201
|
Walker MT, Green JE, Ferrie RP, Queener AM, Kaplan MH, Cook-Mills JM. Mechanism for initiation of food allergy: Dependence on skin barrier mutations and environmental allergen costimulation. J Allergy Clin Immunol 2018; 141:1711-1725.e9. [PMID: 29454836 PMCID: PMC5938139 DOI: 10.1016/j.jaci.2018.02.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/23/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Mechanisms for the development of food allergy in neonates are unknown but clearly linked in patient populations to a genetic predisposition to skin barrier defects. Whether skin barrier defects contribute functionally to development of food allergy is unknown. OBJECTIVE The purpose of the study was to determine whether skin barrier mutations, which are primarily heterozygous in patient populations, contribute to the development of food allergy. METHODS Mice heterozygous for the filaggrin (Flg)ft and Tmem79ma mutations were skin sensitized with environmental and food allergens. After sensitization, mice received oral challenge with food allergen, and then inflammation, inflammatory mediators, and anaphylaxis were measured. RESULTS We define development of inflammation, inflammatory mediators, and food allergen-induced anaphylaxis in neonatal mice with skin barrier mutations after brief concurrent cutaneous exposure to food and environmental allergens. Moreover, neonates of allergic mothers have increased responses to suboptimal sensitization with food allergens. Importantly, responses to food allergens by these neonatal mice were dependent on genetic defects in skin barrier function and on exposure to environmental allergens. ST2 blockade during skin sensitization inhibited the development of anaphylaxis, antigen-specific IgE, and inflammatory mediators. Neonatal anaphylactic responses and antigen-specific IgE were also inhibited by oral pre-exposure to food allergen, but interestingly, this was blunted by concurrent pre-exposure of the skin to environmental allergen. CONCLUSION These studies uncover mechanisms for food allergy sensitization and anaphylaxis in neonatal mice that are consistent with features of human early-life exposures and genetics in patients with clinical food allergy and demonstrate that changes in barrier function drive development of anaphylaxis to food allergen.
Collapse
Affiliation(s)
- Matthew T Walker
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Jeremy E Green
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Ryan P Ferrie
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Ashley M Queener
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Mark H Kaplan
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Ind
| | - Joan M Cook-Mills
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
202
|
Yanagibashi T, Satoh M, Nagai Y, Koike M, Takatsu K. Allergic diseases: From bench to clinic - Contribution of the discovery of interleukin-5. Cytokine 2018; 98:59-70. [PMID: 28863833 DOI: 10.1016/j.cyto.2016.11.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 01/21/2023]
Abstract
T helper 2 cells produce a number of cytokines including inteleukin (IL)-5, IL-4 and IL-13. Group 2 innate lymphoid cells (ILC2s) also produce IL-5 under sterile conditions. IL-5 is interdigitating homodimeric glycoprotein and a member of the four α helical bundle motifs conserved among hematopoietic cytokines. IL-5 exerts its effects on target cells via IL-5 receptor (IL-5R), composed of an IL-5R α and βc subunit. The membrane proximal proline-rich motif of the cytoplasmic domain of both IL-5R α and βc subunits is essential for IL-5 signal transduction. Although IL-5 was initially identified by its ability to support the growth and terminal differentiation of mouse B cells into antibody-secreting cells, recombinant IL-5 exerts pleiotropic activities on various target cells. For example, IL-5 is now recognized as the major maturation and differentiation factor for eosinophils in mice and humans. Overexpression of IL-5 in mouse significantly increases eosinophil numbers and antibody levels in vivo, while mice lacking a functional gene for IL-5 or IL-5R display developmental and functional impairments in B cell and eosinophil lineages. In mice, the role of the IL-5/IL-5R system in the production and secretion of Immunoglobulin (Ig) M and IgA in mucosal tissues has been reported. Although eosinophils protect against invading pathogens including virus, bacteria and helminthes, they are also involved in the pathogenesis of various diseases, such as food allergy, asthma, and inflammatory bowel diseases. The recent expansion in our understanding in the context of IL-5 and IL-5-producing ILC2s in eosinophil activation and the pathogenesis of eosinophil-dependent inflammatory diseases has led to advances in therapeutic options. A new therapy currently under invetigarion in clinical trials uses humanized monoclonal antibodies against IL-5 or the IL-5R. In this review, we summarize our current understanding of the functions of IL-5 and its receptor, the innate regulation of IL-5-producing cells, and therapeutic potential of anti-IL-5 and anti-eosinophil (IL-5R) antibodies.
Collapse
Affiliation(s)
- Tsutomu Yanagibashi
- Toyama Prefectural Institute of Pharmaceutical Research, 17-1 Nakataikouyama, Imizu City, Toyama 939-0363, Japan; Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| | - Mitsuo Satoh
- Kyowa Hakko Kirin Co., Ltd., Otemachi Finamcial City Grand Cube, 1-9-2, Chiyoda-ku, Tokyo 100-8185, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan; JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Masamichi Koike
- Kyowa Hakko Kirin Co., Ltd., Otemachi Finamcial City Grand Cube, 1-9-2, Chiyoda-ku, Tokyo 100-8185, Japan
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute of Pharmaceutical Research, 17-1 Nakataikouyama, Imizu City, Toyama 939-0363, Japan; Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan.
| |
Collapse
|
203
|
The effects of resiquimod in an ovalbumin-induced allergic rhinitis model. Int Immunopharmacol 2018; 59:233-242. [PMID: 29665497 DOI: 10.1016/j.intimp.2018.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
Abstract
Growing evidence indicates that the Toll-like receptor7/8(TLR7/8) agonist resiquimod (R848) is a potential inhibitor of type-2 immunity. However, the mechanisms mediating its therapeutic effects are not fully understood. This study investigated the effects of R848 on OVA-induced allergic rhinitis(AR) mice and the expression of IL-25, IL-33, TSLP, T-cell immunoglobulin mucin1 (TIM1) and T-cell immunoglobulin mucin3 (TIM3). BALB/c mice were intranasally sensitized and challenged with ovalbumin (OVA), and R848 was intraperitoneally injected into AR mice. Histological changes in the nasal mucosa were evaluated by hematoxylin and eosin (H & E) and Periodic Acid-Schiff (PAS) staining; cytokine levels in serum were measured with enzyme-linked immunosorbent assays (ELISAs);the mRNA expression levels of IFN-γ, IL-17 and Foxp3 in the spleen determined by quantitative real-time RT-PCR (qRT-PCR); the proportions of Th1, Th2, Th17, Treg and TIM3 + IFN-γ + Th1 cells in the spleen were assessed with flow cytometry; TIM1, TIM3 and IL-33 expression levels in the nasal mucosa were evaluated with immunofluorescence staining(IF).R848 alleviated the nasal allergic symptoms; reduced eosinophil cell infiltration, goblet cell hyperplasia in the nasal mucosa; reduced IL-13, IL-17, IL-25 and IL-33 levels in serum; upregulated the relative mRNA expression of IFN-γ and Foxp3, and downregulated the relative mRNA expression of IL-17 in the spleen; decreased Th2, Th17 and TIM3 + IFN-γ + Th1 cells ratios, increased the proportion of Th1 and Treg cells in the spleen; suppressed TIM1 and TIM3,but increased IL-33 expression in the nasal mucosa in OVA-induced AR mice. R848 suppresses IL-25, IL-33 released and TIM1, TIM3 expression, which may contribute to its anti-allergic effects.
Collapse
|
204
|
Cai T, Qiu J, Ji Y, Li W, Ding Z, Suo C, Chang J, Wang J, He R, Qian Y, Guo X, Zhou L, Sheng H, Shen L, Qiu J. IL-17-producing ST2 + group 2 innate lymphoid cells play a pathogenic role in lung inflammation. J Allergy Clin Immunol 2018; 143:229-244.e9. [PMID: 29625134 DOI: 10.1016/j.jaci.2018.03.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/01/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND IL-17 plays a pathogenic role in asthma. ST2- inflammatory group 2 innate lymphoid cells (ILC2s) driven by IL-25 can produce IL-17, whereas ST2+ natural ILC2s produce little IL-17. OBJECTIVE We characterized ST2+IL-17+ ILC2s during lung inflammation and determined the pathogenesis and molecular regulation of ST2+IL-17+ ILC2s. METHODS Lung inflammation was induced by papain or IL-33. IL-17 production by lung ILC2s from wild-type, Rag1-/-, Rorcgfp/gfp, and aryl hydrocarbon receptor (Ahr)-/- mice was examined by using flow cytometry. Bone marrow transfer experiments were performed to evaluate hematopoietic myeloid differentiation primary response gene-88 (MyD88) signaling in regulating IL-17 production by ILC2s. mRNA expression of IL-17 was analyzed in purified naive ILC2s treated with IL-33, leukotrienes, and inhibitors for nuclear factor of activated T cells, p38, c-Jun N-terminal kinase, or nuclear factor κ light-chain enhancer of activated B cells. The pathogenesis of IL-17+ ILC2s was determined by transferring wild-type or Il17-/- ILC2s to Rag2-/-Il2rg-/- mice, which further induced lung inflammation. Finally, expression of 106 ILC2 signature genes was compared between ST2+IL-17+ ILC2s and ST2+IL-17- ILC2s. RESULTS Papain or IL-33 treatment boosted IL-17 production from ST2+ ILC2s (referred to by us as ILC217s) but not ST2- ILC2s. Ahr, but not retinoic acid receptor-related orphan receptor γt, facilitated the production of IL-17 by ILC217s. The hematopoietic compartment of MyD88 signaling is essential for ILC217 induction. IL-33 works in synergy with leukotrienes, which signal through nuclear factor of activated T-cell activation to promote IL-17 in ILC217s. Il17-/- ILC2s were less pathogenic in lung inflammation. ILC217s concomitantly expressed IL-5 and IL-13 but expressed little GM-CSF. CONCLUSION During lung inflammation, IL-33 and leukotrienes synergistically induce ILC217s. ILC217s are a highly pathogenic and unexpected source for IL-17 in lung inflammation.
Collapse
Affiliation(s)
- Ting Cai
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinxin Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Ji
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjing Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaoyun Ding
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Caixia Suo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiali Chang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jingjing Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Youcun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Fla
| | - Huiming Sheng
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
205
|
Schuijs MJ, Halim TYF. Group 2 innate lymphocytes at the interface between innate and adaptive immunity. Ann N Y Acad Sci 2018; 1417:87-103. [PMID: 29492980 DOI: 10.1111/nyas.13604] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/22/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022]
Abstract
Group 2 innate lymphoid cells (ILC2) are innate immune cells that respond rapidly to their environment through soluble inflammatory mediators and cell-to-cell interactions. As tissue-resident sentinels, ILC2 help orchestrate localized type 2 immune responses. These ILC2-driven type 2 responses are now recognized in diverse immune processes, different anatomical locations, and homeostatic or pathological settings. ILC2-derived cytokines and cell surface signaling molecules function as key regulators of innate and adaptive immunity. Conversely, ILC2 are governed by their environment. As such, ILC2 form an important nexus of the immune system and may present an attractive target for immune modulation in disease.
