependent on lung iNKT cells, which couldbe partially related to the downregulation of CD40L expression on lung iNKT cells in asthmatic mice.
Collapse
Affiliation(s)
- Qibin Lin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Tong Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Xiaoshu Zuo
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Haiyang Ni
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Jieying Zhong
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hong Cheng
- Department of Parmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Yi Huang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Xuhong Ding
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hongying Yu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China.
| |
Collapse
|
5
|
Choi YA, Dhakal H, Lee S, Kim N, Lee B, Kwon TK, Khang D, Kim SH. IRF3 Activation in Mast Cells Promotes FcεRI-Mediated Allergic Inflammation. Cells 2023; 12:1493. [PMID: 37296614 PMCID: PMC10252328 DOI: 10.3390/cells12111493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
(1) Background: This study aims to elucidate a novel non-transcriptional action of IRF3 in addition to its role as a transcription factor in mast cell activation and associated allergic inflammation; (2) Methods: For in vitro experiments, mouse bone-marrow-derived mast cells (mBMMCs) and a rat basophilic leukemia cell line (RBL-2H3) were used for investigating the underlying mechanism of IRF3 in mast-cell-mediated allergic inflammation. For in vivo experiments, wild-type and Irf3 knockout mice were used for evaluating IgE-mediated local and systemic anaphylaxis; (3) Results: Passive cutaneous anaphylaxis (PCA)-induced tissues showed highly increased IRF3 activity. In addition, the activation of IRF3 was observed in DNP-HSA-treated mast cells. Phosphorylated IRF3 by DNP-HSA was spatially co-localized with tryptase according to the mast cell activation process, and FcεRI-mediated signaling pathways directly regulated that activity. The alteration of IRF3 affected the production of granule contents in the mast cells and the anaphylaxis responses, including PCA- and ovalbumin-induced active systemic anaphylaxis. Furthermore, IRF3 influenced the post-translational processing of histidine decarboxylase (HDC), which is required for granule maturation; and (4) Conclusion: Through this study, we demonstrated the novel function of IRF3 as an important factor inducing mast cell activation and as an upstream molecule for HDC activity.
Collapse
Affiliation(s)
- Young-Ae Choi
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Hima Dhakal
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea;
| | - Namkyung Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea;
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| |
Collapse
|
6
|
Tamagawa-Mineoka R, Ueta M, Arakawa Y, Nakanishi M, Nishigaki H, Katoh N. Roles of interferon regulatory factor 3 in skin inflammation: Possible involvement of regulatory mechanisms. Exp Dermatol 2023; 32:715-717. [PMID: 36933947 DOI: 10.1111/exd.14792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/17/2023] [Accepted: 03/06/2023] [Indexed: 03/20/2023]
Affiliation(s)
- Risa Tamagawa-Mineoka
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mayumi Ueta
- Department of Ophthalmology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukiyasu Arakawa
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mari Nakanishi
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiromi Nishigaki
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Norito Katoh
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
7
|
Morozova AA, Kosyakova NI, Prokhorenko IR. Participation of MAPK and PI3K in Regulation of Cytokine Secretion by Peripheral Blood Monocular Cells in Response to Escherichia coli LPS and rDer p 2 Combination. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:538-547. [PMID: 35790413 DOI: 10.1134/s0006297922060050] [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: 11/10/2021] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 06/15/2023]
Abstract
Search for the effective approaches to treat acute inflammation caused by combination of allergens and infectious agents is an important task for public health worldwide. House dust mites Dermatophagoides pteronyssinus are the source of allergens of the Der p groups and of microbial compounds, in particular, lipopolysaccharides (LPS). LPS and Der p 2 induce secretion of pro-inflammatory cytokines via activation of kinases p38 MAPK, MEK1/2, and PI3K. Participation of these kinases in the regulation of cells response to combined exposure to LPS and Der p 2 has not been sufficiently studied. We studied the effects of kinases (p38 MAPK, MEK1/2, and PI3K) inhibition on secretion of cytokines (TNF, IL-8, and IL-6) by peripheral blood mononuclear cells (PBMC) of healthy volunteers in response to E. coli LPS and rDer p 2. Contribution of kinases to the regulation of cell response to different agents (rDer p 2 and/or LPS) was revealed. It was found that p38 MAPK plays a key role in the regulation of secretion TNF by PBMC in response to the combination of LPS and rDer p 2. MEK1/2-dependent signaling is the main pathway for the synthesis of TNF and IL-8 in response to LPS and rDer p 2. PI3K-dependent signaling negatively regulates TNF production during rDer p 2-induced cell activation, but is not involved in the response to the combination of LPS and rDer p 2. PI3K-dependent signaling in the regulation of PBMC cytokine synthesis is most pronounced in response to their activation by rDer p 2. Understanding the mechanisms of immune cell responses to combinations of inflammatory agents could facilitate the search for new intracellular targets for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Anastasia A Morozova
- Hospital of Pushchino Scientific Center, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
- Institute of Basic Biological Problems, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Ninel I Kosyakova
- Hospital of Pushchino Scientific Center, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Isabella R Prokhorenko
- Institute of Basic Biological Problems, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
8
|
Chang Y, Kang JS, Jung K, Chung DH, Ha SJ, Kim YJ, Kim HY. OASL1-Mediated Inhibition of Type I IFN Reduces Influenza A Infection-Induced Airway Inflammation by Regulating ILC2s. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:99-116. [PMID: 34983110 PMCID: PMC8724833 DOI: 10.4168/aair.2022.14.1.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/27/2021] [Accepted: 10/08/2021] [Indexed: 11/20/2022]
Abstract
Purpose Three observations drove this study. First, 2′-5′-oligoadenylate synthetase-like protein (OASL) is a negative regulator of type I interferon (IFN). Second, type I IFN plays a central role during virus infections and the pathogenesis of various diseases, including asthma. Third, influenza A virus (IAV) causes non-eosinophilic asthma. To evaluate the potential relationships between OASL, type I IFN, and pulmonary innate immune cells in IAV-induced acute airway inflammation by using Oasl1-/- mice. Methods Asthma was induced in wild-type (WT) and Oasl1-/- mice with IAV or ovalbumin (OVA). Airway hyperreactivity (AHR) and immune cell infiltration in the bronchoalveolar lavage (BAL) fluids were measured. The immune cells in the lungs were analyzed by flow cytometry. To investigate the ability of type I IFN to shape the response of lung type 2 innate lymphoid cells (ILC2s), IFN-α was treated intratracheally. Plasmacytoid dendritic cells (pDCs) sorted from bone marrow and ILC2s sorted from lungs of naive mice were co-cultured with/without interferon-alpha receptor subunit 1 (IFNAR-1)-blocking antibodies. Results In the IAV-induced asthma model, Oasl1-/- mice developed greater AHR and immune cell infiltration in the BAL fluids than WT mice. This was not observed in OVA-induced asthma, a standard model of allergen-induced asthma. The lungs of infected Oasl1-/- mice also had elevated DC numbers and Ifna expression and depressed IAV-induced ILC2 responses, namely, proliferation and type 2 cytokine and amphiregulin production. Intratracheal administration of type I IFN in naïve mice suppressed lung ILC2 production of type 2 cytokines and amphiregulin. Co-culture of ILC2s with pDCs showed that pDCs inhibit the function of ILC2s by secreting type I IFN. Conclusions OASL1 may impede the IAV-induced acute airway inflammation that drives AHR by inhibiting IAV-induced type I IFN production from lung DCs, thereby preserving the functions of lung ILC2s, including their amphiregulin production.
