1
|
Xu S, Wang D, Tan L, Lu J. The role of NLRP3 inflammasome in type 2 inflammation related diseases. Autoimmunity 2024; 57:2310269. [PMID: 38332696 DOI: 10.1080/08916934.2024.2310269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
Type 2 inflammation related diseases, such as atopic dermatitis, asthma, and allergic rhinitis, are diverse and affect multiple systems in the human body. It is common for individuals to have multiple co-existing type 2 inflammation related diseases, which can impose a significant financial and living burden on patients. However, the exact pathogenesis of these diseases is still unclear. The NLRP3 inflammasome is a protein complex composed of the NLRP3 protein, ASC, and Caspase-1, and is activated through various mechanisms, including the NF-κB pathway, ion channels, and lysosomal damage. The NLRP3 inflammasome plays a role in the immune response to pathogens and cellular damage. Recent studies have indicated a strong correlation between the abnormal activation of NLRP3 inflammasome and the onset of type 2 inflammation. Additionally, it has been demonstrated that suppressing NLRP3 expression effectively diminishes the inflammatory response, highlighting its promising therapeutic applications. Therefore, this article reviews the role of NLRP3 inflammasome in the development and therapy of multiple type 2 inflammation related diseases.
Collapse
Affiliation(s)
- Shenming Xu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Jianyun Lu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
2
|
Chen T, Shi S, Li X, Zhou L, Yu Y, Cai Y, Wang J, Kan H, Xu Y, Huang C, Tan Y, Meng X, Zhao Z. Improved ambient air quality is associated with decreased prevalence of childhood asthma and infancy shortly after weaning is a sensitive exposure window. Allergy 2024; 79:1166-1179. [PMID: 37458141 DOI: 10.1111/all.15815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND The urban ambient air quality has been largely improved in the past decade. It is unknown whether childhood asthma prevalence is still increasing in ever top-ranking city of Shanghai, whether the improved air quality is beneficial for children's asthma and what time window of exposure plays critical roles. METHODS Using a repeat cross-sectional design, we analyzed the association between early life exposure to particles and wheezing/asthma in each individual and combined surveys in 2011 and 2019, respectively, in 11,825 preschool children in Shanghai. RESULTS A significantly lower prevalence of doctor-diagnosed asthma (DDA) (6.6% vs. 10.5%, p < 0.001) and wheezing (10.5% vs. 23.2%, p < 0.001) was observed in 2019 compared to 2011. Exposure to fine particulate matter (PM2.5), coarse particles (PM2.5-10) and inhalable particles (PM10) was decreased in 2019 by 6.3%, 35.4%, and 44.7% in uterus and 24.3%, 20.2%, and 31.8% in infancy, respectively. Multilevel log-binomial regression analysis showed exposure in infancy had independent association with wheezing/DDA adjusting for exposure in uterus. For each interquartile range (IQR) increase of infancy PM2.5, PM2.5-10 and PM10 exposure, the odds ratios were 1.39 (95% confidence interval (CI): 1.24-1.56), 1.51 (95% CI:1.15-1.98) and 1.53 (95% CI:1.27-1.85) for DDA, respectively. The distributed lag non-linear model showed the sensitive exposure window (SEW) was 5.5-11 months after birth. Stratified analysis showed the SEWs were at or shortly after weaning, but only in those with <6 months of exclusive breastfeeding. CONCLUSIONS Improved ambient PM benefits in decreasing childhood asthma prevalence. We firstly reported the finding of SEW to PM at or closely after weaning on childhood asthma.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Su Shi
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Xinyue Li
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Lu Zhou
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Biostatistics, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yunfei Cai
- Department of General Management and Statistics, Shanghai Environment Monitoring Center, Shanghai, China
| | - Jing Wang
- Pudong New Area Center for Disease Control and Prevention, Shanghai, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health WMO/IGAC MAP-AQ Asian Office Shanghai, Fudan University, Shanghai, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health WMO/IGAC MAP-AQ Asian Office Shanghai, Fudan University, Shanghai, China
| | - Chen Huang
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai, China
| | - Yongqiang Tan
- Department of Pediatrics, Chongming Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xia Meng
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health WMO/IGAC MAP-AQ Asian Office Shanghai, Fudan University, Shanghai, China
| | - Zhuohui Zhao
- Department of Environmental Health, School of Public Health, the Key Laboratory of Public Health Safety of the Ministry of Education, and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health WMO/IGAC MAP-AQ Asian Office Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Chen N, Xie QM, Song SM, Guo SN, Fang Y, Fei GH, Wu HM. Dexamethasone protects against asthma via regulating Hif-1α-glycolysis-lactate axis and protein lactylation. Int Immunopharmacol 2024; 131:111791. [PMID: 38460304 DOI: 10.1016/j.intimp.2024.111791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Abstract
PURPOSE Asthma can not be eradicated till now and its control primarily relies on the application of corticosteroids. Recently, glycolytic reprogramming has been reportedly contributed to asthma, this study aimed to reveal whether the effect of corticosteroids on asthma control is related to their regulation of glycolysis and glycolysis-dependent protein lactylation. METHODS Ovalbumin (OVA) aeroallergen was used to challenge mice and stimulate human macrophage cell line THP-1 following dexamethasone (DEX) treatment. Airway hyperresponsiveness, airway inflammation, the expressions of key glycolytic enzymes and pyroptosis markers, the level of lactic acid, real-time glycolysis and oxidative phosphorylation (OXPHOS), and protein lactylation were analyzed. RESULTS DEX significantly attenuated OVA-induced eosinophilic airway inflammation, including airway hyperresponsiveness, leukocyte infiltration, goblet cell hyperplasia, Th2 cytokines production and pyroptosis markers expression. Meanwhile, OVA-induced Hif-1α-glycolysis axis was substantially downregulated by DEX, which resulted in low level of lactic acid. Besides, key glycolytic enzymes in the lungs of asthmatic mice were notably co-localized with F4/80-positive macrophages, indicating metabolic shift to glycolysis in lung macrophages during asthma. This was confirmed in OVA-stimulated THP-1 cells that DEX treatment resulted in reductions in pyroptosis, glycolysis and lactic acid level. Finally, protein lactylation was found significantly increased in the lungs of asthmatic mice and OVA-stimulated THP-1 cells, which were both inhibited by DEX. CONCLUSION Our present study revealed that the effect of DEX on asthma control was associated with its suppressing of Hif-1α-glycolysis-lactateaxis and subsequent protein lactylation, which may open new avenues for the therapy of eosinophilic asthma.