Collapse
|
206
|
Salimi M, Wang R, Yao X, Li X, Wang X, Hu Y, Chang X, Fan P, Dong T, Ogg G. Activated innate lymphoid cell populations accumulate in human tumour tissues. BMC Cancer 2018; 18:341. [PMID: 29587679 PMCID: PMC5870240 DOI: 10.1186/s12885-018-4262-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/20/2018] [Indexed: 01/25/2023] Open
Abstract
Background Innate lymphoid cells (ILC) are part of a heterogeneous family of haematopoietic effector cells which lack re-arranged antigen-specific receptors. They promote host defense and contribute to tissue and metabolic homeostasis, wound healing and immune surveillance. Their role in human cancer immunity is less defined, and therefore we aimed to identify the frequency and phenotype of distinct ILC groups in various types of cancer. Methods Tissue samples and peripheral blood were collected from patients undergoing surgical resection of gastrointestinal and breast tumours. Single cell suspension of tumour tissue was immediately obtained following surgery using tumour dissociation. Results We observed significantly higher frequencies of ILC2 (p value: 0.04) in malignant breast cancer tissue and significantly higher frequencies of group 1 ILC (p value: 0.001) in malignant gastrointestinal tumours. Tumour infiltrating ILC were found to show an activated phenotype with higher expression of MHC-II, KLRG1, early activation marker CD69 and CD44. Conclusions Activated innate lymphoid cells infiltrate tumours dependent on tumour type and location. Electronic supplementary material The online version of this article (10.1186/s12885-018-4262-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maryam Salimi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ruozheng Wang
- Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China
| | - Xuan Yao
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Xi Li
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Xiyan Wang
- Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China
| | - Yuhui Hu
- Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China
| | - Xumei Chang
- Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China
| | - Peiwen Fan
- Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China
| | - Tao Dong
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.,Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, China.,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China.,Chinese Academy of Medical Sciences-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Graham Ogg
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK. .,Key Laboratory of Cancer Immunity and Radiotherapy of Chinese Academy of Medical Sciences, Ürümqi, China. .,Chinese Academy of Medical Sciences-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
207
|
Abstract
Allergic inflammation is a type 2 immune disorder classically characterized by high levels of immunoglobulin E (IgE) and the development of Th2 cells. Asthma is a pulmonary allergic inflammatory disease resulting in bronchial hyper-reactivity. Atopic asthma is defined by IgE antibody-mediated mast cell degranulation, while in non-atopic asthma there is no allergen-specific IgE and more involvement of innate immune cells, such as basophils, group 2 innate lymphoid cells (ILC2), and eosinophils. Recently, protease allergens were shown to cause asthmatic responses in the absence of Th2 cells, suggesting that an innate cell network (IL-33/TSLP-basophil-ILC2-IL-5/IL-13 axis) can facilitate the sensitization phase of type 2 inflammatory responses. Recent evidence also indicates that in the chronic phase, these innate immune cells directly or indirectly contribute to the adaptive Th2 cell responses. In this review, we discuss the role of Th2 cytokines (IL-4 and IL-13) and innate immune cells (mast cells, basophils, ILC2s, and dendritic cells) in the cross-talk between innate and adaptive inflammatory responses.
Collapse
Affiliation(s)
- Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Japan.,Laboratory for Cytokine Regulation, Research Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| |
Collapse
|
208
|
Dong M, Ma C, Wang WQ, Chen J, Wei Y. Regulation of the IL-33/ST2 pathway contributes to the anti-inflammatory effect of acupuncture in the ovalbumin-induced murine asthma model. Acupunct Med 2018; 36:319-326. [PMID: 29581139 DOI: 10.1136/acupmed-2017-011377] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Bronchial asthma is a chronic airway inflammatory disease which has three main pathological features: airway hyperresponsiveness (AHR), airway remodelling, and chronic inflammation. Acupuncture is known to be an effective integrative medical therapy that has been used in the treatment of several chronic diseases, including bronchial asthma. The aim of the current study was to evaluate the effects of acupuncture on inflammation and regulation of the IL-33/ST2 pathway in a mouse model of asthma. METHODS The murine asthma model was established by both injection and inhalation of ovalbumin (OVA). Within 24 hours of the last OVA challenge, lung function was assessed by measurement of the airway resistance (RL) and lung dynamic compliance (Cdyn). Pulmonary tissues were collected for the detection of pathological changes and mucus secretion. Serum levels of tumour necrosis factor α (TNF-α), interleukin (IL)-1β, IL-33 and sST2 (secreted ST2) were detected by ELISA. Th17 cell proportions and counts in bronchoalveolar lavage fluid (BALF) were analysed by flow cytometry. RESULTS The results showed that AHR, chronic inflammation and mucus secretion were significantly suppressed by acupuncture treatment. RL decreased while Cdyn increased after acupuncture treatment. There was an apparent decrease in the serum concentrations of certain pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-33, and an increase in sST2 level compared with untreated asthmatic mice. Acupuncture also reduced the CD4 +IL-17A+ cell proportion and counts in BALF. CONCLUSION Acupuncture effectively protects lung function and attenuates airway inflammation in the OVA-induced mouse model of asthma, which supports the role of acupuncture as a potential therapy in asthma treatment.
Collapse
Affiliation(s)
- Ming Dong
- Gumei Community Health Center of Minhang District of Shanghai, Shanghai, China
| | - Cheng Ma
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| | - Wen-Qian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| | - Juan Chen
- Department of Pediatric Neurological Rehabilitation, Maternal and Child Health Hospital of Dengfeng, Henan, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| |
Collapse
|
209
|
Abstract
The growth and maturity of the peripheral immune system and subsequent development of pulmonary immunity in early life is dictated by host, environmental and microbial factors. Dysregulation during the critical window of immune development in the postnatal years results in disease which impacts on lifelong lung health. Asthma is a common disease in childhood and is often preceded by wheezing illnesses during the preschool years. However, the mechanisms underlying development of wheeze and how and why only some children progress to asthma is unknown. Human studies to date have generally focused on peripheral immune development, with little assessment of local tissue pathology in young children. Moreover, mechanisms underlying the interactions between inflammation and tissue repair at mucosal surfaces in early life remain unknown. Disappointingly, mechanistic studies in mice have predominantly used adult models. This review will consider the aspects of the neonatal immune system which might contribute to the development of early life wheezing disorders and asthma, and discuss the external environmental factors which may influence this process.
Collapse
Affiliation(s)
- Clare M Lloyd
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, Royal Brompton Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
210
|
Drake LY, Kita H. IL-33: biological properties, functions, and roles in airway disease. Immunol Rev 2018; 278:173-184. [PMID: 28658560 DOI: 10.1111/imr.12552] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Interleukin (IL)-33 is a key cytokine involved in type 2 immunity and allergic airway diseases. Abundantly expressed in lung epithelial cells, IL-33 plays critical roles in both innate and adaptive immune responses in mucosal organs. In innate immunity, IL-33 and group 2 innate lymphoid cells (ILC2s) provide an essential axis for rapid immune responses and tissue homeostasis. In adaptive immunity, IL-33 interacts with dendritic cells, Th2 cells, follicular T cells, and regulatory T cells, where IL-33 influences the development of chronic airway inflammation and tissue remodeling. The clinical findings that both the IL-33 and ILC2 levels are elevated in patients with allergic airway diseases suggest that IL-33 plays an important role in the pathogenesis of these diseases. IL-33 and ILC2 may also serve as biomarkers for disease classification and to monitor the progression of diseases. In this article, we reviewed the current knowledge of the biology of IL-33 and discussed the roles of the IL-33 in regulating airway immune responses and allergic airway diseases.
Collapse
Affiliation(s)
- Li Yin Drake
- Division of Allergic Diseases and Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hirohito Kita
- Division of Allergic Diseases and Department of Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
211
|
Zhou Y, Wang W, Zhao C, Wang Y, Wu H, Sun X, Guan Y, Zhang Y. Prostaglandin E 2 Inhibits Group 2 Innate Lymphoid Cell Activation and Allergic Airway Inflammation Through E-Prostanoid 4-Cyclic Adenosine Monophosphate Signaling. Front Immunol 2018; 9:501. [PMID: 29593738 PMCID: PMC5857904 DOI: 10.3389/fimmu.2018.00501] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022] Open
Abstract
Evidence is accumulating that group 2 innate lymphoid cells (ILC2) play an important role in allergic airway inflammation by producing a large amount of type 2 cytokines. But it remains poorly understood how its activities are properly controlled in vivo. Here, we demonstrated that prostaglandin E2 (PGE2) had a profound inhibitory effect on IL-33-induced ILC2 expansion and IL-5 and IL-13 production in vitro. This effect was mimicked by PGE1-alcohol but attenuated by ONO-AE3-208, indicating a selective action through the E-prostanoid 4 (EP4) receptor. In the IL-33-induced asthma model, coadministration of PGE2 or PGE1-alcohol resulted in diminished IL-5 and IL-13 production, reduced eosinophilia and alleviated lung pathology. In contrast, EP4-deficient mice displayed an exacerbated inflammatory response in another ILC2-mediated asthma model induced by Alternaria extract. Mechanistic studies demonstrated that the PGE2-mediated inhibition of ILC2 was dependent on cyclic adenosine monophosphate (cAMP) production. Further downstream, PGE2-EP4-cAMP signaling led to suppression of GATA3 and ST2 expression, which is known to be critical for ILC2 activation. These findings reveal a novel function of PGE2 as a negative regulator of ILC2 activation and highlight an endogenous counter-regulatory mechanism for the control of innate allergic inflammatory responses.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China
| | - Conghui Zhao
- Department of Oral Pathology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China
| | - Haoming Wu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China
| | - Xiuyuan Sun
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yu Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Medical Immunology, Ministry of Health (Peking University), Beijing, China.,Institute of Biological Sciences, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
212
|
Abstract
Eosinophils are the prominent cells in asthma, allergic bronchopulmonary mycosis (ABPMs), and fungal-sensitization-associated asthma, but their roles in the immunopathology of these disorders are not well understood. Moreover, the immunological mechanisms underlying the molecular direct effector interactions between fungi and eosinophils are rare and not fully known. Here, we provide an overview of eosinophil contributions to allergic asthma and ABPMs. We also revise the major general mechanisms of fungal recognition by eosinophils and consider past and recent advances in our understanding of the molecular mechanisms associated with eosinophil innate effector responses to different fungal species relevant to ABPMs (Alternaria alternata, Candida albicans, and Aspergillus fumigatus). We further examine and speculate about the therapeutic relevance of these findings in fungus-associated allergic pulmonary diseases.