Collapse
Affiliation(s)
- Yuna Chang
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Seon Kang
- Genome Research Center, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Department for Integrated OMICs for Biomedical Science, Yonsei University, Seoul, Korea
| | - Keehoon Jung
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Jun Ha
- System Immunology Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Young-Joon Kim
- Genome Research Center, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Department for Integrated OMICs for Biomedical Science, Yonsei University, Seoul, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
9
|
Petro TM. IFN Regulatory Factor 3 in Health and Disease. THE JOURNAL OF IMMUNOLOGY 2021; 205:1981-1989. [PMID: 33020188 DOI: 10.4049/jimmunol.2000462] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022]
Abstract
Immunity to viruses requires an array of critical cellular proteins that include IFN regulatory factor 3 (IRF3). Consequently, most viruses that infect vertebrates encode proteins that interfere with IRF3 activation. This review describes the cellular pathways linked to IRF3 activation and where those pathways are targeted by human viral pathogens. Moreover, key regulatory pathways that control IRF3 are discussed. Besides viral infections, IRF3 is also involved in resistance to some bacterial infections, in anticancer immunity, and in anticancer therapies involving DNA damage agents. A recent finding shows that IRF3 is needed for T cell effector functions that are involved in anticancer immunity and also in T cell autoimmune diseases. In contrast, unregulated IRF3 activity is clearly not beneficial, considering it is implicated in certain interferonopathies, in which heightened IRF3 activity leads to IFN-β-induced disease. Therefore, IRF3 is involved largely in maintaining health but sometimes contributing to disease.
Collapse
Affiliation(s)
- Thomas M Petro
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583; and Nebraska Center for Virology, University of Nebraska Medical Center, Lincoln, NE 68583
| |
Collapse
|
10
|
Mosayyebi S, Sarac BE, Akel Bilgic H, Sahiner UM, Sackesen C, Kalayci O, Karaaslan C. The Genetic Variants of Interferon Regulatory Factor-3 in Children with Asthma. J Interferon Cytokine Res 2020; 40:570-577. [PMID: 33337935 DOI: 10.1089/jir.2020.0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interferon Regulatory Factor-3 (IRF-3) is one of the key players in the inflammatory response mediated by the innate immune system. Although many studies have implicated a role for IRF-3 in the pathogenesis of inflammatory airway diseases, information about the possible association of IRF-3 genetic variants with asthma is scarce. We aimed to investigate the potential effects of IRF-3 polymorphisms in childhood asthma and asthma-related phenotypes. IRF-3 polymorphisms were first determined by sequencing 25 asthmatic and 25 healthy children. For further analysis, 609 asthmatic children and 191 healthy controls were screened for the genetic variants, such as rs2304204, rs2304205, rs320440, rs34739574, and rs7251. In addition, the relationship between these polymorphisms and asthma-related phenotypic features, including forced expiratory volume in one second values, eosinophil counts, and IgE levels was determined. rs7251 was associated with asthma in the codominant (P = 0.049) and G dominant (P = 0.025) model, however this significance was lost after False Discovery Rate analysis. Other investigated single nucleotide polymorphisms (SNPs) showed no significant association with asthma or asthma-related phenotypes. In conclusion, the seven SNPs of IRF-3 gene are not associated with asthma or asthma-related phenotypes in Turkish asthmatic children.
Collapse
Affiliation(s)
- Solmaz Mosayyebi
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Basak Ezgi Sarac
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Hayriye Akel Bilgic
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Umit Murat Sahiner
- Department of Pediatric Allergy, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Cansın Sackesen
- Department of Pediatric Allergy, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,Division of Pediatric Allergy, School of Medicine, Koc University, Istanbul, Turkey
| | - Omer Kalayci
- Department of Pediatric Allergy, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| |
Collapse
|
11
|
A Comparative Usability Study of Bare Hand Three-Dimensional Object Selection Techniques in Virtual Environment. Symmetry (Basel) 2020. [DOI: 10.3390/sym12101723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Object selection is the basis of natural user–computer interaction (NUI) in a virtual environment (VE). Among the three-dimensional object selection techniques employed in virtual reality (VR), bare hand-based finger clicking interaction and ray-casting are two convenient approaches with a high level of acceptance. This study involved 14 participants, constructed a virtual laboratory environment in VR, and compared the above two finger-based interaction techniques in terms of aspects of the task performance, including the success rate, total reaction time, operational deviation, and accuracy, at different spatial positions. The results indicated that the applicable distance range of finger clicking interaction and finger ray-casting was 0.2 to 1.4 m and over 0.4 m, respectively. Within the shared applicable distance, the finger clicking interaction achieved a shorter total reaction time and higher clicking accuracy. The performance of finger clicking interaction varied remarkably at the center and edge of the horizontal field of view, while no significant difference was found among ray-casting at various horizontal azimuths. The current findings could be directly applied to the application of bare-hand interaction in VR environments.
Collapse
|
12
|
Lejeune S, Deschildre A, Le Rouzic O, Engelmann I, Dessein R, Pichavant M, Gosset P. Childhood asthma heterogeneity at the era of precision medicine: Modulating the immune response or the microbiota for the management of asthma attack. Biochem Pharmacol 2020; 179:114046. [PMID: 32446884 PMCID: PMC7242211 DOI: 10.1016/j.bcp.2020.114046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Exacerbations are a main characteristic of asthma. In childhood, the risk is increasing with severity. Exacerbations are a strong phenotypic marker, particularly of severe and therapy-resistant asthma. These early-life events may influence the evolution and be involved in lung function decline. In children, asthma attacks are facilitated by exposure to allergens and pollutants, but are mainly triggered by microbial agents. Multiple studies have assessed immune responses to viruses, and to a lesser extend bacteria, during asthma exacerbation. Research has identified impairment of innate immune responses in children, related to altered pathogen recognition, interferon release, or anti-viral response. Influence of this host-microbiota dialog on the adaptive immune response may be crucial, leading to the development of biased T helper (Th)2 inflammation. These dynamic interactions may impact the presentations of asthma attacks, and have long-term consequences. The aim of this review is to synthesize studies exploring immune mechanisms impairment against viruses and bacteria promoting asthma attacks in children. The potential influence of the nature of infectious agents and/or preexisting microbiota on the development of exacerbation is also addressed. We then discuss our understanding of how these diverse host-microbiota interactions in children may account for the heterogeneity of endotypes and clinical presentations. Finally, improving the knowledge of the pathophysiological processes induced by infections has led to offer new opportunities for the development of preventive or curative therapeutics for acute asthma. A better definition of asthma endotypes associated with precision medicine might lead to substantial progress in the management of severe childhood asthma.