Collapse
Affiliation(s)
- Ning Chen
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Si-Ming Song
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Si-Nuo Guo
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Yu Fang
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Guang-He Fei
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China.
| |
Collapse
|
4
|
Zhang L, Wu Q, Huang Y, Zheng J, Guo S, He L. Formononetin ameliorates airway inflammation by suppressing ESR1/NLRP3/Caspase-1 signaling in asthma. Biomed Pharmacother 2023; 168:115799. [PMID: 37922653 DOI: 10.1016/j.biopha.2023.115799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Since inhaled glucocorticoids are the first-line treatment for asthma, asthma management becomes extremely difficult when asthma does not react well to glucocorticoids. Formononetin, a bioactive isoflavone and typical phytoestrogen, has been shown to have an anti-inflammatory impact while alleviating epithelial barrier dysfunction, which plays a role in the pathogenesis of allergic illnesses like asthma. However, the biological mechanisms behind this impact are unknown. As a result, we set out to investigate the effects of formononetin on airway inflammation and epithelial barrier repair in house dust mite (HDM)-induced asthmatic mice. We further expanded on formononetin's putative mode of action in reducing airway inflammation by modifying epithelial barrier dysfunction. In the current study, researchers discovered that formononetin significantly lowered total IgE levels in serum and interleukin (IL)-4, IL-6, and IL-17A levels in bronchoalveolar lavage fluid (BALF) in HDM-challenged asthmatic mice. Experiments on cell proliferation, migration, and apoptosis were performed in vitro to determine the effect of formononetin on bronchial epithelial barrier repair. Furthermore, in lipopolysaccharide (LPS)-stimulated 16HBE cells, formononetin increased cell proliferation and migration while preventing apoptosis and lowering the Bax/Bcl-2 ratio. In vitro and in vivo, formononetin significantly inhibited toll-like receptor 4 (TLR4) and estrogen receptor (ESR1)/Nod-like receptor family pyrin domain-containing protein 3 (NLRP3)/Caspase-1 signaling. These findings show that formononetin can reduce airway inflammation in HDM-challenged asthmatic mice by promoting epithelial barrier repair and possibly by inhibiting ESR1/NLRP3/Caspase-1 signaling as the underlying mechanism; formononetin could be a promising alternative treatment for asthma.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Wu
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zheng
- Department of Respiratory Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Guo
- Department of Endocrine, Genetics and Metabolism, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Li He
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
5
|
Hu M, Zhao X, Liu Y, Zhou H, You Y, Xue Z. Complex interplay of gut microbiota between obesity and asthma in children. Front Microbiol 2023; 14:1264356. [PMID: 38029078 PMCID: PMC10655108 DOI: 10.3389/fmicb.2023.1264356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is an important risk factor and common comorbidity of childhood asthma. Simultaneously, obesity-related asthma, a distinct asthma phenotype, has attracted significant attention owing to its association with more severe clinical manifestations, poorer disease control, and reduced quality of life. The establishment of the gut microbiota during early life is essential for maintaining metabolic balance and fostering the development of the immune system in children. Microbial dysbiosis influences host lipid metabolism, triggers chronic low-grade inflammation, and affects immune responses. It is intimately linked to the susceptibility to childhood obesity and asthma and plays a potentially crucial transitional role in the progression of obesity-related asthma. This review article summarizes the latest research on the interplay between asthma and obesity, with a particular focus on the mediating role of gut microbiota in the pathogenesis of obesity-related asthma. This study aims to provide valuable insight to enhance our understanding of this condition and offer preliminary evidence to support the development of therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Yannan You
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Xue
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Li Z, Wang X, Zhang W, Yang W, Xu B, Hu W. Excretory/Secretory Products from Schistosoma japonicum Eggs Alleviate Ovalbumin-Induced Allergic Airway Inflammation. PLoS Negl Trop Dis 2023; 17:e0011625. [PMID: 37788409 PMCID: PMC10547495 DOI: 10.1371/journal.pntd.0011625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Excretory/secretory products (ESPs) derived from helminths have been reported to effectively control allergic inflammation, which have better therapeutic prospects than live parasite infections. However, it remains unknown whether ESPs from schistosome eggs can protect against allergies, despite reports alleging that schistosome infection could alleviate disordered allergic inflammation. METHOD In the present study, we investigated the protective effects of ESPs from Schistosoma