Collapse
Affiliation(s)
- Rodrigo T Figueiredo
- Institute of Biomedical Sciences/Unit of Xerem, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Josiane S Neves
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
213
|
Thio CLP, Chi PY, Lai ACY, Chang YJ. Regulation of type 2 innate lymphoid cell-dependent airway hyperreactivity by butyrate. J Allergy Clin Immunol 2018. [PMID: 29522844 DOI: 10.1016/j.jaci.2018.02.032] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Allergic asthma is characterized by airway hyperreactivity (AHR) and inflammation driven by aberrant TH2 responses. Type 2 innate lymphoid cells (ILC2s) are a critical source of the TH2 cytokines IL-5 and IL-13, which promote acute asthma exacerbation. Short-chain fatty acids (SCFAs) have been shown to attenuate T cell-mediated allergic airway inflammation. However, their role in regulation of ILC2-driven AHR and lung inflammation remains unknown. OBJECTIVE We investigated the immunomodulatory role of SCFAs in regulation of ILC2-induced AHR and airway inflammation and delineated the mechanism involved. METHODS We assessed the role of SCFAs in regulating survival, proliferation, and cytokine production in lung sorted ILC2s. The SCFA butyrate was administered through drinking water or intranasally in BALB/c mice to evaluate its role in the ILC2-driven inflammatory response in IL-33 and Alternaria alternata models of allergic inflammation. We further confirmed our findings in human ILC2s. RESULTS We show that butyrate, but not acetate or propionate, inhibited IL-13 and IL-5 production by murine ILC2s. Systemic and local administration of butyrate significantly ameliorated ILC2-driven AHR and airway inflammation. We further demonstrate that butyrate inhibited ILC2 proliferation and GATA3 expression but did not induce cell apoptosis, likely through histone deacetylase (HDAC) inhibition, because trichostatin A, a pan-HDAC inhibitor, exerted similar effects on ILC2s. Importantly, cotreatment with trichostatin A and butyrate did not result in an additive effect. Finally, we show that butyrate reduces cytokine production in human ILC2s. CONCLUSION Our findings identify butyrate as a critical regulator of ILC2 proliferation and function through its HDAC inhibitory activity and can serve as a potential therapeutic target for asthma.
Collapse
Affiliation(s)
- Christina Li-Ping Thio
- Taiwan International Graduate Program (TIGP) in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Yu Chi
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Ya-Jen Chang
- Taiwan International Graduate Program (TIGP) in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
214
|
Barbour M, Wood R, Hridi SU, Wilson C, McKay G, Bushell TJ, Jiang HR. The therapeutic effect of anti-CD52 treatment in murine experimental autoimmune encephalomyelitis is associated with altered IL-33 and ST2 expression levels. J Neuroimmunol 2018. [PMID: 29526407 DOI: 10.1016/j.jneuroim.2018.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) mice were administered with murine anti-CD52 antibody to investigate its therapeutic effect and whether the treatment modulates IL-33 and ST2 expression. EAE severity and central nervous system (CNS) inflammation were reduced following the treatment, which was accompanied by peripheral T and B lymphocyte depletion and reduced production of various cytokines including IL-33, while sST2 was increased. In spinal cords of EAE mice, while the number of IL-33+ cells remained unchanged, the extracellular level of IL-33 protein was significantly reduced in anti-CD52 antibody treated mice compared with controls. Furthermore the number of ST2+ cells in the spinal cord of treated EAE mice was downregulated due to decreased inflammation and immune cell infiltration in the CNS. These results suggest that treatment with anti-CD52 antibody differentially alters expression of IL-33 and ST2, both systemically and within the CNS, which may indicate IL-33/ST2 axis is involved in the action of the antibody in inhibiting EAE.
Collapse
Affiliation(s)
- Mark Barbour
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Rachel Wood
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Shehla U Hridi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Chelsey Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Grant McKay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Trevor J Bushell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Hui-Rong Jiang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
215
|
Hinks TSC, Batty P, Klenerman P, Pavord ID, Xue L. Cytometric Gating Stringency Impacts Studies of Type 2 Innate Lymphoid Cells in Asthma. Am J Respir Cell Mol Biol 2018; 57:745-747. [PMID: 29192833 DOI: 10.1165/rcmb.2017-0201le] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Timothy S C Hinks
- 1 University of Oxford Oxford, United Kingdom and.,2 University of Southampton Southampton, United Kingdom
| | - Paul Batty
- 1 University of Oxford Oxford, United Kingdom and
| | | | - Ian D Pavord
- 1 University of Oxford Oxford, United Kingdom and
| | - Luzheng Xue
- 1 University of Oxford Oxford, United Kingdom and
| |
Collapse
|
216
|
Type 2 Innate Lymphocytes Actuate Immunity Against Tumours and Limit Cancer Metastasis. Sci Rep 2018; 8:2924. [PMID: 29440650 PMCID: PMC5811448 DOI: 10.1038/s41598-018-20608-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Type 2 innate lymphoid cells (ILC2) potentiate immune responses, however, their role in mediating adaptive immunity in cancer has not been assessed. Here, we report that mice genetically lacking ILC2s have significantly increased tumour growth rates and conspicuously higher frequency of circulating tumour cells (CTCs) and resulting metastasis to distal organs. Our data support the model that IL-33 dependent tumour-infiltrating ILC2s are mobilized from the lungs and other tissues through chemoattraction to enter tumours, and subsequently mediate tumour immune-surveillance by cooperating with dendritic cells to promote adaptive cytolytic T cell responses. We conclude that ILC2s play a fundamental, yet hitherto undescribed role in enhancing anti-cancer immunity and controlling tumour metastasis.
Collapse
|
217
|
von Moltke J, Pepper M. Sentinels of the Type 2 Immune Response. Trends Immunol 2018; 39:99-111. [PMID: 29122456 PMCID: PMC6181126 DOI: 10.1016/j.it.2017.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/25/2022]
Abstract
Type 2 immune responses have evolved to sense and respond to large, non-replicating infections or non-microbial noxious compounds in tissues. The development of these responses therefore depends upon highly coordinated and tightly regulated tissue-residing cellular sensors and responders. Multiple exposure to type 2 helper T cell (Th2)-inducing stimuli further enhances both the diversity and potency of the response. This review discusses advances in our understanding of the interacting cellular subsets that comprise both primary and secondary type 2 responses. Current knowledge regarding type 2 immune responses in the lung are initially presented and are then contrasted with what is known about the small intestine. The studies described portray an immune response that depends upon well-organized tissue structures, and suggest their modulation as a therapeutic strategy.
Collapse
Affiliation(s)
- Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
218
|
Teufelberger AR, Nordengrün M, Braun H, Maes T, De Grove K, Holtappels G, O'Brien C, Provoost S, Hammad H, Gonçalves A, Beyaert R, Declercq W, Vandenabeele P, Krysko DV, Bröker BM, Bachert C, Krysko O. The IL-33/ST2 axis is crucial in type 2 airway responses induced by Staphylococcus aureus –derived serine protease–like protein D. J Allergy Clin Immunol 2018; 141:549-559.e7. [DOI: 10.1016/j.jaci.2017.05.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/09/2023]
|
219
|
Li BWS, Beerens DMJM, Brem MD, Hendriks RW. Characterization of Group 2 Innate Lymphoid Cells in Allergic Airway Inflammation Models in the Mouse. Methods Mol Biol 2018; 1559:169-183. [PMID: 28063044 DOI: 10.1007/978-1-4939-6786-5_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Allergic asthma is a chronic inflammatory lung disease mediated by type 2 cytokines produced by T helper 2 (Th2) cells as well as the recently discovered group 2 innate lymphoid cells (ILC2). Due to a lack of unique markers, the accurate phenotypic characterization and quantification of ILC2 requires a comprehensive panel of fluorescently labeled antibodies. The markers that are currently used to characterize ILC2 have not been standardized and often vary between research groups, which poses significant challenges when comparing data. Intranasal administration of the pro-inflammatory cytokine IL-33 in mice is associated with strong, Th2 cell-independent ILC2 activation. ILC2 are also activated in mouse models of allergic asthma based on the physiologically relevant house dust mite (HDM) allergen, which parallel eosinophilic airway inflammation observed in asthma patients. Here, we describe the analysis of ILC2 by flow cytometry in these two commonly used allergic airway inflammation models in the mouse.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Dior M J M Beerens
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Maarten D Brem
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands.
| |
Collapse
|
220
|
Mohammadi H, Sharafkandi N, Hemmatzadeh M, Azizi G, Karimi M, Jadidi-Niaragh F, Baradaran B, Babaloo Z. The role of innate lymphoid cells in health and disease. J Cell Physiol 2018; 233:4512-4529. [PMID: 29058773 DOI: 10.1002/jcp.26250] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/13/2017] [Indexed: 12/13/2022]
Abstract
Innate lymphoid cells (ILCs) are kind of innate immune cells which can be divided into three main subsets according to their cytokine release profile, transcription factors, and surface markers. ILCs affect the initial stages of immunity in response to microbes and participate in immunity, inflammation, and tissue repair. ILCs modulate immunity through resistance to the pathogens and regulation of autoimmune inflammation and metabolic homeostasis. Therefore dysregulation of ILCs may lead to chronic pathologies such as allergies (i.e., asthma), inflammation (i.e., inflammatory bowel disease), and autoimmunity (i.e., psoriasis, atopic dermatitis, rheumatoid arthritis, multiple sclerosis, and ankylosing spondylitis). Regarding the critical role of ILCs in the regulation of immune system, the elucidation of their function in different conditions makes an interesting target for improvement of novel therapeutic approach to modulate an immune response in different disease context.
Collapse
Affiliation(s)
- Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadia Sharafkandi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Karimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
221
|
Toki S, Goleniewska K, Reiss S, Zhang J, Bloodworth MH, Stier MT, Zhou W, Newcomb DC, Ware LB, Stanwood GD, Galli A, Boyd KL, Niswender KD, Peebles RS. Glucagon-like peptide 1 signaling inhibits allergen-induced lung IL-33 release and reduces group 2 innate lymphoid cell cytokine production in vivo. J Allergy Clin Immunol 2018; 142:1515-1528.e8. [PMID: 29331643 DOI: 10.1016/j.jaci.2017.11.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 10/19/2017] [Accepted: 11/01/2017] [Indexed: 01/26/2023]
Abstract
BACKGROUND IL-33 is one of the most consistently associated gene candidates for asthma identified by using a genome-wide association study. Studies in mice and in human cells have confirmed the importance of IL-33 in inducing type 2 cytokine production from both group 2 innate lymphoid cells (ILC2s) and TH2 cells. However, there are no pharmacologic agents known to inhibit IL-33 release from airway cells. OBJECTIVE We sought to determine the effect of glucagon-like peptide 1 receptor (GLP-1R) signaling on aeroallergen-induced airway IL-33 production and release and on innate type 2 airway inflammation. METHODS BALB/c mice were challenged intranasally with Alternaria extract for 4 consecutive days. GLP-1R agonist or vehicle was administered starting either 2 days before the first Alternaria extract challenge or 1 day after the first Alternaria extract challenge. RESULTS GLP-1R agonist treatment starting 2 days before the first Alternaria extract challenge decreased IL-33 release in the bronchoalveolar lavage fluid and dual oxidase 1 (Duox1) mRNA expression 1 hour after the first Alternaria extract challenge and IL-33 expression in lung epithelial cells 24 hours after the last Alternaria extract challenge. Furthermore, GLP-1R agonist significantly decreased the number of ILC2s expressing IL-5 and IL-13, lung protein expression of type 2 cytokines and chemokines, the number of perivascular eosinophils, mucus production, and airway responsiveness compared with vehicle treatment. GLP-1R agonist treatment starting 1 day after the first Alternaria extract challenge also significantly decreased eosinophilia and type 2 cytokine and chemokine expression in the airway after 4 days of Alternaria extract challenge. CONCLUSION These results reveal that GLP-1R signaling might be a therapy to reduce IL-33 release and inhibit the ILC2 response to protease-containing aeroallergens, such as Alternaria.