Collapse
Affiliation(s)
- Stéphanie Lejeune
- CHU Lille, Univ. Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, F-59000 Lille, France; Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Antoine Deschildre
- CHU Lille, Univ. Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, F-59000 Lille, France; Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Olivier Le Rouzic
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France; CHU Lille, Univ. Lille, Department of Respiratory Diseases, F-59000 Lille Cedex, France
| | - Ilka Engelmann
- Univ. Lille, Virology Laboratory, EA3610, Institute of Microbiology, CHU Lille, F-59037 Lille Cedex, France
| | - Rodrigue Dessein
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France; Univ. Lille, Bacteriology Department, Institute of Microbiology, CHU Lille, F-59037 Lille Cedex, France
| | - Muriel Pichavant
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Philippe Gosset
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France.
| |
Collapse
|
13
|
IRF3 and IRF7 contribute to diesel exhaust particles‐induced pulmonary inflammation by mediating mTORC1 activation and restraining autophagy in mice. Eur J Immunol 2020; 50:1142-1153. [DOI: 10.1002/eji.201948415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/10/2020] [Accepted: 03/03/2020] [Indexed: 01/29/2023]
|
14
|
He J, Yang Q, Xiao Q, Lei A, Li X, Zhou P, Liu T, Zhang L, Shi K, Yang Q, Dong J, Zhou J. IRF-7 Is a Critical Regulator of Type 2 Innate Lymphoid Cells in Allergic Airway Inflammation. Cell Rep 2019; 29:2718-2730.e6. [PMID: 31775040 DOI: 10.1016/j.celrep.2019.10.077] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/16/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Allergic asthma is a highly prevalent airway disease triggered by hyperresponsiveness to inhaled allergens. Interferon regulatory factor 7 (IRF7) has been shown to be highly expressed in nasal aspirates from children with asthma. Type 2 innate lymphoid cells (ILC2s) represent the major player in allergic airway inflammation. The role of IRF7 in ILC2-driven asthma remains to be explored. Here, we report that IRF7 expression in murine lung ILC2s is dramatically induced upon papain or interleukin-33 (IL-33) stimulation. ILC2s from asthma patients display a much higher level of IRF7 than those from healthy donors. Deficiency of IRF7 in mice significantly impairs the expansion and function of lung ILC2s in multiple models of allergic asthma. Furthermore, the regulation of ILC2s by IRF7 is cell intrinsic and mediated by the transcription factor Bcl11b. These observations identify IRF7 as a regulator of lung ILC2s, which may have immunotherapeutic value in allergic asthma.
Collapse
Affiliation(s)
- Juan He
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiong Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xiao
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Aihua Lei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pan Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Zhang
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Kun Shi
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Junchao Dong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Radermecker C, Sabatel C, Vanwinge C, Ruscitti C, Maréchal P, Perin F, Schyns J, Rocks N, Toussaint M, Cataldo D, Johnston SL, Bureau F, Marichal T. Locally instructed CXCR4 hi neutrophils trigger environment-driven allergic asthma through the release of neutrophil extracellular traps. Nat Immunol 2019; 20:1444-1455. [PMID: 31591573 PMCID: PMC6859073 DOI: 10.1038/s41590-019-0496-9] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Low exposure to microbial products, respiratory viral infections and air pollution is a major risk factor for allergic asthma, yet the mechanistic links between such conditions and host susceptibility to type 2 allergic disorders remain unclear. Through the use of single-cell RNA sequencing (scRNA-seq), we characterized lung neutrophils in mice exposed to a pro-allergic, low dose of lipopolysaccharides (LPSlo) or a protective, high dose of LPS (LPShi) before exposure to house dust mite (HDM). Unlike exposure to LPShi, exposure to LPSlo instructed recruited neutrophils to upregulate the expression of the chemokine receptor CXCR4 and to release neutrophil extracellular traps (NETs). The LPSlo-induced neutrophils and NETs potentiated the uptake of HDM by CD11b+Ly-6C+ dendritic cells (DCs) and type 2 allergic airway inflammation in response to HDM. NETs derived from CXCR4hi neutrophils were also needed to mediate allergic asthma triggered by infection with influenza virus or exposure to ozone. Our study indicates that apparently unrelated environmental risk factors can shape recruited lung neutrophils to promote the initiation of allergic asthma.
Collapse
Affiliation(s)
- Coraline Radermecker
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Catherine Sabatel
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Céline Vanwinge
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Cecilia Ruscitti
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Pauline Maréchal
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Fabienne Perin
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Joey Schyns
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium.,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Natacha Rocks
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Marie Toussaint
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK.,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Didier Cataldo
- Laboratory of Tumor and Development Biology, GIGA Institute, Liege University, Liege, Belgium
| | - Sebastian L Johnston
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK.,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.,Imperial College Healthcare National Health Service Trust, London, UK
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium. .,Faculty of Veterinary Medicine, Liege University, Liege, Belgium. .,Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium.
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, Liege University, Liege, Belgium. .,Laboratory of Immunophysiology, GIGA Institute, Liege University, Liege, Belgium. .,Faculty of Veterinary Medicine, Liege University, Liege, Belgium. .,Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium.
| |
Collapse
|
16
|
She L, Alanazi HH, Yan L, Zou Y, Sun Y, Dube PH, Brooks EG, Barrera GD, Lai Z, Chen Y, Liu Y, Zhang X, Li XD. Immune Sensing of Aeroallergen-Associated Double-Stranded RNA Triggers an IFN Response and Modulates Type 2 Lung Inflammation. THE JOURNAL OF IMMUNOLOGY 2019; 203:2520-2531. [PMID: 31562213 DOI: 10.4049/jimmunol.1900720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/26/2019] [Indexed: 01/10/2023]
Abstract
The innate immune sensing of allergens or allergen-associated components regulate the development of type 2 inflammatory responses. However, the underlying molecular basis by which allergens or allergen-associated components are detected by innate immune receptors remains elusive. In this study, we report that the most common aeroallergen, house dust mite (HDM), harbors a dsRNA species (HDM-dsRNA) that can activate TLR3-mediated IFN responses and counteract the development of an uncontrolled type 2 immune response. We demonstrate that the mouse strains defective in the dsRNA-sensing pathways show aggravated type 2 inflammation defined by severe eosinophilia, elevated level of type 2 cytokines, and mucus overproduction in a model of allergic lung inflammation. The inability to sense HDM-dsRNA resulted in significant increases in airway hyperreactivity. We further show that the administration of the purified HDM-dsRNA at a low dose is sufficient to induce an immune response to prevent the onset of a severe type 2 lung inflammation. Collectively, these results unveil a new role for the HDM-dsRNA/TLR3-signaling axis in the modulation of a type 2 lung inflammation in mice.