japonicum eggs (ESP-SJE) on asthmatic inflammation. Firstly, we successfully established an allergic airway inflammation model in mice by alum-adjuvanted ovalbumin (OVA) sensitization and challenge. ESP-SJE were administered intraperitoneally on days -1 and 13 (before sensitization), on day 20 (before challenge), and on days 21-24 (challenge phase). RESULTS The results showed that ESP-SJE treatment significantly reduced the infiltration of inflammatory cells, especially eosinophils into the lung tissue, inhibited the production of the total and OVA-specific IgE during OVA-sensitized and -challenged phases, respectively, and suppressed the secretion of Th2-type inflammatory cytokines (IL-4). Additionally, ESP-SJE treatment significantly upregulated the regulatory T cells (Tregs) in the lung tissue during OVA challenge. Furthermore, using liquid chromatography-mass spectrometry analysis and Treg induction experiments in vitro, we might identify nine potential therapeutic proteins against allergic inflammation in ESP-SJE. The targets of these candidate proteins included glutathione S-transferase, egg protein CP422 precursor, tubulin alpha-2/alpha-4 chain, actin-2, T-complex protein 1 subunit beta, histone H₄, whey acidic protein core region, and molecular chaperone HtpG. CONCLUSION Taken together, the results discussed herein demonstrated that ESP-SJE could significantly alleviate OVA-induced asthmatic inflammation in a murine model, which might be mediated by the upregulation of Treg in lung tissues that may be induced by the potential modulatory proteins. Therefore, potential proteins in ESP-SJE might be the best candidates to be tested for therapeutic application of asthma, thus pointing out to a possible new therapy for allergic airway inflammation.
Collapse
Affiliation(s)
- Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, P. R. China
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Xiaoling Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wenbin Yang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Bin Xu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Wei Hu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
7
|
Liu L, Zhou L, Wang LL, Zheng PD, Zhang FQ, Mao ZY, Zhang HJ, Liu HG. Programmed Cell Death in Asthma: Apoptosis, Autophagy, Pyroptosis, Ferroptosis, and Necroptosis. J Inflamm Res 2023; 16:2727-2754. [PMID: 37415620 PMCID: PMC10321329 DOI: 10.2147/jir.s417801] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
Bronchial asthma is a complex heterogeneous airway disease, which has emerged as a global health issue. A comprehensive understanding of the different molecular mechanisms of bronchial asthma may be an efficient means to improve its clinical efficacy in the future. Increasing research evidence indicates that some types of programmed cell death (PCD), including apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, contributed to asthma pathogenesis, and may become new targets for future asthma treatment. This review briefly discusses the molecular mechanism and signaling pathway of these forms of PCD focuses on summarizing their roles in the pathogenesis and treatment strategies of asthma and offers some efficient means to improve clinical efficacy of therapeutics for asthma in the near future.
Collapse
Affiliation(s)
- Lu Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ling-Ling Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Peng-Dou Zheng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Feng-Qin Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhen-Yu Mao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Huo-Jun Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Hui-Guo Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
8
|
Fukaya T, Uto T, Mitoma S, Takagi H, Nishikawa Y, Tominaga M, Choijookhuu N, Hishikawa Y, Sato K. Gut dysbiosis promotes the breakdown of oral tolerance mediated through dysfunction of mucosal dendritic cells. Cell Rep 2023; 42:112431. [PMID: 37099426 DOI: 10.1016/j.celrep.2023.112431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/21/2023] [Accepted: 04/10/2023] [Indexed: 04/27/2023] Open
Abstract
While dysbiosis in the gut is implicated in the impaired induction of oral tolerance generated in mesenteric lymph nodes (MesLNs), how dysbiosis affects this process remains unclear. Here, we describe that antibiotic-driven gut dysbiosis causes the dysfunction of CD11c+CD103+ conventional dendritic cells (cDCs) in MesLNs, preventing the establishment of oral tolerance. Deficiency of CD11c+CD103+ cDCs abrogates the generation of regulatory T cells in MesLNs to establish oral tolerance. Antibiotic treatment triggers the intestinal dysbiosis linked to the impaired generation of colony-stimulating factor 2 (Csf2)-producing group 3 innate lymphoid cells (ILC3s) for regulating the tolerogenesis of CD11c+CD103+ cDCs and the reduced expression of tumor necrosis factor (TNF)-like ligand 1A (TL1A) on CD11c+CD103+ cDCs for generating Csf2-producing ILC3s. Thus, antibiotic-driven intestinal dysbiosis leads to the breakdown of crosstalk between CD11c+CD103+ cDCs and ILC3s for maintaining the tolerogenesis of CD11c+CD103+ cDCs in MesLNs, responsible for the failed establishment of oral tolerance.