Collapse
Affiliation(s)
- Shinji Toki
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Sara Reiss
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Jian Zhang
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Melissa H Bloodworth
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Matthew T Stier
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Weisong Zhou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Dawn C Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Gregg D Stanwood
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University, Tallahassee, Fla
| | - Aurelio Galli
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tenn; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kelli L Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kevin D Niswender
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tenn; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tenn; Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, Tenn; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tenn.
| | - R Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tenn; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tenn.
| |
Collapse
|
222
|
Zhang Y, Feng Y, Li L, Ye X, Wang J, Wang Q, Li P, Li N, Zheng X, Gao X, Li C, Li F, Sun B, Lai K, Su Z, Zhong N, Chen L, Feng L. Immunization with an adenovirus-vectored TB vaccine containing Ag85A-Mtb32 effectively alleviates allergic asthma. J Mol Med (Berl) 2018; 96:249-263. [PMID: 29302700 PMCID: PMC5859035 DOI: 10.1007/s00109-017-1614-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/12/2017] [Accepted: 12/13/2017] [Indexed: 12/31/2022]
Abstract
Abstract Current treatments for allergic asthma primarily ameliorate symptoms rather than inhibit disease progression. Regulating the excessive T helper type 2 (Th2) responses may prevent asthma exacerbation. In this study, we investigated the protective effects of Ad5-gsgAM, an adenovirus vector carrying two mycobacterial antigens Ag85A and Mtb32, against allergic asthma. Using an ovalbumin (OVA)-induced asthmatic mouse model, we found that Ad5-gsgAM elicited much more Th1-biased CD4+T and CD8+T cells than bacillus Calmette-Guérin (BCG). After OVA challenge, Ad5-gsgAM-immunized mice showed significantly lowered airway inflammation in comparison with mice immunized with or without BCG. Total serum immunoglobulin E and pulmonary inducible-nitric-oxide-synthase were efficiently reduced. The cytokine profiles in bronchial-alveolar-lavage-fluids (BALFs) were also modulated, as evidenced by the increased level of interferon-γ (IFN-γ) and the decreased level of interleukin (IL)-4, IL-5, and IL-13. Anti-inflammatory cytokine IL-10 was sharply increased, whereas pro-inflammatory cytokine IL-33 was significantly decreased. Importantly, exogenous IL-33 abrogated the protective effects of Ad5-gsgAM, revealing that the suppression of IL-33/ST2 axis substantially contributed to protection against allergic inflammation. Moreover, regulatory T cells were essential for regulating aberrant Th2 responses as well as IL-33/ST2 axis. These results suggested that modulating the IL-33/ST2 axis via adenovirus-vectored mycobacterial antigen vaccination may provide clinical benefits in allergic inflammatory airways disease. Key messages •Ad5-gsgAM elicits Th1 responses and suppresses Th2-mediated allergic asthma in mice. •Ad5-gsgAM inhibits IL-33/ST2 axis by reducing IL-33 secretion but not ILC2 recruiting. •Treg is essential for modulating Th2 responses and IL-33/ST2 axis by Ad5-gsgAM. Electronic supplementary material The online version of this article (10.1007/s00109-017-1614-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiling Zhang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China.,Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Ying Feng
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Liang Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Xianmiao Ye
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Jinlin Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Qian Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Na Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuehua Zheng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Xiang Gao
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Chufang Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feng Li
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Baoqing Sun
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhong Su
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China. .,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China.
| | - Liqiang Feng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China.
| |
Collapse
|
223
|
Jeong JS, Kim SR, Lee YC. Can Controlling Endoplasmic Reticulum Dysfunction Treat Allergic Inflammation in Severe Asthma With Fungal Sensitization? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:106-120. [PMID: 29411551 PMCID: PMC5809759 DOI: 10.4168/aair.2018.10.2.106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/05/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022]
Abstract
Severe asthma is a heterogeneous disease entity to which diverse cellular components and pathogenetic mechanisms contribute. Current asthma therapies, including new biologic agents, are mainly targeting T helper type 2 cell-dominant inflammation, so that they are often unsatisfactory in the treatment of severe asthma. Respiratory fungal exposure has long been regarded as a precipitating factor for severe asthma phenotype. Moreover, as seen in clinical definitions of allergic bronchopulmonary aspergillosis (ABPA) and severe asthma with fungal sensitization (SAFS), fungal allergy-associated severe asthma phenotype is increasingly thought to have distinct pathobiologic mechanisms requiring different therapeutic approaches other than conventional treatment. However, there are still many unanswered questions on the direct causality of fungal sensitization in inducing severe allergic inflammation in SAFS. Recently, growing evidence suggests that stress response from the largest organelle, endoplasmic reticulum (ER), is closely interconnected to diverse cellular immune/inflammatory platforms, thereby being implicated in severe allergic lung inflammation. Interestingly, a recent study on this issue has suggested that ER stress responses and several associated molecular platforms, including phosphoinositide 3-kinase-δ and mitochondria, may be crucial players in the development of severe allergic inflammation in the SAFS. Defining emerging roles of ER and associated cellular platforms in SAFS may offer promising therapeutic options in the near future.
Collapse
Affiliation(s)
- Jae Seok Jeong
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea
| | - So Ri Kim
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea.,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Medical School, Jeonju, Korea
| | - Yong Chul Lee
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea.,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
224
|
Mitthamsiri W, Pradubpongsa P, Sangasapaviliya A, Boonpiyathad T. Decreased CRTH2 Expression and Response to Allergen Re-stimulation on Innate Lymphoid Cells in Patients With Allergen-Specific Immunotherapy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:662-674. [PMID: 30306748 PMCID: PMC6182198 DOI: 10.4168/aair.2018.10.6.662] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/02/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Purpose Group 2 innate lymphoid cells (ILC2s) have been implicated in the pathogenesis of allergic disease. However, the effect of allergen-specific immunotherapy (AIT) on ILCs remains to be clarified. The aim of this study was to evaluate the levels of ILC subsets in allergic rhinitis (AR) patients in response to house dust mite (HDM)-specific immunotherapy. Methods We enrolled 37 AR patients undergoing AIT (16 responders and 11 non-responders) for 2 years, 35 HDM AR patients and 28 healthy subjects. Peripheral blood mononuclear cells (PBMCs) were analyzed by flow cytometry to identify ILC subsets. Stimulation of ILC2s with recombinant allergen-specific protein was used to determine ILC2's activation (CD69 expression). Results Responder AIT patients and healthy subjects had a decreased frequency of circulating ILC2s compared to non-responder AIT and AR patients. Conversely, ILC1s from responder AIT patients and healthy subjects showed increased frequency compared to non-responder AIT and AR patients. The frequency of ILC3s natural cytotoxicity receptor (NCR)+ and NCR− in responder AIT patients was significantly lower compared to AR patients and healthy subjects. The ILC1: ILC2 proportion in responder AIT patients was similar to that of healthy subjects. PBMCs from patients who were responders to AIT had a significantly lower expression of the activation marker CD69 on ILC2s in response to allergen re-stimulation compared to AR patients, but no difference compared to non-responder AIT patients and healthy subjects. Conclusions We propose that AIT might affect ILC responses. The activation of ILC2s was reduced in AR patients treated with AIT. Our results indicate that a relative ILC1/ILC2 skewed response is a possible key to successful AIT.
Collapse
Affiliation(s)
- Wat Mitthamsiri
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Panitan Pradubpongsa
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Atik Sangasapaviliya
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Tadech Boonpiyathad
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand.
| |
Collapse
|
225
|
Geslewitz WE, Percopo CM, Rosenberg HF. Eosinophil persistence in vivo and sustained viability ex vivo in response to respiratory challenge with fungal allergens. Clin Exp Allergy 2018; 48:29-38. [PMID: 29068539 PMCID: PMC5746458 DOI: 10.1111/cea.13050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Eosinophils are immunomodulatory leucocytes that contribute to the pathogenesis of Th2-driven asthma and allergic lung diseases. OBJECTIVE Our goal was to identify unique properties of eosinophils recruited to the lungs and airways of mice in response to challenge with asthma-associated fungal allergens. METHODS Mice were challenged intranasally on days 0, 3 and 6 with a filtrate of Alternaria alternata. Recruited eosinophils were enumerated in bronchoalveolar lavage fluid. Eosinophils were also isolated from lungs of mice sensitized and challenged with Aspergillus fumigatus and evaluated ex vivo in tissue culture. RESULTS Eosinophils persist in the airways for several weeks in response to brief provocation with A. alternata in wild-type, Gm-csf- and eotaxin-1-gene-deleted mice, while eosinophils are recruited but do not persist in the absence of IL-13. Eosinophils isolated from the lungs A. alternata-challenged mice are cytokine-enriched compared to those from IL5tg mice, including 800-fold higher levels of eotaxin-1. Furthermore, eosinophils from the lungs and spleen of fungal allergen-challenged wild-type mice are capable of prolonged survival ex vivo, in contrast to eosinophils from both untreated and fungal allergen-challenged IL5tg mice, which undergo rapid demise in the absence of exogenous cytokine support. TNF-α (but not IL5, IL-3, eotaxin-1 or GM-CSF) was detected in supernatants of ex vivo eosinophil cultures from the lungs of fungal allergen-challenged wild-type mice. However, neither TNF-α gene deletion nor anti-TNF-α neutralizing antibodies had any impact sustained eosinophil survival ex vivo. CONCLUSION AND CLINICAL RELEVANCE Eosinophils are phenotypically and functionally heterogeneous. As shown here, eosinophils from fungal allergen-challenged wild-type mice maintain a distinct cytokine profile, and, unlike eosinophils isolated from IL5tg mice, they survive ex vivo in the absence of exogenous pro-survival cytokine support. As treatments for asthma currently in development focus on limiting eosinophil viability via strategic cytokine blockade, the molecular mechanisms underlying differential survival merit further investigation.