Collapse
Affiliation(s)
- Li She
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229.,Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hamad H Alanazi
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Liping Yan
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Yi Zou
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229; and
| | - Yilun Sun
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Peter H Dube
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Edward G Brooks
- Division of Immunology and Infectious Disease, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Gema D Barrera
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229; and
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229; and
| | - Yong Liu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xin Zhang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiao-Dong Li
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229;
| |
Collapse
|
17
|
Ozasa K, Temizoz B, Kusakabe T, Kobari S, Momota M, Coban C, Ito S, Kobiyama K, Kuroda E, Ishii KJ. Cyclic GMP-AMP Triggers Asthma in an IL-33-Dependent Manner That Is Blocked by Amlexanox, a TBK1 Inhibitor. Front Immunol 2019; 10:2212. [PMID: 31616416 PMCID: PMC6775192 DOI: 10.3389/fimmu.2019.02212] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/02/2019] [Indexed: 01/01/2023] Open
Abstract
Extracellular host-derived DNA, as one of damage associated molecular patterns (DAMPs), is associated with allergic type 2 immune responses. Immune recognition of such DNA generates the second messenger cyclic GMP-AMP (cGAMP) and induces type-2 immune responses; however, its role in allergic diseases, such as asthma, has not been fully elucidated. This study aimed to determine whether cGAMP could induce asthma when used as an adjuvant. We intranasally sensitized mice with cGAMP together with house dust mite antigen (HDM), followed by airway challenge with HDM. We then assessed the levels of eosinophils in the broncho-alveolar lavage fluid (BALF) and serum HDM-specific antibodies. cGAMP promoted HDM specific allergic asthma, characterized by significantly increased HDM specific IgG1 and total IgE in the serum and infiltration of eosinophils in the BALF. cGAMP stimulated lung fibroblast cells to produce IL-33 in vitro, and mice deficient for IL-33 or IL-33 receptor (ST2) failed to develop asthma enhancement by cGAMP. Not only Il-33 -/- mice, but also Sting -/-, Tbk1 -/-, and Irf3 -/- Irf7 -/- mice which lack the cGAMP-mediated innate immune activation failed to increase eosinophils in the BALF than that from wild type mice. Consistently, intranasal and oral administration of amlexanox, a TBK1 inhibitor, decreased cGAMP-induced lung allergic inflammation. Thus, cGAMP functions as a type 2 adjuvant in the lung and can promote allergic asthma in manners that dependent on the intracellular STING/TBK1/IRF3/7 signaling pathway and the resultant intercellular signaling pathway via IL-33 and ST2 might be a novel therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Koji Ozasa
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Burcu Temizoz
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takato Kusakabe
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shingo Kobari
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masatoshi Momota
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cevayir Coban
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Malaria Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuichi Ito
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
18
|
Tibbitt CA, Stark JM, Martens L, Ma J, Mold JE, Deswarte K, Oliynyk G, Feng X, Lambrecht BN, De Bleser P, Nylén S, Hammad H, Arsenian Henriksson M, Saeys Y, Coquet JM. Single-Cell RNA Sequencing of the T Helper Cell Response to House Dust Mites Defines a Distinct Gene Expression Signature in Airway Th2 Cells. Immunity 2019; 51:169-184.e5. [PMID: 31231035 DOI: 10.1016/j.immuni.2019.05.014] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/28/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
Naive CD4+ T cells differentiate into functionally diverse T helper (Th) cell subsets. Th2 cells play a pathogenic role in asthma, yet a clear picture of their transcriptional profile is lacking. We performed single-cell RNA sequencing (scRNA-seq) of T helper cells from lymph node, lung, and airways in the house dust mite (HDM) model of allergic airway disease. scRNA-seq resolved transcriptional profiles of naive CD4+ T, Th1, Th2, regulatory T (Treg) cells, and a CD4+ T cell population responsive to type I interferons. Th2 cells in the airways were enriched for transcription of many genes, including Cd200r1, Il6, Plac8, and Igfbp7, and their mRNA profile was supported by analysis of chromatin accessibility and flow cytometry. Pathways associated with lipid metabolism were enriched in Th2 cells, and experiments with inhibitors of key metabolic pathways supported roles for glucose and lipid metabolism. These findings provide insight into the differentiation of pathogenic Th2 cells in the context of allergy.
Collapse
Affiliation(s)
| | - Julian Mario Stark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Liesbet Martens
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Junjie Ma
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Jeff Eron Mold
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Kim Deswarte
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Ganna Oliynyk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Xiaogang Feng
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Bart Norbert Lambrecht
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Pieter De Bleser
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | | | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Jonathan Marie Coquet
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden.
| |
Collapse
|
19
|
Burleson JD, Siniard D, Yadagiri VK, Chen X, Weirauch MT, Ruff BP, Brandt EB, Hershey GKK, Ji H. TET1 contributes to allergic airway inflammation and regulates interferon and aryl hydrocarbon receptor signaling pathways in bronchial epithelial cells. Sci Rep 2019; 9:7361. [PMID: 31089182 PMCID: PMC6517446 DOI: 10.1038/s41598-019-43767-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 05/01/2019] [Indexed: 01/10/2023] Open
Abstract
Previous studies have suggested a role for Tet1 in the pathogenesis of childhood asthma. However, how Tet1 contributes to asthma remains unknown. Here we used mice deficient for Tet1 in a well-established model of allergic airway inflammation and demonstrated that loss of Tet1 increased disease severity including airway hyperresponsiveness and lung eosinophilia. Increased expression of Muc5ac, Il13, Il33, Il17a, Egfr, and Tff2 were observed in HDM-challenged Tet1-deficient mice compared to Tet1+/+ littermates. Further, transcriptomic analysis of lung RNA followed by pathway and protein network analysis showed that the IFN signaling pathway was significantly upregulated and the aryl hydrocarbon receptor (AhR) pathway was significantly downregulated in HDM-challenged Tet1-/- mice. This transcriptional regulation of the IFN and AhR pathways by Tet1 was also present in human bronchial epithelial cells at base line and following HDM challenges. Genes in these pathways were further associated with changes in DNA methylation, predicted binding of transcriptional factors with relevant functions in their promoters, and the presence of histone marks generated by histone enzymes that are known to interact with Tet1. Collectively, our data suggest that Tet1 inhibits HDM-induced allergic airway inflammation by direct regulation of the IFN and AhR pathways.
Collapse
Affiliation(s)
- J D Burleson
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dylan Siniard
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Pyrosequencing lab for genomic and epigenomic research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Veda K Yadagiri
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brandy P Ruff
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eric B Brandt
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hong Ji
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Pyrosequencing lab for genomic and epigenomic research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, USA. .,California National Primate Research Center, Davis, CA, USA.
| |
Collapse
|
20
|
Hu Q, Gilley RP, Dube PH. House dust mite exposure attenuates influenza A infection in a mouse model of pulmonary allergic inflammation. Microb Pathog 2019; 129:242-249. [PMID: 30776411 DOI: 10.1016/j.micpath.2019.02.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 11/16/2022]
Abstract
Environmental allergens elicit complex immune responses in the lungs that can promote the development of asthma or exacerbate preexisting asthma in susceptible individuals. House dust mites are one of the most common indoor allergens and are a significant driver of allergic disease. Respiratory infections are known factors in acute exacerbations of asthma but the impact of allergen on the pathogen is not well understood. We investigated the pathogenesis of influenza A infection following exposure to house dust mites. Mice exposed to house dust mites lose less weight following infection and had more transcription of interferon-lambda than controls. These data correlated with less transcription of the influenza polymerase acidic gene suggesting diminished viral replication in house dust mite exposed mice. Altogether, these data suggest that exposure to environmental allergens can influence the pathogenesis of influenza infection.