Collapse
Affiliation(s)
- Tomohiro Fukaya
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Tomofumi Uto
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Shuya Mitoma
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hideaki Takagi
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yotaro Nishikawa
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Dermatology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Moe Tominaga
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Narantsog Choijookhuu
- Division of Histochemistry and Cell Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshitaka Hishikawa
- Division of Histochemistry and Cell Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan; Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
9
|
Li X, Zhao C, Li C, Zhang M, Xie Y, Feng F, Yao W, Wang N. Detection and analysis of lung microbiota in mice with lung cancer lacking the NLRP3 gene. Biochem Biophys Res Commun 2023; 639:117-125. [PMID: 36481355 DOI: 10.1016/j.bbrc.2022.11.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
To explore whether the lung microbiota have changed in the process of NLRP3 inflammasome promoting cancer, we constructed a murine lung cancer model using tracheal instillation of benzo(a)pyrene and an equal volume of tricaprylin, and characterized lung microbiota in bronchoalveolar lavage fluid from 24 SPF wild-type and NLRP3 gene knockout (NLRP3-/-) C57BL/6 mice. 16SrDNA sequencing was used to analyze the changes in the microbiota. The wild-type and the NLRP3-/- lung cancer group had statistically significant differences in tumor formation rate, tumor number, and tumor size. At the phylum and the genus level, the relative abundance of Proteobacteria and Sphingomonas were the highest in each group respectively. Simpson (P = 0.002) and Shannon (P = 0.008) indexes showed that the diversity of microbiota in the lung cancer group was lower than that in the control group under the NLRP3-/- background. According to the ANOSIM and MRPP analysis, there was a difference between the NLRP3-/- lung cancer group and the NLRP3-/- control group (P < 0.05). The knockout of the NLRP3 gene caused changes in the lung microbiota of mice. There may be a regulatory relationship between the NLRP3 inflammasome and the lung microbiota, which affects the occurrence and development of lung cancer.
Collapse
Affiliation(s)
- Xinyan Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Congcong Zhao
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Chao Li
- President's Office, Shandong Cancer Hospital, Jinan, 250117, China
| | - Mengmeng Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanchen Xie
- Henan Red Cross Blood Center, Zhengzhou, 450053, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Na Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
10
|
Li N, Chen J, Xie S, Zhang M, Shi T, He Y, Jie Z, Su X. Oral antibiotics relieve allergic asthma in post-weaning mice via reducing iNKT cells and function of ADRB2. Front Immunol 2022; 13:1024235. [PMID: 36389706 PMCID: PMC9640740 DOI: 10.3389/fimmu.2022.1024235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
The role of normal gut microbiota in asthma or ovalbumin (OVA)-induced asthma tolerance (OT) remains unclear. Here, we established mouse models of asthma and OT followed by 2 weeks of antibiotic treatment, to clear the gut microbiota. Antibiotic treatment was found to alleviate allergic asthma accompanied with a reduction of invariant natural killer (iNKT) cells. By RNA-seq analysis, we found that β-adrenergic receptor (ADRB) genes, including Adrb1, Adrb2, and Adrb3, were downregulated in asthmatic lungs, but these changes were reversed in OT lungs. Moreover, Adrb2 and Adrb3 were significantly upregulated in asthmatic lungs after antibiotic treatment. Surprisingly, blocking ADRB with propranolol relieved allergic asthma while reducing T helper 2 (Th2) and Treg cell numbers. Further analyses using flow cytometry and immunofluorescence showed that the protein expression level of ADRB2 was higher in asthmatic lungs than that in the control and OT lungs. Notably, dendritic cells (DCs), especially the ADRB2+ DCs, were increased in asthmatic lungs compared to that in the control and OT lungs. In addition, ADRB2+ DCs were significantly reduced following the administration of the ADRB2-specific antagonist ICI118551. Our findings suggest that antibiotic treatment can alleviate OVA-induced allergic asthma via reducing the frequency of iNKT cells and function of ADRB2.
Collapse
Affiliation(s)
- Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Sitao Xie
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- *Correspondence: Xiao Su, ; Zhijun Jie,
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Xiao Su, ; Zhijun Jie,
| |
Collapse
|
11
|
Tuazon JA, Kilburg-Basnyat B, Oldfield LM, Wiscovitch-Russo R, Dunigan-Russell K, Fedulov AV, Oestreich KJ, Gowdy KM. Emerging Insights into the Impact of Air Pollution on Immune-Mediated Asthma Pathogenesis. Curr Allergy Asthma Rep 2022; 22:77-92. [PMID: 35394608 PMCID: PMC9246904 DOI: 10.1007/s11882-022-01034-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Increases in ambient levels of air pollutants have been linked to lung inflammation and remodeling, processes that lead to the development and exacerbation of allergic asthma. Conventional research has focused on the role of CD4+ T helper 2 (TH2) cells in the pathogenesis of air pollution-induced asthma. However, much work in the past decade has uncovered an array of air pollution-induced non-TH2 immune mechanisms that contribute to allergic airway inflammation and disease. RECENT FINDINGS In this article, we review current research demonstrating the connection between common air pollutants and their downstream effects on non-TH2 immune responses emerging as key players in asthma, including PRRs, ILCs, and non-TH2 T cell subsets. We also discuss the proposed mechanisms by which air pollution increases immune-mediated asthma risk, including pre-existing genetic risk, epigenetic alterations in immune cells, and perturbation of the composition and function of the lung and gut microbiomes. Together, these studies reveal the multifaceted impacts of various air pollutants on innate and adaptive immune functions via genetic, epigenetic, and microbiome-based mechanisms that facilitate the induction and worsening of asthma.