Collapse
Affiliation(s)
- Wendy E. Geslewitz
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland
| | - Caroline M. Percopo
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland
| | - Helene F. Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
226
|
IFN-γ increases susceptibility to influenza A infection through suppression of group II innate lymphoid cells. Mucosal Immunol 2018; 11:209-219. [PMID: 28513592 PMCID: PMC5693789 DOI: 10.1038/mi.2017.41] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/02/2017] [Indexed: 02/04/2023]
Abstract
Increased levels of interferon-γ (IFN-γ) are routinely observed in the respiratory tract following influenza virus infection, yet its potential role remains unclear. We now demonstrate that influenza-induced IFN-γ restricts protective innate lymphoid cell group II (ILC2) function in the lung following challenge with the pandemic H1N1 A/CA/04/2009 (CA04) influenza virus. Specifically, IFN-γ deficiency resulted in enhanced ILC2 activity, characterized by increased production of interleukin (IL)-5 and amphiregulin, and improved tissue integrity, yet no change in ILC2 numbers, viral load or clearance. We further found that IFN-γ-deficient mice, as well as wild-type animals treated with neutralizing anti-IFN-γ antibody, exhibited decreased susceptibility to lethal infection with H1N1 CA04 influenza virus, and moreover that survival was dependent on the presence of IL-5. The beneficial effects of IFN-γ neutralization were not observed in ILC2-deficient animals. These data support the novel concept that IFN-γ can have a detrimental role in the pathogenesis of influenza through a restriction in ILC2 activity. Thus, regulation of ILC2 activity is a potential target for post-infection therapy of influenza.
Collapse
|
227
|
Stier MT, Zhang J, Goleniewska K, Cephus JY, Rusznak M, Wu L, Van Kaer L, Zhou B, Newcomb DC, Peebles RS. IL-33 promotes the egress of group 2 innate lymphoid cells from the bone marrow. J Exp Med 2017; 215:263-281. [PMID: 29222107 PMCID: PMC5748848 DOI: 10.1084/jem.20170449] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/03/2017] [Accepted: 11/03/2017] [Indexed: 12/27/2022] Open
Abstract
ILC2s are potent mucosal effector cells that participate in type 2 inflammatory responses. Stier et al. demonstrate that IL-33 negatively regulates CXCR4, mediating the egress of ILC2 lineage cells from the bone marrow for potential hematogenous trafficking. Group 2 innate lymphoid cells (ILC2s) are effector cells within the mucosa and key participants in type 2 immune responses in the context of allergic inflammation and infection. ILC2s develop in the bone marrow from common lymphoid progenitor cells, but little is known about how ILC2s egress from the bone marrow for hematogenous trafficking. In this study, we identified a critical role for IL-33, a hallmark peripheral ILC2-activating cytokine, in promoting the egress of ILC2 lineage cells from the bone marrow. Mice lacking IL-33 signaling had normal development of ILC2s but retained significantly more ILC2 progenitors in the bone marrow via augmented expression of CXCR4. Intravenous injection of IL-33 or pulmonary fungal allergen challenge mobilized ILC2 progenitors to exit the bone marrow. Finally, IL-33 enhanced ILC2 trafficking to the lungs in a parabiosis mouse model of tissue disruption and repopulation. Collectively, these data demonstrate that IL-33 plays a critical role in promoting ILC2 egress from the bone marrow.
Collapse
Affiliation(s)
- Matthew T Stier
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jian Zhang
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jacqueline Y Cephus
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Mark Rusznak
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lan Wu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Baohua Zhou
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Dawn C Newcomb
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN .,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
228
|
Maric J, Ravindran A, Mazzurana L, Björklund ÅK, Van Acker A, Rao A, Friberg D, Dahlén SE, Heinemann A, Konya V, Mjösberg J. Prostaglandin E 2 suppresses human group 2 innate lymphoid cell function. J Allergy Clin Immunol 2017; 141:1761-1773.e6. [PMID: 29217133 PMCID: PMC5929462 DOI: 10.1016/j.jaci.2017.09.050] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 08/30/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022]
Abstract
Background Group 2 innate lymphoid cells (ILC2s) are involved in the initial phase of type 2 inflammation and can amplify allergic immune responses by orchestrating other type 2 immune cells. Prostaglandin (PG) E2 is a bioactive lipid that plays protective roles in the lung, particularly during allergic inflammation. Objective We set out to investigate how PGE2 regulates human ILC2 function. Methods The effects of PGE2 on human ILC2 proliferation and intracellular cytokine and transcription factor expression were assessed by means of flow cytometry. Cytokine production was measured by using ELISA, and real-time quantitative PCR was performed to detect PGE2 receptor expression. Results PGE2 inhibited GATA-3 expression, as well as production of the type 2 cytokines IL-5 and IL-13, from human tonsillar and blood ILC2s in response to stimulation with a combination of IL-25, IL-33, thymic stromal lymphopoietin, and IL-2. Furthermore, PGE2 downregulated the expression of IL-2 receptor α (CD25). In line with this observation, PGE2 decreased ILC2 proliferation. These effects were mediated by the combined action of E-type prostanoid receptor (EP) 2 and EP4 receptors, which were specifically expressed on ILC2s. Conclusion Our findings reveal that PGE2 limits ILC2 activation and propose that selective EP2 and EP4 receptor agonists might serve as a promising therapeutic approach in treating allergic diseases by suppressing ILC2 function.
Collapse
Affiliation(s)
- Jovana Maric
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria; Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Avinash Ravindran
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Luca Mazzurana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Åsa K Björklund
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Aline Van Acker
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anna Rao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Danielle Friberg
- Department of Oto-Rhino-Laryngology, Karolinska University Hospital and CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Experimental Asthma and Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Viktoria Konya
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria; Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
229
|
Gupta RK, Gupta K, Dwivedi PD. Pathophysiology of IL-33 and IL-17 in allergic disorders. Cytokine Growth Factor Rev 2017; 38:22-36. [PMID: 29153708 DOI: 10.1016/j.cytogfr.2017.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022]
Abstract
Allergic diseases are among common clinical conditions, affecting millions of children and adults throughout the world. Food allergies, skin allergies (atopic dermatitis), and respiratory allergies (allergic rhinitis and asthma) are the common types of allergies. Recently discovered cytokines IL-17 and IL-33 have been found to play an important role in the pathogenicity of various hypersensitive disorders. After exposure to allergens or infection with parasites or viruses, IL-17 and IL-33 producing cells, such as Th17 and specialized epithelial cells respectively, become activated and trigger the pathogenic immune responses in different susceptible conditions. Potent inhibitors of these cytokines have been identified recently that may represent potential therapeutic agents to overcome the clinical complications of allergies. In the present review, we have discussed the cellular sources, modes of action and regulation of IL-17 and IL-33 in the context of hypersensitive diseases. We have also assessed the therapeutic potential of inhibitory molecules that may alter production of both these cytokines, and thus modulate susceptible conditions.
Collapse
Affiliation(s)
- Rinkesh Kumar Gupta
- Food Toxicology Laboratory; Food, Drug and Chemical Toxicology Group; CSIR-Indian Institute of Toxicology Research (CSIR-IITR); Vishvigyan Bhawan; 31; Mahatma Gandhi Marg; Lucknow 226 001; Uttar Pradesh; India
| | - Kriti Gupta
- Food Toxicology Laboratory; Food, Drug and Chemical Toxicology Group; CSIR-Indian Institute of Toxicology Research (CSIR-IITR); Vishvigyan Bhawan; 31; Mahatma Gandhi Marg; Lucknow 226 001; Uttar Pradesh; India
| | - Premendra D Dwivedi
- Food Toxicology Laboratory; Food, Drug and Chemical Toxicology Group; CSIR-Indian Institute of Toxicology Research (CSIR-IITR); Vishvigyan Bhawan; 31; Mahatma Gandhi Marg; Lucknow 226 001; Uttar Pradesh; India.
| |
Collapse
|
230
|
Li BWS, Stadhouders R, de Bruijn MJW, Lukkes M, Beerens DMJM, Brem MD, KleinJan A, Bergen I, Vroman H, Kool M, van IJcken WFJ, Rao TN, Fehling HJ, Hendriks RW. Group 2 Innate Lymphoid Cells Exhibit a Dynamic Phenotype in Allergic Airway Inflammation. Front Immunol 2017; 8:1684. [PMID: 29250067 PMCID: PMC5716969 DOI: 10.3389/fimmu.2017.01684] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2) are implicated in allergic asthma as an early innate source of the type 2 cytokines IL-5 and IL-13. However, their induction in house dust mite (HDM)-mediated airway inflammation additionally requires T cell activation. It is currently unknown whether phenotypic differences exist between ILC2s that are activated in a T cell-dependent or T cell-independent fashion. Here, we compared ILC2s in IL-33- and HDM-driven airway inflammation. Using flow cytometry, we found that surface expression levels of various markers frequently used to identify ILC2s were dependent on their mode of activation, highly variable over time, and differed between tissue compartments, including bronchoalveolar lavage (BAL) fluid, lung, draining lymph nodes, and spleen. Whereas in vivo IL-33-activated BAL fluid ILC2s exhibited an almost uniform CD25+CD127+T1/ST2+ICOS+KLRG1+ phenotype, at a comparable time point after HDM exposure BAL fluid ILC2s had a very heterogeneous surface marker phenotype. A major fraction of HDM-activated ILC2s were CD25lowCD127+T1/ST2low ICOSlowKLRG1low, but nevertheless had the capacity to produce large amounts of type 2 cytokines. HDM-activated CD25low ILC2s in BAL fluid and lung rapidly reverted to CD25high ILC2s upon in vivo stimulation with IL-33. Genome-wide transcriptional profiling of BAL ILC2s revealed ~1,600 differentially expressed genes: HDM-stimulated ILC2s specifically expressed genes involved in the regulation of adaptive immunity through B and T cell interactions, whereas IL-33-stimulated ILC2s expressed high levels of proliferation-related and cytokine genes. In both airway inflammation models ILC2s were present in the lung submucosa close to epithelial cells, as identified by confocal microscopy. In chronic HDM-driven airway inflammation ILC2s were also found inside organized cellular infiltrates near T cells. Collectively, our findings show that ILC2s are phenotypically more heterogeneous than previously thought, whereby their surface marker and gene expression profile are highly dynamic.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | | | - Melanie Lukkes
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | - Maarten D Brem
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Ingrid Bergen
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | | | | | | |
Collapse
|
231
|
Bartemes K, Chen CC, Iijima K, Drake L, Kita H. IL-33-Responsive Group 2 Innate Lymphoid Cells Are Regulated by Female Sex Hormones in the Uterus. THE JOURNAL OF IMMUNOLOGY 2017; 200:229-236. [PMID: 29133293 DOI: 10.4049/jimmunol.1602085] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 10/14/2017] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in multiple organs in the body, where they play roles in immunity, tissue homeostasis, and metabolic regulation. However, little is known about the regulatory mechanisms of ILC2s in different organs. Here, we identified ILC2s in the mouse uterus and found that they express cell surface molecules, including the IL-33 receptor, ST2, that are roughly comparable to those expressed by lung ILC2s. Both in vivo and in vitro treatment with IL-33 induced type 2 cytokine production in uterine ILC2s, suggesting that they respond to IL-33 in a manner similar to ILC2s in other organs. Importantly, uterine ILC2s were nearly absent in ovariectomized mice and were increased in wild-type mice by estrogen administration, whereas lung ILC2s were unaffected by both ovariectomy and estrogen administration. Likewise, a marked reduction in uterine ILC2s was observed in mice deficient in estrogen receptor α or estrogen receptor β. Furthermore, uterine ILC2s highly expressed estrogen receptor α, and in vitro culture of isolated uterine ILC2s with 17β-estradiol modified expression of a number of genes. Finally, an increased prevalence in neonatal mortality was observed in litters from dams lacking the IL-33 receptor, ST2. Taken together, our findings indicate that unlike lung IL2Cs, uterine ILC2s are regulated by female sex hormones, which may specialize them for specific physiological functions.