Collapse
Affiliation(s)
- Qiyao Hu
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410000, PR China
| | - Ryan P Gilley
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Peter H Dube
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
21
|
Machelart A, Potemberg G, Van Maele L, Demars A, Lagneaux M, De Trez C, Sabatel C, Bureau F, De Prins S, Percier P, Denis O, Jurion F, Romano M, Vanderwinden JM, Letesson JJ, Muraille E. Allergic Asthma Favors Brucella Growth in the Lungs of Infected Mice. Front Immunol 2018; 9:1856. [PMID: 30147700 PMCID: PMC6095999 DOI: 10.3389/fimmu.2018.01856] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
Allergic asthma is a chronic Th2 inflammatory disease of the lower airways affecting a growing number of people worldwide. The impact of infections and microbiota composition on allergic asthma has been investigated frequently. Until now, however, there have been few attempts to investigate the impact of asthma on the control of infectious microorganisms and the underlying mechanisms. In this work, we characterize the consequences of allergic asthma on intranasal (i.n.) infection by Brucella bacteria in mice. We observed that i.n. sensitization with extracts of the house dust mite Dermatophagoides farinae or the mold Alternaria alternata (Alt) significantly increased the number of Brucella melitensis, Brucella suis, and Brucella abortus in the lungs of infected mice. Microscopic analysis showed dense aggregates of infected cells composed mainly of alveolar macrophages (CD11c+ F4/80+ MHCII+) surrounded by neutrophils (Ly-6G+). Asthma-induced Brucella susceptibility appears to be dependent on CD4+ T cells, the IL-4/STAT6 signaling pathway and IL-10, and is maintained in IL-12- and IFN-γR-deficient mice. The effects of the Alt sensitization protocol were also tested on Streptococcus pneumoniae and Mycobacterium tuberculosis pulmonary infections. Surprisingly, we observed that Alt sensitization strongly increases the survival of S. pneumoniae infected mice by a T cell and STAT6 independent signaling pathway. In contrast, the course of M. tuberculosis infection is not affected in the lungs of sensitized mice. Our work demonstrates that the impact of the same allergic sensitization protocol can be neutral, negative, or positive with regard to the resistance of mice to bacterial infection, depending on the bacterial species.
Collapse
Affiliation(s)
- Arnaud Machelart
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
| | - Georges Potemberg
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
| | - Laurye Van Maele
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Aurore Demars
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
| | - Maxime Lagneaux
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
| | - Carl De Trez
- Department of Molecular and Cellular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Catherine Sabatel
- Laboratory of Cellular and Molecular Immunology, GIGA- Research & WELBIO, University of Liège, Liège, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA- Research & WELBIO, University of Liège, Liège, Belgium
| | - Sofie De Prins
- Immunology Unit, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | - Pauline Percier
- Immunology Unit, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | - Olivier Denis
- Immunology Unit, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | - Fabienne Jurion
- Immunology Unit, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | - Marta Romano
- Immunology Unit, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | | | - Jean-Jacques Letesson
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de Microbiologie, NAmur Research Institute for Life Sciences (NARILIS), Université de Namur, Namur, Belgium
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
22
|
Pellefigues C, Tang SC, Schmidt A, White RF, Lamiable O, Connor LM, Ruedl C, Dobrucki J, Le Gros G, Ronchese F. Toll-Like Receptor 4, but Not Neutrophil Extracellular Traps, Promote IFN Type I Expression to Enhance Th2 Responses to Nippostrongylus brasiliensis. Front Immunol 2017; 8:1575. [PMID: 29201030 PMCID: PMC5696323 DOI: 10.3389/fimmu.2017.01575] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022] Open
Abstract
The induction of Th2 responses is thought to be multifactorial, and emerge from specific pathways distinct from those associated with antagonistic antibacterial or antiviral Th1 responses. Here, we show that the recognition of non-viable Nippostrongylus brasiliensis (Nb) in the skin induces a strong recruitment of monocytes and neutrophils and the release of neutrophil extracellular traps (NETs). Nb also activates toll-like receptor 4 (TLR4) signaling with expression of Ifnb transcripts in the skin and the development of an IFN type I signature on helminth antigen-bearing dendritic cells in draining lymph nodes. Co-injection of Nb together with about 10,000 Gram-negative bacteria amplified this TLR4-dependent but NET-independent IFN type I response and enhanced the development of Th2 responses. Thus, a limited activation of antibacterial signaling pathways is able to boost antihelminthic responses, suggesting a role for bacterial sensing in the optimal induction of Th2 immunity.
Collapse
Affiliation(s)
| | | | - Alfonso Schmidt
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Ruby F White
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Lisa M Connor
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jurek Dobrucki
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biophysics, Jagiellonian University, Kraków, Poland
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
23
|
Exposure to Bacterial CpG DNA Protects from Airway Allergic Inflammation by Expanding Regulatory Lung Interstitial Macrophages. Immunity 2017; 46:457-473. [PMID: 28329706 DOI: 10.1016/j.immuni.2017.02.016] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/21/2016] [Accepted: 01/24/2017] [Indexed: 12/24/2022]
Abstract
Living in a microbe-rich environment reduces the risk of developing asthma. Exposure of humans or mice to unmethylated CpG DNA (CpG) from bacteria reproduces these protective effects, suggesting a major contribution of CpG to microbe-induced asthma resistance. However, how CpG confers protection remains elusive. We found that exposure to CpG expanded regulatory lung interstitial macrophages (IMs) from monocytes infiltrating the lung or mobilized from the spleen. Trafficking of IM precursors to the lung was independent of CCR2, a chemokine receptor required for monocyte mobilization from the bone marrow. Using a mouse model of allergic airway inflammation, we found that adoptive transfer of IMs isolated from CpG-treated mice recapitulated the protective effects of CpG when administered before allergen sensitization or challenge. IM-mediated protection was dependent on IL-10, given that Il10-/- CpG-induced IMs lacked regulatory effects. Thus, the expansion of regulatory lung IMs upon exposure to CpG might underlie the reduced risk of asthma development associated with a microbe-rich environment.
Collapse
|
24
|
Toussaint M, Jackson DJ, Swieboda D, Guedán A, Tsourouktsoglou TD, Ching YM, Radermecker C, Makrinioti H, Aniscenko J, Bartlett NW, Edwards MR, Solari R, Farnir F, Papayannopoulos V, Bureau F, Marichal T, Johnston SL. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat Med 2017; 23:681-691. [PMID: 28459437 PMCID: PMC5821220 DOI: 10.1038/nm.4332] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
Respiratory viral infections represent the most common cause of allergic asthma exacerbations. Amplification of the type-2 immune response is strongly implicated in asthma exacerbation, but how virus infection boosts type-2 responses is poorly understood. We report a significant correlation between the release of host double-stranded DNA (dsDNA) following rhinovirus infection and the exacerbation of type-2 allergic inflammation in humans. In a mouse model of allergic airway hypersensitivity, we show that rhinovirus infection triggers dsDNA release associated with the formation of neutrophil extracellular traps (NETs), known as NETosis. We further demonstrate that inhibiting NETosis by blocking neutrophil elastase or by degrading NETs with DNase protects mice from type-2 immunopathology. Furthermore, the injection of mouse genomic DNA alone is sufficient to recapitulate many features of rhinovirus-induced type-2 immune responses and asthma pathology. Thus, NETosis and its associated extracellular dsDNA contribute to the pathogenesis and may represent potential therapeutic targets of rhinovirus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Marie Toussaint
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- Guy's and St Thomas' NHS Trust, London, UK
| | - Dawid Swieboda
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Anabel Guedán
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - Yee Man Ching
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Coraline Radermecker
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Heidi Makrinioti
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Nathan W Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Michael R Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Roberto Solari
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Frédéric Farnir
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Fundamental and Applied Research for Animals &Health, University of Liège, Liège, Belgium
| | | | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
25
|
Lambrecht BN, Persson EK, Hammad H. Myeloid Cells in Asthma. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0053-2016. [PMID: 28102118 PMCID: PMC11687443 DOI: 10.1128/microbiolspec.mchd-0053-2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Indexed: 12/24/2022] Open
Abstract
Asthma is a heterogeneous chronic inflammatory disorder of the airways, and not surprisingly, many myeloid cells play a crucial role in pathogenesis. Antigen-presenting dendritic cells are the first to recognize the allergens, pollutants, and viruses that are implicated in asthma pathogenesis, and subsequently initiate the adaptive immune response by migrating to lymph nodes. Eosinophils are the hallmark of type 2 inflammation, releasing toxic compounds in the airways and contributing to airway remodeling. Mast cells and basophils control both the early- and late-phase allergic response and contribute to alterations in smooth muscle reactivity. Finally, relatively little is known about neutrophils and macrophages in this disease. Although many of these myeloid cells respond well to treatment with inhaled steroids, there is now an increasing armamentarium of targeted biologicals that can specifically eliminate only one myeloid cell population, like eosinophils. It is only with those new tools that we will be able to fully understand the role of myeloid cells in chronic asthma in humans.