Collapse
Affiliation(s)
- J A Tuazon
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, 43210, USA
| | - B Kilburg-Basnyat
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, 27858, USA
| | - L M Oldfield
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
- Department of Synthetic Genomics, Replay Holdings LLC, San Diego, 92121, USA
| | - R Wiscovitch-Russo
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
| | - K Dunigan-Russell
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA
| | - A V Fedulov
- Division of Surgical Research, Department of Surgery, Alpert Medical School, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - K J Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, The James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - K M Gowdy
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Shen X, Zhang H, Xie H, Chen L, Li S, Zheng J, Chai R, Wang Z, Zang Y, He S. Reduced CCR6 +IL-17A +Treg Cells in Blood and CCR6-Dependent Accumulation of IL-17A +Treg Cells in Lungs of Patients With Allergic Asthma. Front Immunol 2021; 12:710750. [PMID: 34497608 PMCID: PMC8419235 DOI: 10.3389/fimmu.2021.710750] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Human regulatory T (Treg) cells play a central role in controlling allergic inflammation in the airways. A reduced number of peripheral Treg cells and decreased suppressive function have been previously reported in the pathogenesis of allergic asthma. However, the characteristic role of specific Treg cell subsets and their mechanisms in the pathogenesis of allergic asthma remain unclear. In this study, we examined the proportion of different Treg cell subsets in both healthy subjects and patients with allergic asthma using flow cytometry and single-cell RNA sequencing. The migration function of the cells was compared using cell sorting and Transwell experiments. Furthermore, two allergen-challenged mouse models and a cell transfer experiment were used to examine the role of these Treg subsets. We found that the proportion of CD25+Foxp3+CD127- Treg cells in the peripheral blood of patients with allergic asthma was lower than in those of healthy subjects. Furthermore, the circulating Treg cells expressed lower levels of CCR6 and IL-17 compared with healthy subjects. The chemokine from the airway mucosa, CCL20, was abundantly expressed, and Transwell experiments further proved that this chemokine promoted CCR6+ Treg cell migration in vitro. A mouse model induced by house dust mite (HDM) revealed that the number of CCR6+ Treg cells in the lung tissue increased remarkably. The incidence of allergic asthma may be related to an increase in Treg cells secreting IL-17 in the lung tissue. Recruited CCR6+ Treg cells are likely to differentiate into Th17-like cells under the Th17 environment present in the lungs. IL-17 derived from Th17-like cells could be associated with the pathology of allergic asthma by promoting Th17 responses, thereby favoring HDM-induced asthma exacerbations.
Collapse
Affiliation(s)
- Xiaokun Shen
- Institute of Translation Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Huiyun Zhang
- Institute of Translation Medicine, Shenyang Medical College, Shenyang, China
| | - Hua Xie
- People's Liberation Army (PLA) Center of Respiratory and Allergic Disease Diagnosing Management, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Liping Chen
- Respiratory Medicine Department, Second Affiliated Hospital of Shenyang Medical College, Shenyang, China
| | - Shinan Li
- Institute of Translation Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Junjuan Zheng
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ruonan Chai
- People's Liberation Army (PLA) Center of Respiratory and Allergic Disease Diagnosing Management, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Zhao Wang
- Institute of Translation Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanyan Zang
- Institute of Translation Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Shaoheng He
- Institute of Translation Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Institute of Translation Medicine, Shenyang Medical College, Shenyang, China
| |
Collapse
|
13
|
Wang LQ, Liu T, Yang S, Sun L, Zhao ZY, Li LY, She YC, Zheng YY, Ye XY, Bao Q, Dong GH, Li CW, Cui J. Perfluoroalkyl substance pollutants activate the innate immune system through the AIM2 inflammasome. Nat Commun 2021; 12:2915. [PMID: 34006824 PMCID: PMC8131593 DOI: 10.1038/s41467-021-23201-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/15/2021] [Indexed: 02/03/2023] Open
Abstract
Perfluoroalkyl substances (PFAS) are widely used in various manufacturing processes. Accumulation of these chemicals has adverse effects on human health, including inflammation in multiple organs, yet how PFAS are sensed by host cells, and how tissue inflammation eventually incurs, is still unclear. Here, we show that the double-stranded DNA receptor AIM2 is able to recognize perfluorooctane sulfonate (PFOS), a common form of PFAS, to trigger IL-1β secretion and pyroptosis. Mechanistically, PFOS activates the AIM2 inflammasome in a process involving mitochondrial DNA release through the Ca2+-PKC-NF-κB/JNK-BAX/BAK axis. Accordingly, Aim2-/- mice have reduced PFOS-induced inflammation, as well as tissue damage in the lungs, livers, and kidneys in both their basic condition and in an asthmatic exacerbation model. Our results thus suggest a function of AIM2 in PFOS-mediated tissue inflammation, and identify AIM2 as a major pattern recognition receptor in response to the environmental organic pollutants.