Collapse
Affiliation(s)
- Kathleen Bartemes
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Chien-Chang Chen
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Koji Iijima
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Li Drake
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and .,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
232
|
Singh PB, Pua HH, Happ HC, Schneider C, von Moltke J, Locksley RM, Baumjohann D, Ansel KM. MicroRNA regulation of type 2 innate lymphoid cell homeostasis and function in allergic inflammation. J Exp Med 2017; 214:3627-3643. [PMID: 29122948 PMCID: PMC5716040 DOI: 10.1084/jem.20170545] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/05/2017] [Accepted: 09/21/2017] [Indexed: 12/30/2022] Open
Abstract
Singh et al. examined microRNA expression and physiological requirements in type 2 innate lymphoid cells (ILC2s). The miR-17∼92 cluster promotes ILC2 growth, cytokine expression, and function in allergic inflammation. MicroRNAs (miRNAs) exert powerful effects on immunity through coordinate regulation of multiple target genes in a wide variety of cells. Type 2 innate lymphoid cells (ILC2s) are tissue sentinel mediators of allergic inflammation. We established the physiological requirements for miRNAs in ILC2 homeostasis and immune function and compared the global miRNA repertoire of resting and activated ILC2s and T helper type 2 (TH2) cells. After exposure to the natural allergen papain, mice selectively lacking the miR-17∼92 cluster in ILC2s displayed reduced lung inflammation. Moreover, miR-17∼92–deficient ILC2s exhibited defective growth and cytokine expression in response to IL-33 and thymic stromal lymphopoietin in vitro. The miR-17∼92 cluster member miR-19a promoted IL-13 and IL-5 production and inhibited expression of several targets, including SOCS1 and A20, signaling inhibitors that limit IL-13 and IL-5 production. These findings establish miRNAs as important regulators of ILC2 biology, reveal overlapping but nonidentical miRNA-regulated gene expression networks in ILC2s and TH2 cells, and reinforce the therapeutic potential of targeting miR-19 to alleviate pathogenic allergic responses.
Collapse
Affiliation(s)
- Priti B Singh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Heather H Pua
- Department of Pathology, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Hannah C Happ
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Christoph Schneider
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Jakob von Moltke
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, CA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Dirk Baumjohann
- Institute for Immunology, Biomedical Center Munich, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - K Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA .,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
233
|
HpARI Protein Secreted by a Helminth Parasite Suppresses Interleukin-33. Immunity 2017; 47:739-751.e5. [PMID: 29045903 PMCID: PMC5655542 DOI: 10.1016/j.immuni.2017.09.015] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/09/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022]
Abstract
Infection by helminth parasites is associated with amelioration of allergic reactivity, but mechanistic insights into this association are lacking. Products secreted by the mouse parasite Heligmosomoides polygyrus suppress type 2 (allergic) immune responses through interference in the interleukin-33 (IL-33) pathway. Here, we identified H. polygyrus Alarmin Release Inhibitor (HpARI), an IL-33-suppressive 26-kDa protein, containing three predicted complement control protein (CCP) modules. In vivo, recombinant HpARI abrogated IL-33, group 2 innate lymphoid cell (ILC2) and eosinophilic responses to Alternaria allergen administration, and diminished eosinophilic responses to Nippostrongylus brasiliensis, increasing parasite burden. HpARI bound directly to both mouse and human IL-33 (in the cytokine’s activated state) and also to nuclear DNA via its N-terminal CCP module pair (CCP1/2), tethering active IL-33 within necrotic cells, preventing its release, and forestalling initiation of type 2 allergic responses. Thus, HpARI employs a novel molecular strategy to suppress type 2 immunity in both infection and allergy. HpARI is a suppressor of IL-33 release and consequent allergic sensitization HpARI binds active IL-33 and nuclear DNA, tethering IL-33 within necrotic cells HpARI is active against both human and murine IL-33
Collapse
|
234
|
Everaere L, Ait Yahia S, Bouté M, Audousset C, Chenivesse C, Tsicopoulos A. Innate lymphoid cells at the interface between obesity and asthma. Immunology 2017; 153:21-30. [PMID: 28880992 DOI: 10.1111/imm.12832] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity and asthma prevalence has dramatically and concomitantly increased over the last 25 years, and many epidemiological studies have highlighted obesity as an important risk factor for asthma. Although many studies have been performed, the underlying mechanisms remain poorly understood. Innate mechanisms have been involved in both diseases, in particular through the recently described innate lymphoid cells (ILCs). ILCs are subdivided into three groups that are defined by their cytokine production and by their master transcription factor expression, in sharp correlation with their T helper counterparts. However, unlike T helper cells, ILCs do not express antigen-specific receptors, but respond to damage-induced signals. ILCs have been found in target tissues of both diseases, and data have implicated these cells in the pathogenesis of both diseases. In particular group 2 ILCs (ILC2) are activated in both the adipose and lung tissues under the effect of interleukin-33 and interleukin-25 expression. However, counter-intuitively to the well-known association between obesity and asthma, ILC2 are beneficial for obesity but deleterious for asthma. This review will examine the roles of ILCs in each disease and recent data highlighting ILCs as a putative link between obesity and asthma.
Collapse
Affiliation(s)
- Laetitia Everaere
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France
| | - Saliha Ait Yahia
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France
| | - Mélodie Bouté
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France
| | - Camille Audousset
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France.,Clinique des Maladies Respiratoires et Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Cécile Chenivesse
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France.,Clinique des Maladies Respiratoires et Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Anne Tsicopoulos
- Institut National de la Santé et de la Recherche Médicale, Lille, France.,CNRS, UMR 8204, Centre for Infection and Immunity of Lille, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université de Lille, Lille, France.,Clinique des Maladies Respiratoires et Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| |
Collapse
|
235
|
Chen R, Smith SG, Salter B, El-Gammal A, Oliveria JP, Obminski C, Watson R, O'Byrne PM, Gauvreau GM, Sehmi R. Allergen-induced Increases in Sputum Levels of Group 2 Innate Lymphoid Cells in Subjects with Asthma. Am J Respir Crit Care Med 2017; 196:700-712. [PMID: 28422515 DOI: 10.1164/rccm.201612-2427oc] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RATIONALE Group 2 innate lymphoid cells (ILC2), a major source of type 2 cytokines, initiate eosinophilic inflammatory responses in murine models of asthma. OBJECTIVES To investigate the role of ILC2 in allergen-induced airway eosinophilic responses in subjects with atopy and asthma. METHODS Using a diluent-controlled allergen challenge crossover study, where all subjects (n = 10) developed allergen-induced early and late responses, airway eosinophilia, and increased methacholine airway responsiveness, bone marrow, blood, and sputum samples were collected before and after inhalation challenge. MEASUREMENTS AND MAIN RESULTS ILC2 (lin-FcεRI-CD45+CD127+ST2+) and CD4+T lymphocytes were enumerated by flow cytometry, as well as intracellular IL-5 and IL-13 expression. Steroid sensitivity of ILC2 and CD4+ T cells was investigated in vitro. A significant increase in total, IL-5+, IL-13+, and CRTH2+ ILC2 was found in sputum, 24 hours after allergen, coincident with a significant decrease in blood ILC2. Total, IL-5+, and IL-13+, but not CRTH2+, CD4+ T cells significantly increased at 24 and 48 hours after allergen in sputum. In blood and bone marrow, only CD4+ cells demonstrated increased activation after allergen. Airway eosinophilia correlated with IL-5+ ILC2 at all time points and allergen-induced changes in IL-5+ CD4+ cells at 48 hours after allergen. Dexamethasone significantly attenuated IL-2- and IL-33-stimulated IL-5 and IL-13 production by both cell types. CONCLUSIONS Innate and adaptive immune cells are increased in the airways associated with allergic asthmatic responses. Total and type 2 cytokine-positive ILC2 are increased only within the airways, whereas CD4+ T lymphocytes demonstrated local and systemic increases. Steroid sensitivity of both cells may explain effectiveness of this therapy in those with mild asthma.
Collapse
Affiliation(s)
- Ruchong Chen
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and.,2 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Steven G Smith
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Brittany Salter
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Amani El-Gammal
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - John Paul Oliveria
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Caitlin Obminski
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Rick Watson
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Paul M O'Byrne
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Gail M Gauvreau
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| | - Roma Sehmi
- 1 Asthma Research Group, Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada; and
| |
Collapse
|
236
|
Zhong H, Fan XL, Yu QN, Qin ZL, Chen D, Xu R, Chen DH, Lin ZB, Wen W, Fu QL. Increased innate type 2 immune response in house dust mite-allergic patients with allergic rhinitis. Clin Immunol 2017; 183:293-299. [PMID: 28917723 DOI: 10.1016/j.clim.2017.09.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 08/11/2017] [Accepted: 09/12/2017] [Indexed: 01/02/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are essential in initiating and driving allergic immune responses. However, there were inconsistent findings of the ILC2 levels in allergic rhinitis (AR) patients. This study investigated the ILC2 levels in the peripheral blood of house dust mite (HDM)-sensitized AR patients and their ability to secrete type 2 cytokines. The levels of ILC2s with phenotypic ILC2 characteristics were increased in the HDM-AR patients. The AR patients' symptom score and IL-13 levels were positively associated with the ILC2s in HDM-AR patients. The epithelial cytokine stimulation induced dramatic production of IL-5 and IL-13 in PBMCs of AR patients. We successfully sorted ILC2s from AR patients and identified their ability of type 2 cytokines production. The number of ILC2s increased in the HDM-AR patients and ILC2s produced the amount of TH2 cytokines in the presence of epithelial cytokines, which suggested the important role of ILC2 in AR patients.