Collapse
Affiliation(s)
- Bart N Lambrecht
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Emma K Persson
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
| | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| |
Collapse
|
26
|
Janss T, Mesnil C, Pirottin D, Lemaitre P, Marichal T, Bureau F, Desmet CJ. Interferon response factor-3 promotes the pro-Th2 activity of mouse lung CD11b + conventional dendritic cells in response to house dust mite allergens. Eur J Immunol 2016; 46:2614-2628. [PMID: 27546168 DOI: 10.1002/eji.201646513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/07/2016] [Accepted: 08/16/2016] [Indexed: 11/07/2022]
Abstract
Very few transcription factors have been identified that are required by antigen-presenting cells (APCs) to induce T helper type 2 (Th2) responses. Because lung CD11b+ conventional dendritic cells (CD11b+ cDCs) are responsible for priming Th2 responses in house-dust mite (HDM)-induced airway allergy, we used them as a model to identify transcriptional events regulating the pro-Th2 activity of cDCs. Transcriptomic profiling of lung CD11b+ cDCs exposed to HDM in vivo revealed first that HDM triggers an antiviral defence-like response, and second that the majority of HDM-induced transcriptional changes depend on the transcription factor Interferon Response Factor-3 (Irf3). Validating the functional relevance of these observations, Irf3-deficient CD11b+ cDCs displayed reduced pro-allergic activity. Indeed, Irf3-deficient CD11b+ cDCs induced less Th2, more regulatory T cell, and similar Th1 differentiation in naïve CD4+ T cells compared to their wild-type counterparts. The altered APC activity of Irf3 CD11b+ cDCs was associated with reduced expression of CD86 and was phenocopied by blocking CD86 activity in wild-type CD11b+ cDCs. Altogether, these results establish Irf3, known mostly for its role in antiviral responses, as a transcription factor involved in the induction of Th2 responses through the promotion of pro-Th2 costimulation in CD11b+ DCs.
Collapse
Affiliation(s)
- Thibaut Janss
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Claire Mesnil
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Dimitri Pirottin
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Pierre Lemaitre
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Christophe J Desmet
- Laboratory of Cellular and Molecular Immunology, GIGA-I3, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium.
| |
Collapse
|
27
|
Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA, Pirottin D, Janss T, Starkl P, Ramery E, Henket M, Schleich FN, Radermecker M, Thielemans K, Gillet L, Thiry M, Belvisi MG, Louis R, Desmet C, Marichal T, Bureau F. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 2016; 126:3279-95. [PMID: 27548519 DOI: 10.1172/jci85664] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Increases in eosinophil numbers are associated with infection and allergic diseases, including asthma, but there is also evidence that eosinophils contribute to homeostatic immune processes. In mice, the normal lung contains resident eosinophils (rEos), but their function has not been characterized. Here, we have reported that steady-state pulmonary rEos are IL-5-independent parenchymal Siglec-FintCD62L+CD101lo cells with a ring-shaped nucleus. During house dust mite-induced airway allergy, rEos features remained unchanged, and rEos were accompanied by recruited inflammatory eosinophils (iEos), which were defined as IL-5-dependent peribronchial Siglec-FhiCD62L-CD101hi cells with a segmented nucleus. Gene expression analyses revealed a more regulatory profile for rEos than for iEos, and correspondingly, mice lacking lung rEos showed an increase in Th2 cell responses to inhaled allergens. Such elevation of Th2 responses was linked to the ability of rEos, but not iEos, to inhibit the maturation, and therefore the pro-Th2 function, of allergen-loaded DCs. Finally, we determined that the parenchymal rEos found in nonasthmatic human lungs (Siglec-8+CD62L+IL-3Rlo cells) were phenotypically distinct from the iEos isolated from the sputa of eosinophilic asthmatic patients (Siglec-8+CD62LloIL-3Rhi cells), suggesting that our findings in mice are relevant to humans. In conclusion, our data define lung rEos as a distinct eosinophil subset with key homeostatic functions.
Collapse
|
28
|
Ysebrant de Lendonck L, Martinet V, Goriely S. Interferon regulatory factor 3 in adaptive immune responses. Cell Mol Life Sci 2014; 71:3873-83. [PMID: 24879293 PMCID: PMC11113752 DOI: 10.1007/s00018-014-1653-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/06/2014] [Accepted: 05/16/2014] [Indexed: 12/13/2022]
Abstract
Interferon regulatory factor (IRF) 3 plays a key role in innate responses against viruses. Indeed, activation of this transcription factor triggers the expression of type I interferons and downstream interferon-stimulated genes in infected cells. Recent evidences indicate that this pathway also modulates adaptive immune responses. This review focuses on the different mechanisms that are implicated in this process. We discuss the role of IRF3 within antigen-presenting cells and T lymphocytes in the polarization of the cellular immune response and its implication in the pathogenesis of immune disorders.
Collapse
Affiliation(s)
- Laure Ysebrant de Lendonck
- WELBIO and Institute for Medical Immunology (IMI), Université Libre de Bruxelles, 8 rue Adrienne Bolland, 6041 Charleroi-Gosselies, Belgium
| | - Valerie Martinet
- WELBIO and Institute for Medical Immunology (IMI), Université Libre de Bruxelles, 8 rue Adrienne Bolland, 6041 Charleroi-Gosselies, Belgium
| | - Stanislas Goriely
- WELBIO and Institute for Medical Immunology (IMI), Université Libre de Bruxelles, 8 rue Adrienne Bolland, 6041 Charleroi-Gosselies, Belgium
| |
Collapse
|
29
|
Innate immune cells in asthma. Trends Immunol 2013; 34:540-7. [PMID: 24035576 DOI: 10.1016/j.it.2013.08.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 12/17/2022]
Abstract
Asthma is an inflammatory disease of the airways associated with a T helper (Th)2 response. Such a response in the lungs requires complex interactions between innate cells and structural cells. Dendritic cells (DCs) are pivotal during sensitization to allergens but clearly require epithelium-derived signals to become activated. Epithelial cells also contribute to the activation and the survival of mast cells (MCs), basophils, and eosinophils and group 2 innate lymphoid cells (ILC2s). In turn, these innate cells can activate DCs to sustain Th2 immunity. Here, we review the role played by these different populations of immune cells in the pathogenesis of asthma and how they interact to orchestrate Th2 immunity.