Collapse
Affiliation(s)
- Li-Qiu Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lin Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi-Yao Zhao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li-Yue Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuan-Chu She
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan-Yan Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Yan Ye
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qing Bao
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chun-Wei Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Dai R, Niu M, Wang N, Wang Y. Syringin alleviates ovalbumin-induced lung inflammation in BALB/c mice asthma model via NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:433-444. [PMID: 33146439 DOI: 10.1002/tox.23049] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
Asthma is an allergic chronic inflammatory disease of the pulmonary airways, characterized by the infiltration of white blood cells and release of inflammatory cytokines of complex pathways linked to its pathogenesis. Syringin extracted from various medicinal plants has been used extensively for the treatment of inflammatory diseases. Hence, this study was conducted to further explore the protective effects of the syringin in ovalbumin (OVA) induced-asthma mice model. OVA-sensitized BALB mice were treated intraperitonealy with three doses (25, 50 and 100 mg/kg) of the syringin which was validated by the alteration in the immunoglobulin E (IgE) levels, cytokines levels, histopathological evaluation inflammatory cell count, lung weight, nitrite (NO) levels, oxidative stress biomarkers and gene markers. The treatment of syringin intensely reduced the increased IgE, inflammatory cytokines, WBC count and restored the antioxidant stress markers OVA stimulated animals. In addition, a significant reduction in inflammation and mucus production was evidenced in histopathological analysis which was further validated by suppression NF-κB pathway activation by syringin. These results suggest that syringin may improve asthma symptoms in OVA-induced mice by modulating NF-κB pathway activation.
Collapse
Affiliation(s)
- Rui Dai
- Department of Pediatric, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Manman Niu
- Department of Pediatric, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ningling Wang
- Department of Pediatric, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Wang
- Department of Pediatric, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Tsang MSM, Hou T, Chan BCL, Wong CK. Immunological Roles of NLR in Allergic Diseases and Its Underlying Mechanisms. Int J Mol Sci 2021; 22:1507. [PMID: 33546184 PMCID: PMC7913164 DOI: 10.3390/ijms22041507] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Our understanding on the immunological roles of pathogen recognition in innate immunity has vastly increased over the past 20 years. Nucleotide-binding oligomerization domain (NOD)-like receptors (NLR) are cytosolic pattern recognition receptors (PRR) that are responsible for sensing microbial motifs and endogenous damage signals in mammalian cytosol for immune surveillance and host defense. The accumulating discoveries on these NLR sensors in allergic diseases suggest that the pathogenesis of allergic diseases may not be confined to the adaptive immune response. Therapy targeting NLR in murine models also shields light on its potential in the treatment of allergies in man. In this review, we herein summarize the recent understanding of the role of NLR sensors and their molecular mechanisms involved in allergic inflammation, including atopic dermatitis and allergic asthma.
Collapse
Affiliation(s)
- Miranda Sin-Man Tsang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; (M.S.-M.T.); (T.H.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China;
| | - Tianheng Hou
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; (M.S.-M.T.); (T.H.)
| | - Ben Chung-Lap Chan
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China;
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; (M.S.-M.T.); (T.H.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China;
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Yan B, Gao J, Guo J, Yang D, Li D. Interleukin-28B dampens protease-induced lung inflammation via IL-25 and TSLP inhibition in epithelial cells. Sci Rep 2020; 10:20973. [PMID: 33262394 PMCID: PMC7708501 DOI: 10.1038/s41598-020-77844-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022] Open
Abstract
Asthma is a chronic respiratory disease with high heterogeneity in human. Different mouse models have been applied for investigation of pathogenesis and treatment of asthma, which target on different cells, receptors and pathways. Interleukin (IL-) 28B, a member of λ-interferons, have been shown to play a protective role in OVA-induced asthma, which is antigen-specific and adaptive immune system dominant. However, the roles of IL-28B in protease-induced asthma, an adaptive immune system independent asthma, are still unclear. Here, we used plant-derived cysteine protease, papain to induce asthma in mice and found that IL-28B was capable of alleviating papain-induced asthma. Papain challenge lead to activation of epithelial cells and production of alarmin, such as IL-25 and thymic stromal lymphopoietin and IL-28B treatment down-regulated their production. Further mechanism was proved to be that IL-28B inhibited the phosphorylation of Erk in epithelial cells via interaction with their receptors. Our results reveal a protective role of IL-28B via regulation of epithelial cells in protease induced asthma.