Collapse
Affiliation(s)
- Hua Zhong
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China
| | - Xing-Liang Fan
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China
| | - Qiu-Ning Yu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China
| | - Zi-Li Qin
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China
| | - Dong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Xu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - De-Hua Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhi-Bin Lin
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiping Wen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China.
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
237
|
Vincent M, Percier P, De Prins S, Huygen K, Potemberg G, Muraille E, Romano M, Michel O, Denis O. Investigation of inflammatory and allergic responses to common mold species: Results from in vitro experiments, from a mouse model of asthma, and from a group of asthmatic patients. INDOOR AIR 2017; 27:933-945. [PMID: 28370571 DOI: 10.1111/ina.12385] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/25/2017] [Indexed: 06/07/2023]
Abstract
Most studies on molds focus on Alternaria alternata and Aspergillus fumigatus. Here, we report on inflammatory and allergenic properties of more typical indoor species Aspergillus versicolor, P. chrysogenum, C. cladosporioïdes, and C. sphaerospermum that were compared to A. alternata and A. fumigatus. In a mouse model, after intranasal instillation, A. alternaria, A. versicolor, and C. sphaerospermum induced the early recruitment of neutrophils and the strong expression of inflammatory markers in the bronchoalveolar lavages fluids. A. fumigatus also induced the early accumulation of neutrophils but with lower levels of inflammatory markers. Chronic treatment induced variable response according to species: P. chrysogenum and A. fumigatus appeared strong pro-allergenic inducers compared to A. alternata and C. sphaerospermum while A. versicolor and C. cladosporioides induced a mixed pro-allergenic/pro-inflammatory response. In mold-sensitized asthmatics, mold-specific Immunoglobulin E (IgE) were detected with an in-house dot-blot assay. A. fumigatus and A. alternata were the most frequent sensitizers. Altogether, P. chrysogenum, P. brevicompactum, C. sphaerospermum, and C. cladosporïoides were the "major sensitizer" (defined as the strongest response against a single mold species) for almost 30% of the asthmatics. These results show that, not only A. alternata and A. fumigatus, but also indoor species have strong inflammatory and allergic properties and a harmful potency.
Collapse
Affiliation(s)
- M Vincent
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| | - P Percier
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| | - S De Prins
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| | - K Huygen
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| | - G Potemberg
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - E Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
| | - M Romano
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| | - O Michel
- Clinic of Immunology and Allergology, CHU Brugmann, Université Libre de Bruxelles, Bruxelles, Belgium
| | - O Denis
- Scientific Service Immunology, WIV-ISP, Bruxelles, Belgium
| |
Collapse
|
238
|
Karami J, Mahmoudi M, Amirzargar A, Gharshasbi M, Jamshidi A, Aslani S, Nicknam MH. Promoter hypermethylation of BCL11B gene correlates with downregulation of gene transcription in ankylosing spondylitis patients. Genes Immun 2017; 18:170-175. [DOI: 10.1038/gene.2017.17] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 01/05/2023]
|
239
|
Cavagnero K, Doherty TA. Cytokine and Lipid Mediator Regulation of Group 2 Innate Lymphoid Cells (ILC2s) in Human Allergic Airway Disease. ACTA ACUST UNITED AC 2017; 2. [PMID: 28959799 PMCID: PMC5614509 DOI: 10.4172/2576-3881.1000116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The recent discovery of group 2 innate lymphoid cells (ILC2s) has caused a paradigm shift in the understanding of allergic airway disease pathogenesis. Prior to the discovery of ILC2s, Th2 cells were largely thought to be the primary source of type 2 cytokines; however, activated ILC2s have since been shown to contribute significantly, and in some cases, dominantly to type 2 cytokine production. Since the discovery of ILC2s in 2010, many mediators have been shown to regulate their effector functions. Initial studies identified the epithelial derived cytokines IL-25, IL-33, and TSLP as activators of ILC2s, and recent studies have identified many additional cytokine and lipid mediators that are involved in ILC2 regulation. ILC2s and their mediators represent novel therapeutic targets for allergic airway diseases and intensive investigation is underway to better understand ILC2 biology and upstream and downstream pathways that lead to ILC2-driven airway pathology. In this review, we will focus on the cytokine and lipid mediators that regulate ILC2s in human allergic airway disease, as well as highlight newly discovered mediators of mouse ILC2s that may eventually translate to humans.
Collapse
|
240
|
Aron JL, Akbari O. Regulatory T cells and type 2 innate lymphoid cell-dependent asthma. Allergy 2017; 72:1148-1155. [PMID: 28160290 DOI: 10.1111/all.13139] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2017] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a recently identified group of cells with the potent capability to produce Th2-type cytokines such as interleukin (IL)-5 and IL-13. Several studies suggest that ILC2s play an important role in the development of allergic diseases and asthma. Activation of pulmonary ILC2s in murine models lacking T and B cells induces eosinophilia and airway hyper-reactivity (AHR), which are cardinal features of asthma. More importantly, numerous recent studies have highlighted the role of ILC2s in asthma persistence and exacerbation among human subjects, and thus, regulation of pulmonary ILC2s is a major area of investigation aimed at curbing allergic lung inflammation and exacerbation. Emerging evidence reveals that a group of regulatory T cells, induced Tregs (iTregs), effectively suppress the production of ILC2-driven, pro-inflammatory cytokines IL-5 and IL-13. The inhibitory effects of iTregs are blocked by preventing direct cellular contact or by inhibiting the ICOS-ICOS-ligand (ICOSL) pathway, suggesting that both direct contact and ICOS-ICOSL interaction are important in the regulation of ILC2 function. Also, cytokines such as IL-10 and TGF-β1 significantly reduce cytokine secretion by ILC2s. Altogether, these new findings uncover iTregs as potent regulators of ILC2 activation and implicate their utility as a therapeutic approach for the treatment of ILC2-mediated allergic asthma and respiratory disease.
Collapse
Affiliation(s)
- J. L. Aron
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| | - O. Akbari
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles CA USA
| |
Collapse
|
241
|
Oczypok EA, Perkins TN, Oury TD. Alveolar Epithelial Cell-Derived Mediators: Potential Direct Regulators of Large Airway and Vascular Responses. Am J Respir Cell Mol Biol 2017; 56:694-699. [PMID: 28080134 DOI: 10.1165/rcmb.2016-0151ps] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchial epithelial cells and pulmonary endothelial cells are thought to be the primary modulators of conducting airways and vessels, respectively. However, histological examination of both mouse and human lung tissue reveals that alveolar epithelial cells (AECs) line the adventitia of large airways and vessels and thus are also in a position to directly regulate these structures. The primary purpose of this perspective is to highlight the fact that AECs coat the adventitial surface of every vessel and airway in the lung parenchyma. This localization is ideal for transmitting signals that can contribute to physiologic and pathologic responses in vessels and airways. A few examples of mediators produced by AECs that may contribute to vascular and airway responses are provided to illustrate some of the potential effects that AECs may modulate.
Collapse
Affiliation(s)
- Elizabeth A Oczypok
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
242
|
Morita H, Moro K, Koyasu S. Innate lymphoid cells in allergic and nonallergic inflammation. J Allergy Clin Immunol 2017; 138:1253-1264. [PMID: 27817797 DOI: 10.1016/j.jaci.2016.09.011] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/26/2022]
Abstract
In the last decade, the full picture of the role of innate lymphoid cells (ILCs) has been gradually revealed. ILCs are classified into 3 groups based on their transcription factors and cytokine production patterns, which mirror helper T-cell subsets. Unlike T cells and B cells, ILCs do not have antigen receptors. They promptly respond to multiple tissue-derived factors, such as cytokines and alarmins, and produce multiple proinflammatory and immunoregulatory cytokines. It has been reported that ILC-derived cytokines are important for the induction and regulation of inflammation. Accumulating evidence suggests that ILCs play substantial roles in protection against infection and the pathogenesis of inflammatory diseases, such as allergic diseases and autoimmune diseases. Different ILC subsets localize in distinct tissue/organ niches and receive tissue-derived signals on different types of inflammation, which allows them to acquire diverse phenotypes with specialized effector capacities. In this review we highlight the roles of ILCs in a variety of organs, such as the airway, skin, and gastrointestinal tract, in the context of allergic and nonallergic inflammation.
Collapse
Affiliation(s)
- Hideaki Morita
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
243
|
Abstract
PURPOSE OF REVIEW Many phenotypes of asthma exist, ranging from mild asthma with onset during childhood to severe asthma with later onset, making asthma a broad disease with different pathologies. A gender disparity exists in asthma prevalence. As adults, women have an increased asthma prevalence compared to men. Further, women are more likely to have severe asthma and a later onset of asthma compared to men. Here, we review clinical and animal studies that have defined the role of sex hormones in airway inflammation, smooth muscle contraction, mucus production, and airway mechanics associated with asthma pathogenesis. RECENT FINDINGS Clinical evidence shows that increased asthma symptoms occur in females starting at puberty compared to those in boys. However, after puberty, the role for sex hormones in regulating asthma symptoms during menstruation, pregnancy, and menopause is not as clear. Animal studies have shown that estrogen increases and testosterone decreases Th2-mediated airway inflammation, and that females have increased IL-17A-mediated airway inflammation compared to males. Further, females had increased DC and Mϕ function compared to males. However, the mechanisms driving the types of allergic inflammation are not fully elucidated. Overall, ovarian hormones increased and testosterone decreased airway inflammation in asthma, but the mechanisms remain unclear. Delineating these pathways using animal models as well as women and men with various phenotypes of asthma will help determine if women with asthma should take (or avoid) hormonal contraceptives as well as predict changes in asthma symptoms during life phases, including pregnancy and menopause, when sex hormones are dramatically changing.
Collapse
|
244
|
Abstract
Innate lymphoid cells are functionally diverse subsets of immune cells including the conventional natural killer cells, lymphoid tissue inducers, type 1, 2, and 3 with significant roles in immunity and pathogenesis of inflammatory diseases. Type 2 innate lymphoid cells (ILC2s) resemble type 2 helper (Th2) cells in cytokine production and contribute to anti-helminth immunity, maintaining mucosal tissue integrity, and adipose tissue browning. ILC2s play important roles in the pathogenesis of allergic diseases and asthma. Studying the pathways of activation and regulation of ILC2s are currently a priority for giving a better understanding of pathogenesis of diseases with immunological roots. Recently, our laboratory and others have shown several pathways of regulation of ILC2s by co-stimulatory molecules such as ICOS, regulatory T cells and by compounds such as nicotine. In this review, we summarize the current understanding of the mechanisms of activation and regulation of ILC2s and the role of these cells in health and disease.