Collapse
|
30
|
Murphy AA, Rosato PC, Parker ZM, Khalenkov A, Leib DA. Synergistic control of herpes simplex virus pathogenesis by IRF-3, and IRF-7 revealed through non-invasive bioluminescence imaging. Virology 2013; 444:71-9. [PMID: 23777662 DOI: 10.1016/j.virol.2013.05.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 12/13/2022]
Abstract
Interferon regulatory factors IRF-3 and IRF-7 are central to the establishment of the innate antiviral response. This study examines HSV-1 pathogenesis in IRF-3(-/-), IRF-7(-/-) and double-deleted IRF3/7(-/-) (DKO) mice. Bioluminescence imaging of infection revealed that DKO mice developed visceral infection following corneal inoculation, along with increased viral burdens in all tissues relative to single knockout mice. While all DKO mice synchronously reached endpoint criteria 5 days post infection, the IRF-7(-/-) mice survived longer, indicating that although IRF-7 is dominant, IRF-3 also plays a role in controlling disease. Higher levels of systemic pro-inflammatory cytokines were found in IRF7(-/-) and DKO mice relative to wild-type and IRF-3(-/-) mice, and IL-6 and G-CSF, indicative of sepsis, were increased in the DKO mice relative to wild-type or single-knockout mice. In addition to controlling viral replication, IRF-3 and -7 therefore play coordinating roles in modulation of inflammation during HSV infection.
Collapse
Affiliation(s)
- Aisling A Murphy
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, HB 7556, Lebanon, NH 03756, USA
| | | | | | | | | |
Collapse
|
31
|
Toussaint M, Fievez L, Drion PV, Cataldo D, Bureau F, Lekeux P, Desmet CJ. Myeloid hypoxia-inducible factor 1α prevents airway allergy in mice through macrophage-mediated immunoregulation. Mucosal Immunol 2013; 6:485-97. [PMID: 22968421 DOI: 10.1038/mi.2012.88] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypoxia-inducible factor (HIF) has important roles in promoting pro-inflammatory and bactericidal functions in myeloid cells. Conditional genetic ablation of its major subunit Hif1α in the myeloid lineage consequently results in decreased inflammatory responses in classical models of acute inflammation in mice. By contrast, we report here that mice conditionally deficient for Hif1α in myeloid cells display enhanced sensitivity to the development of airway allergy to experimental allergens and house-dust mite antigens. We support that upon allergen exposure, MyD88-dependent upregulation of Hif1α boosts the expression of the immunosuppressive cytokine interleukin (IL)-10 by lung interstitial macrophages (IMs). Hif1α-dependent IL-10 secretion is required for IMs to block allergen-induced dendritic cell activation and consequently for preventing the development of allergen-specific T-helper cell responses upon allergen exposure. Thus, this study supports that, in addition to its known pro-inflammatory activities, myeloid Hif1α possesses immunoregulatory functions implicated in the prevention of airway allergy.
Collapse
Affiliation(s)
- M Toussaint
- Laboratory of Cellular and Molecular Immunology, GIGA-Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | | | | | | | | | | | | |
Collapse
|
32
|
Krysko O, Maes T, Plantinga M, Holtappels G, Imiru R, Vandenabeele P, Joos G, Krysko DV, Bachert C. The adjuvant-like activity of staphylococcal enterotoxin B in a murine asthma model is independent of IL-1R signaling. Allergy 2013; 68:446-53. [PMID: 23347053 DOI: 10.1111/all.12102] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND Staphylococcal enterotoxin B (SEB) is a superantigen known to be a modulator of chronic airway inflammation in mice and humans, yet little is known about the mechanisms that regulate its interaction with the innate immune system. We investigated this mechanism in a murine model of allergic airway inflammation induced by OVA (ovalbumin) in the presence of SEB. METHODS Superantigen-induced allergic inflammation was studied in IL-1R knockout (KO) mice exposed to OVA+SEB. Multicolor flow cytometry was used to analyze the inflammatory cell profile in airways and lymph nodes. Production of IL-4, IL-5, IL-10, and IL-13 in lymph nodes was assessed by Luminex technology. RESULTS In wild-type mice, endonasal instillation of OVA+SEB induced a pulmonary inflammation, characterized by an increase in the number of eosinophils, T cells, and dendritic cells and in the production of Th2 cytokines and OVA-specific IgE. In IL-1R KO mice exposed to OVA+SEB, attraction of CD4+ cells and production of Th2 cytokines were reduced. However, knocking out IL-1R did not affect any of the features of allergic airway inflammation, such as bronchial eosinophilia, OVA-specific IgE production and goblet cell metaplasia. CONCLUSION We provide new insights into the mechanisms of airways allergy development in the presence of bacterial superantigen. The asthma features induced by OVA+SEB, such as bronchial eosinophilia, goblet cell proliferation, production of OVA-specific IgE and increase in inflammatory dendritic cells, are IL-1R independent. Yet, IL-1R signaling is crucial for CD4 cell accumulation and Th2 cytokine production.
Collapse
Affiliation(s)
- O. Krysko
- Upper Airway Research Laboratory; Department of Oto-Rhino-Laryngology; Ghent University; Ghent; Belgium
| | - T. Maes
- Department of Respiratory Medicine; Ghent University Hospital; Ghent; Belgium
| | - M. Plantinga
- Laboratory of Immunoregulation and Mucosal Immunology; Department of Respiratory Diseases; Ghent University Hospital; Ghent; Belgium
| | - G. Holtappels
- Upper Airway Research Laboratory; Department of Oto-Rhino-Laryngology; Ghent University; Ghent; Belgium
| | - R. Imiru
- Upper Airway Research Laboratory; Department of Oto-Rhino-Laryngology; Ghent University; Ghent; Belgium
| | | | - G. Joos
- Department of Respiratory Medicine; Ghent University Hospital; Ghent; Belgium
| | | | - C. Bachert
- Upper Airway Research Laboratory; Department of Oto-Rhino-Laryngology; Ghent University; Ghent; Belgium
| |
Collapse
|
33
|
Innate immune responses in house dust mite allergy. ISRN ALLERGY 2013; 2013:735031. [PMID: 23724247 PMCID: PMC3658386 DOI: 10.1155/2013/735031] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 11/22/2012] [Indexed: 12/20/2022]
Abstract
Sensitizations to house dust mites (HDM) trigger strong exacerbated allergen-induced inflammation of the skin and airways mucosa from atopic subjects resulting in atopic dermatitis as well as allergic rhinitis and asthma. Initially, the Th2-biased HDM allergic response was considered to be mediated only by allergen B- and T-cell epitopes to promote allergen-specific IgE production as well as IL-4, IL-5, and IL-13 to recruit inflammatory cells. But this general molecular model of HDM allergenicity must be revisited as a growing literature suggests that stimulations of innate immune activation pathways by HDM allergens offer new answers to the following question: what makes an HDM allergen an allergen? Indeed, HDM is a carrier not only for allergenic proteins but also microbial adjuvant compounds, both of which are able to stimulate innate signaling pathways leading to allergy. This paper will describe the multiple ways used by HDM allergens together with microbial compounds to control the initiation of the allergic response through engagement of innate immunity.