Collapse
Affiliation(s)
- Bailing Yan
- Department of Respiratory Medicine, The First Hospital of Jilin University, 1Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Jinying Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, 1Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Jia Guo
- Department of Respiratory Medicine, The First Hospital of Jilin University, 1Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Dong Yang
- Department of Gastroenterology, The First Hospital of Jilin University, 1Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Dan Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, 1Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China.
| |
Collapse
|
17
|
Guan M, Ma H, Fan X, Chen X, Miao M, Wu H. Dexamethasone alleviate allergic airway inflammation in mice by inhibiting the activation of NLRP3 inflammasome. Int Immunopharmacol 2019; 78:106017. [PMID: 31780368 DOI: 10.1016/j.intimp.2019.106017] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/11/2019] [Accepted: 10/29/2019] [Indexed: 01/20/2023]
Abstract
Dexamethasone (DEX) is the mainstay treatment for asthma, which is a common chronic airway inflammation disease. However, the mechanism of DEX resolute symptoms of asthma is not completely clear. Here, we aimed to analyze the effect of DEX on airway inflammation in OVA-induced mice and whether this effect is related to the inhibition of the activation of NLRP3 inflammasome. Female (C57BL/6) mice were used to establish the allergic airway inflammation model by inhalation OVA. The number of inflammatory cells in the bronchi alveolar lavage fluid (BALF) was counted by Swiss-Giemsa staining, and the contents of IL-1β, IL-18, IL-5 and IL-17 were detected by ELISA. The degree of inflammatory cells infiltration and mucous cells proliferation in lung tissue were separately observed by H&E and PAS staining. The proteins expression of NLRP3, pro-caspase-1, caspase-1, IL-1β, IL-6 and IL-17 in lung tissue were detected by Western blotting. We found that DEX significantly inhibited OVA-induced inflammatory cells infiltration, airway mucus secretion and goblet cell proliferation in mice. The total and classified numbers of inflammatory cells and the levels of IL-1β, IL-18, IL-5 and IL-17 in the BALF of the experimental group were significantly lower than those of the model group after DEX treatment. DEX also significantly inhibited the activity of NLRP3 inflammasome and reduced the protein contents of Pro-Caspase-1, Caspase-1, Capase-1/Pro-Caspase-1, IL-1β, IL-6 and IL-17 in lung tissues. Our study suggested that DEX alleviates allergic airway inflammation by inhibiting the activity of NLRP3 inflammasome and the levels of IL-1β and IL-18.
Collapse
Affiliation(s)
- Minglong Guan
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Hengli Ma
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| | - Xu Chen
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Manli Miao
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Huimei Wu
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| |
Collapse
|
18
|
Wu HM, Xie QM, Zhao CC, Xu J, Fan XY, Fei GH. Melatonin biosynthesis restored by CpG oligodeoxynucleotides attenuates allergic airway inflammation via regulating NLRP3 inflammasome. Life Sci 2019; 239:117067. [PMID: 31738882 DOI: 10.1016/j.lfs.2019.117067] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
Abstract
AIMS Both CpG oligodeoxynucleotide (CpG-ODN) and melatonin have been reported to induce Th1 response and contribute to allergic asthma resistance. Here, we aimed to reveal how they confer such effect as well as whether they crosstalk with each other. MAIN METHODS Six-week-old Female C57BL/6 mice were challenged by OVA to induce allergic airway inflammation, and were treated with CpG-ODN, CpG-ODN plus Luzindole or melatonin respectively. Bronchoalveolar lavage fluid (BALF) cellularity was classified and counted by Wright's-Giemsa staining. HE and PAS staining were used to analyze airway inflammation. The levels of IL-4, IL-5, IL-13,GM-CSF and IFN-γ, as well as IL-1β and IL-18 were analyzed by ELISA. Protein expressions of ASMT, AANAT, NLRP3, IL-1β and caspase-1 in lung tissue were detected by Western blotting, expression of ASMT and AANAT were further observed by immunohistochemistry. KEY FINDINGS We found that CpG-ODN considerably suppressed OVA-induced airway leukocytes infiltration, goblet cell hyperplasia and Th2 cytokines production. Furthermore, the resolution effect of CpG-ODN on OVA-induced allergic airway inflammation occurred in parallel with decreased-activation of NLRP3 inflammasome and increased biosynthesis of melatonin. Blocking the effect of endogenous melatonin by Luzindole abolished the suppressive effect of CpG-ODN on OVA-induced airway inflammation and activation of NLRP3 inflammasome, suggesting such effect was mediated by endogenous melatonin. Moreover, exogenous melatonin pronouncedly ameliorated airway inflammation and decreased the activation of NLRP3 inflammasome. SIGNIFICANCE These results proven that CpG-ODN protects against allergic airway inflammation via suppressing the activation of NLRP3 inflammasome, and such effect may be resulted from the restored-production of melatonin.
Collapse
Affiliation(s)
- Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China.