Collapse
Affiliation(s)
- Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California 90033, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California 90033, USA
| |
Collapse
|
245
|
Lewis BW, Sultana R, Sharma R, Noël A, Langohr I, Patial S, Penn AL, Saini Y. Early Postnatal Secondhand Smoke Exposure Disrupts Bacterial Clearance and Abolishes Immune Responses in Muco-Obstructive Lung Disease. THE JOURNAL OF IMMUNOLOGY 2017; 199:1170-1183. [PMID: 28667160 DOI: 10.4049/jimmunol.1700144] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/05/2017] [Indexed: 01/15/2023]
Abstract
Secondhand smoke (SHS) exposure has been linked to the worsening of ongoing lung diseases. However, whether SHS exposure affects the manifestation and natural history of imminent pediatric muco-obstructive airway diseases such as cystic fibrosis remains unclear. To address these questions, we exposed Scnn1b transgenic (Scnn1b-Tg+) mice to SHS from postnatal day (PND) 3-21 and lung phenotypes were examined at PND22. Although a majority of filtered air (FA)-exposed Scnn1b-Tg+ (FA-Tg+) mice successfully cleared spontaneous bacterial infections by PND22, the SHS-exposed Scnn1b-Tg+ (SHS-Tg+) mice failed to resolve these infections. This defect was associated with suppressed antibacterial defenses, i.e., phagocyte recruitment, IgA secretion, and Muc5b expression. Whereas the FA-Tg+ mice exhibited marked mucus obstruction and Th2 responses, SHS-Tg+ mice displayed a dramatic suppression of these responses. Mechanistically, downregulated expression of IL-33, a stimulator of type II innate lymphoid cells, in lung epithelial cells was associated with suppression of neutrophil recruitment, IgA secretions, Th2 responses, and delayed bacterial clearance in SHS-Tg+ mice. Cessation of SHS exposure for 21 d restored previously suppressed responses, including phagocyte recruitment, IgA secretion, and mucous cell metaplasia. However, in contrast with FA-Tg+ mice, the SHS-Tg+ mice had pronounced epithelial necrosis, alveolar space consolidation, and lymphoid hyperplasia; indicating lagged unfavorable effects of early postnatal SHS exposure in later life. Collectively, our data show that early postnatal SHS exposure reversibly suppresses IL-33 levels in airspaces which, in turn, results in reduced neutrophil recruitment and diminished Th2 response. Our data indicate that household smoking may predispose neonates with muco-obstructive lung disease to bacterial exacerbations.
Collapse
Affiliation(s)
- Brandon W Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Razia Sultana
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Rahul Sharma
- National Hansen's Disease Program, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803; and
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Ingeborg Langohr
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803.,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803;
| |
Collapse
|
246
|
Lund SJ, Portillo A, Cavagnero K, Baum RE, Naji LH, Badrani JH, Mehta A, Croft M, Broide DH, Doherty TA. Leukotriene C4 Potentiates IL-33-Induced Group 2 Innate Lymphoid Cell Activation and Lung Inflammation. THE JOURNAL OF IMMUNOLOGY 2017; 199:1096-1104. [PMID: 28667163 DOI: 10.4049/jimmunol.1601569] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 06/06/2017] [Indexed: 01/06/2023]
Abstract
Asthma is a complex disease that is promoted by dysregulated immunity and the presence of many cytokine and lipid mediators. Despite this, there is a paucity of data demonstrating the combined effects of multiple mediators in asthma pathogenesis. Group 2 innate lymphoid cells (ILC2s) have recently been shown to play important roles in the initiation of allergic inflammation; however, it is unclear whether lipid mediators, such as cysteinyl leukotrienes (CysLTs), which are present in asthma, could further amplify the effects of IL-33 on ILC2 activation and lung inflammation. In this article, we show that airway challenges with the parent CysLT, leukotriene C4 (LTC4), given in combination with low-dose IL-33 to naive wild-type mice, led to synergistic increases in airway Th2 cytokines, eosinophilia, and peribronchial inflammation compared with IL-33 alone. Further, the numbers of proliferating and cytokine-producing lung ILC2s were increased after challenge with both LTC4 and IL-33. Levels of CysLT1R, CysLT2R, and candidate leukotriene E4 receptor P2Y12 mRNAs were increased in ILC2s. The synergistic effect of LTC4 with IL-33 was completely dependent upon CysLT1R, because CysLT1R-/- mice, but not CysLT2R-/- mice, had abrogated responses. Further, CysLTs directly potentiated IL-5 and IL-13 production from purified ILC2s stimulated with IL-33 and resulted in NFAT1 nuclear translocation. Finally, CysLT1R-/- mice had reduced lung eosinophils and ILC2 responses after exposure to the fungal allergen Alternaria alternata Thus, CysLT1R promotes LTC4- and Alternaria-induced ILC2 activation and lung inflammation. These findings suggest that multiple pathways likely exist in asthma to activate ILC2s and propagate inflammatory responses.
Collapse
Affiliation(s)
- Sean J Lund
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Alex Portillo
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Kellen Cavagnero
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Rachel E Baum
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Luay H Naji
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Jana H Badrani
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Amit Mehta
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - David H Broide
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Taylor A Doherty
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| |
Collapse
|
247
|
Morikawa T, Fukuoka A, Matsushita K, Yasuda K, Iwasaki N, Akasaki S, Fujieda S, Yoshimoto T. Activation of group 2 innate lymphoid cells exacerbates and confers corticosteroid resistance to mouse nasal type 2 inflammation. Int Immunol 2017; 29:221-233. [DOI: 10.1093/intimm/dxx030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/21/2017] [Indexed: 12/13/2022] Open
|
248
|
Becerra-Díaz M, Wills-Karp M, Heller NM. New perspectives on the regulation of type II inflammation in asthma. F1000Res 2017; 6:1014. [PMID: 28721208 PMCID: PMC5497827 DOI: 10.12688/f1000research.11198.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2017] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the lungs which has been thought to arise as a result of inappropriately directed T helper type-2 (Th2) immune responses of the lungs to otherwise innocuous inhaled antigens. Current asthma therapeutics are directed towards the amelioration of downstream consequences of type-2 immune responses (i.e. β-agonists) or broad-spectrum immunosuppression (i.e. corticosteroids). However, few approaches to date have been focused on the primary prevention of immune deviation. Advances in molecular phenotyping reveal heterogeneity within the asthmatic population with multiple endotypes whose varying expression depends on the interplay between numerous environmental factors and the inheritance of a broad range of susceptibility genes. The most common endotype is one described as "type-2-high" (i.e. high levels of interleukin [IL]-13, eosinophilia, and periostin). The identification of multiple endotypes has provided a potential explanation for the observations that therapies directed at typical Th2 cytokines (IL-4, IL-5, and IL-13) and their receptors have often fallen short when they were tested in a diverse group of asthmatic patients without first stratifying based on disease endotype or severity. However, despite the incorporation of endotype-dependent stratification schemes into clinical trial designs, variation in drug responses are still apparent, suggesting that additional genetic/environmental factors may be contributing to the diversity in drug efficacy. Herein, we will review recent advances in our understanding of the complex pathways involved in the initiation and regulation of type-2-mediated immune responses and their modulation by host factors (genetics, metabolic status, and the microbiome). Particular consideration will be given to how this knowledge could pave the way for further refinement of disease endotypes and/or the development of novel therapeutic strategies for the treatment of asthma .
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Nicola M. Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
249
|
Maazi H, Banie H, Aleman Muench GR, Patel N, Wang B, Sankaranarayanan I, Bhargava V, Sato T, Lewis G, Cesaroni M, Karras J, Das A, Soroosh P, Akbari O. Activated plasmacytoid dendritic cells regulate type 2 innate lymphoid cell-mediated airway hyperreactivity. J Allergy Clin Immunol 2017; 141:893-905.e6. [PMID: 28579374 DOI: 10.1016/j.jaci.2017.04.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Allergic asthma is a prevalent inflammatory disease of the airways caused by dysregulated immune balance in the lungs with incompletely understood pathogenesis. The recently identified type 2 innate lymphoid cells (ILC2s) play significant roles in the pathogenesis of asthma. Although ILC2-activating factors have been identified, the mechanisms that suppress ILC2s remain largely unknown. Plasmacytoid dendritic cells (pDCs) are important in antiviral immunity and in maintaining tolerance to inert antigens. OBJECTIVE We sought to address the role of pDCs in regulating ILC2 function and ILC2-mediated airway hyperreactivity (AHR) and lung inflammation. METHODS We used several murine models, including BDCA-2-diphtheria toxin receptor (DTR) transgenic and IFN-α receptor 1-deficient mice, as well as purified primary ILC2s, to reach our objective. We extended and validated our findings to human ILC2s. RESULTS We show that activation of pDCs through Toll-like receptor 7/8 suppresses ILC2-mediated AHR and airway inflammation and that depletion of pDCs reverses this suppression. We further show that pDCs suppress cytokine production and the proliferation rate while increasing the apoptosis rate of ILC2s through IFN-α production. Transcriptomic analysis of both human and murine ILC2s confirms the activation of regulatory pathways in ILC2s by IFN-α. CONCLUSION Activation of pDCs alleviates AHR and airway inflammation by suppressing ILC2 function and survival. Our findings reveal a novel regulatory pathway in ILC2-mediated pulmonary inflammation with important clinical implications.
Collapse
Affiliation(s)
- Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | | | | | - Nisheel Patel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Bowen Wang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Ishwarya Sankaranarayanan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Vipul Bhargava
- Janssen Research and Development, Spring House, Philadelphia, Pa
| | - Takahiro Sato
- Janssen Research and Development, Spring House, Philadelphia, Pa
| | - Gavin Lewis
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif; Janssen Research and Development, San Diego, Calif
| | - Matteo Cesaroni
- Janssen Research and Development, Spring House, Philadelphia, Pa
| | - James Karras
- Janssen Research and Development, San Diego, Calif
| | - Anuk Das
- Janssen Research and Development, Spring House, Philadelphia, Pa
| | | | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
| |
Collapse
|
250
|
Filbey K, Bouchery T, Le Gros G. The role of ILC2 in hookworm infection. Parasite Immunol 2017; 40. [PMID: 28369954 DOI: 10.1111/pim.12429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/22/2017] [Indexed: 01/01/2023]
Abstract
Hookworm is a major public health concern, yet still relatively little is known about the immunological responses involved in human infection. Animal studies are mainly confined to using the natural rodent helminth Nippostrongylus brasiliensis as this has been proposed as the most accurate model of hookworm infection in the mouse, with both its life cycle and the immune responses it invokes having been extremely well characterized. In this review, we examine the roles that type 2 innate lymphoid cells (ILC2s) play in immunity and host tolerance to hookworm infection, particularly N. brasiliensis. This includes their role in the initiation and regulation of immune responses, as well as in the resolution and limitation of tissue damage required after an infection with a large organism, such as a helminth.
Collapse
Affiliation(s)
- K Filbey
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - T Bouchery
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - G Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|