Collapse
|
34
|
Mesnil C, Sabatel CM, Marichal T, Toussaint M, Cataldo D, Drion PV, Lekeux P, Bureau F, Desmet CJ. Resident CD11b(+)Ly6C(-) lung dendritic cells are responsible for allergic airway sensitization to house dust mite in mice. PLoS One 2012; 7:e53242. [PMID: 23300898 PMCID: PMC3534017 DOI: 10.1371/journal.pone.0053242] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 11/27/2012] [Indexed: 01/17/2023] Open
Abstract
Conventional dendritic cells (DCs) are considered to be the prime initiators of airway allergy. Yet, it remains unclear whether specific DC subsets are preferentially involved in allergic airway sensitization. Here, we systematically assessed the respective pro-allergic potential of individually sorted lung DC subsets isolated from house dust mite antigen (HDM)-treated donor mice, following transfer to naïve recipients. Transfer of lung CD11c(+)CD11b(+) DCs, but not CD11c(+)CD11b(-)CD103(+) DCs, was sufficient to prime airway allergy. The CD11c(+)CD11b(+) DC subpopulation was composed of CD11c(+)CD11b(+)Ly6C(+) inflammatory monocyte-derived cells, whose numbers increase in the lungs following HDM exposure, and of CD11c(+)CD11b(+)Ly6C(-) DCs, which remain stable. Counterintuitively, only CD11c(+)CD11b(+)Ly6C(-) DCs, and not CD11c(+)CD11b(+)Ly6C(+) DCs, were able to convey antigen to the lymph nodes and induce adaptive T cell responses and subsequent airway allergy. Our results thus support that lung resident non-inflammatory CD11c(+)CD11b(+)Ly6C(-) DCs are the essential inducers of allergic airway sensitization to the common aeroallergen HDM in mice.
Collapse
Affiliation(s)
- Claire Mesnil
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Catherine M. Sabatel
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Marie Toussaint
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Didier Cataldo
- Laboratory of Tumours and Developmental Biology, GIGA-Research, University of Liège, Liège, Belgium
| | - Pierre-Vincent Drion
- Laboratory of Preclinical and Biomedical Sciences, University Hospital Center, University of Liège, Liège, Belgium
| | - Pierre Lekeux
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Christophe J. Desmet
- Laboratory of Cellular and Molecular Immunology, GIGA-Research and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
35
|
Wang JY. The innate immune response in house dust mite-induced allergic inflammation. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2012; 5:68-74. [PMID: 23450108 PMCID: PMC3579094 DOI: 10.4168/aair.2013.5.2.68] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/04/2012] [Indexed: 12/05/2022]
Abstract
Hypersensitivity to house dust mite (HDM; Dermatophagoides sp.) allergens is one of the most common allergic responses, affecting up to 85% of asthmatics. Sensitization to indoor allergens is the strongest independent risk factor associated with asthma. Additionally, >50% of children and adolescents with asthma are sensitized to HDM. Although allergen-specific CD4+ Th2 cells orchestrate the HDM allergic response through induction of IgE directed toward mite allergens, activation of innate immunity also plays a critical role in HDM-induced allergic inflammation. This review highlights the HDM components that lead to activation of the innate immune response. Activation may due to HDM proteases. Proteases may be recognized by protease-activation receptors (PARs), Toll-like receptors (TLRs), or C-type lectin receptors (CTRs), or act as a molecular mimic for PAMP activation signaling pathways. Understanding the role of mite allergen-induced innate immunity will facilitate the development of therapeutic strategies that exploit innate immunity receptors and associated signaling pathways for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Jiu-Yao Wang
- Division of Allergy and Clinical Immunology, Department of Pediatrics, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
36
|
Lambrecht BN, Hammad H. Lung dendritic cells in respiratory viral infection and asthma: from protection to immunopathology. Annu Rev Immunol 2012; 30:243-70. [PMID: 22224777 DOI: 10.1146/annurev-immunol-020711-075021] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lung dendritic cells (DCs) bridge innate and adaptive immunity, and depending on context, they also induce a Th1, Th2, or Th17 response to optimally clear infectious threats. Conversely, lung DCs can also mount maladaptive Th2 immune responses to harmless allergens and, in this way, contribute to immunopathology. It is now clear that the various aspects of DC biology can be understood only if we take into account the functional specializations of different DC subsets that are present in the lung in homeostasis or are attracted to the lung as part of the inflammatory response to inhaled noxious stimuli. Lung DCs are heavily influenced by the nearby epithelial cells, and a model is emerging whereby direct communication between DCs and epithelial cells determines the outcome of the pulmonary immune response. Here, we have approached DC biology from the perspective of viral infection and allergy to illustrate these emerging concepts.
Collapse
Affiliation(s)
- Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, Department of Molecular Biomedical Research, VIB, 9052 Ghent, Belgium.
| | | |
Collapse
|
37
|
Willart M, Hammad H. Lung dendritic cell-epithelial cell crosstalk in Th2 responses to allergens. Curr Opin Immunol 2011; 23:772-7. [PMID: 22074731 DOI: 10.1016/j.coi.2011.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 09/28/2011] [Indexed: 12/01/2022]
Abstract
Dendritic cells (DC) have been shown to be responsible for the initiation and maintenance of adaptive Th2 responses in asthma. It is increasingly clear that DC functions are strongly influenced by crosstalk with neighboring cells like epithelial cells, which can release a number of innate cytokines promoting Th2 responses. Clinically relevant allergens often interfere directly or indirectly with the innate immune functions of airway epithelial cells and DC. A better understanding of these interactions might lead to a better prevention and ultimately to new treatments for asthma.
Collapse
Affiliation(s)
- Monique Willart
- Laboratory of Immunoregulation and Mucosal Immunology, University of Ghent, De Pintelaan 185, 9000 Ghent, Belgium
| | | |
Collapse
|
38
|
DNA released from dying host cells mediates aluminum adjuvant activity. Nat Med 2011; 17:996-1002. [PMID: 21765404 DOI: 10.1038/nm.2403] [Citation(s) in RCA: 412] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 05/19/2011] [Indexed: 12/12/2022]
Abstract
Aluminum-based adjuvants (aluminum salts or alum) are widely used in human vaccination, although their mechanisms of action are poorly understood. Here we report that, in mice, alum causes cell death and the subsequent release of host cell DNA, which acts as a potent endogenous immunostimulatory signal mediating alum adjuvant activity. Furthermore, we propose that host DNA signaling differentially regulates IgE and IgG1 production after alum-adjuvanted immunization. We suggest that, on the one hand, host DNA induces primary B cell responses, including IgG1 production, through interferon response factor 3 (Irf3)-independent mechanisms. On the other hand, we suggest that host DNA also stimulates 'canonical' T helper type 2 (T(H)2) responses, associated with IgE isotype switching and peripheral effector responses, through Irf3-dependent mechanisms. The finding that host DNA released from dying cells acts as a damage-associated molecular pattern that mediates alum adjuvant activity may increase our understanding of the mechanisms of action of current vaccines and help in the design of new adjuvants.
Collapse
|
39
|
The role of innate immunity activation in house dust mite allergy. Trends Mol Med 2011; 17:604-11. [PMID: 21741880 DOI: 10.1016/j.molmed.2011.05.014] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/23/2011] [Accepted: 05/31/2011] [Indexed: 12/14/2022]
Abstract
House dust mite (HDM) allergy is a frequent inflammatory disease found worldwide. Although allergen-specific CD4(+) Th2 cells orchestrate the HDM allergic response, notably through induction of IgE directed towards mite allergens, recent studies have demonstrated that innate immunity activation also plays a critical role in HDM-induced allergy pathogenesis. HDM allergens can not only be considered proteins that induce adaptive Th2-biased responses in susceptible subjects but also as strong activators of innate immune cells, including skin keratinocytes and airway epithelial cells. The contribution of microbial adjuvant factors, derived from HDM carriers or the environment, is also essential in such cell stimulation. This review highlights how HDM allergens, together with microbial compounds, promote allergic responses through pattern recognition receptor-dependent pathways.
Collapse
|