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Juan Xu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Xiao-Yun Fan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Guang-He Fei
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China.
| |
Collapse
|
19
|
Jiang S, Wang Q, Wang Y, Song X, Zhang Y. Blockade of CCL2/CCR2 signaling pathway prevents inflammatory monocyte recruitment and attenuates OVA-Induced allergic asthma in mice. Immunol Lett 2019; 214:30-36. [PMID: 31454522 DOI: 10.1016/j.imlet.2019.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022]
Abstract
Recent studies have reported recruitment of inflammatory monocytes by cytokines including chemokine (C-C motif) ligand 2 (CCL2) are critical in allergic responses. We aimed to investigate the role of inflammatory monocytes and CCL2 in mouse model with ovalbumin (OVA)-induced allergic asthma. Mice were sensitized with OVA to induce allergic asthma. The proportion of inflammatory cells in bronchoalveolar lavage fluid (BALF) and peritoneal lavage fluid (PLF) were measured by flow cytometry. The expression of CCL2 and CCL2 receptor (CCR2) were determined by qPCR and western blot. The concentrations of Type 1 helper T (Th1) and Type 2 helper T (Th2) cytokines in PLF were detected by ELISA. Inflammatory monocytes are recruited in PLF, and expression of CCL2 and CCR2 were elevated in OVA-induced mice. In addition, transfer of CCR2 knockdown inflammatory monocytes decreased the levels of allergic asthma biomarkers. Injection of anti-CCL2 or anti-CCR2 antibody decreased the proportion of eosinophils and inflammatory monocytes in BALF. Blockade of CCL2/CCR2 signaling pathway suppressed the allergen-induced Th2 cytokines and enhanced the levels of Th1-associated cytokines. Blockade of CCL2/CCR2 signaling pathway in sensitization-recruited inflammatory monocytes exhibits protective effects in mouse model of OVA-induced allergic asthma by inhibiting the Th2 inflammatory responses.
Collapse
Affiliation(s)
- Shaohong Jiang
- Department of Otolaryngology Head and Neck Surgery, Yuhuangding Hospital, Yantai, 264000, Shandong, China.
| | - Qiang Wang
- Department of Otolaryngology Head and Neck Surgery, Yuhuangding Hospital, Yantai, 264000, Shandong, China.
| | - Yuxin Wang
- Department of Otolaryngology Head and Neck Surgery, Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Xicheng Song
- Department of Otolaryngology Head and Neck Surgery, Yuhuangding Hospital, Yantai, 264000, Shandong, China.
| | - Yu Zhang
- Department of Otolaryngology Head and Neck Surgery, Yuhuangding Hospital, Yantai, 264000, Shandong, China
| |
Collapse
|
20
|
|
21
|
Wang Z, Gao S, Xie J, Li R. Identification of multiple dysregulated metabolic pathways by GC-MS-based profiling of lung tissue in mice with PM 2.5-induced asthma. CHEMOSPHERE 2019; 220:1-10. [PMID: 30572224 DOI: 10.1016/j.chemosphere.2018.12.092] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/05/2018] [Accepted: 12/11/2018] [Indexed: 06/09/2023]
Abstract
The risk of development of asthma, a multi-faceted chronic disease, increases as a result of exposure to PM2.5. However, the mechanism underlying asthma-related metabolic changes caused by PM2.5 exposure is unclear. Here, we investigated the major metabolic changes, metabolic pathways involved, and underlying molecular mechanisms in mice with PM2.5 exposure-induced asthma. Forty-eight adult female mice were randomly assigned to control (C), low concentration-PM2.5 exposure: 0.50 mg kg-1 (L), medium concentration-PM2.5 exposure: 1.58 mg kg-1 (M), and high concentration-PM2.5 exposure: 4.98 mg kg-1 (H) groups. M and H groups presented significantly higher IL-4, IL-8, IL-1β, IL-5, IL-13, and OVA-specific IgE levels, and significantly lower IFN-γ levels, than the C group, as well as significantly increased eosinophil count and MUC5AC expression in the lung tissue. These findings indicate that exposure to medium and high concentrations of PM2.5 induced asthma in mice. Statistical analyses identified 13 asthma-related major metabolites, which were analyzed by gas chromatography-mass spectrometry (GC-MS). Meta Mapp Software revealed 4 major metabolic pathways. PM2.5-induced ATP requirement and oxidative stress may perturb metabolic processes in asthma. The present findings increase our understanding of the toxic effect of PM2.5 in the development of asthma and identify potentially useful biomarkers.
Collapse
Affiliation(s)
- Zhentao Wang
- College of Environment and Resource, Shanxi University, Taiyuan, 030006, PR China
| | - Shaolong Gao
- State Environmental Protection Key Laboratory on Efficient Resource-utilization Techniques of Coal Waste, Institute of Resources and Environment Engineering, Shanxi University, Taiyuan, 030006, PR China
| | - Jingfang Xie
- College of Environment and Resource, Shanxi University, Taiyuan, 030006, PR China.
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, 030006, PR China
| |
Collapse
|
22
|
Huang C, Wang J, Zheng X, Chen Y, Wei H, Sun R, Tian Z. Activation of TLR Signaling in Sensitization-Recruited Inflammatory Monocytes Attenuates OVA-Induced Allergic Asthma. Front Immunol 2018; 9:2591. [PMID: 30510553 PMCID: PMC6252340 DOI: 10.3389/fimmu.2018.02591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
The activation of Toll-like receptor (TLR) signaling is widely reported to be involved in preventing the development of allergic asthma. However, the mechanism of the protective function of TLR signaling remains limited. Here, we studied the mouse model of ovalbumin (OVA)-induced allergic asthma and found that deficiency of TLR signaling or activating TLR signaling with agonist would aggravate or attenuate OVA-induced allergic asthma, respectively, and TLR signaling-mediated protective effect mainly affected the sensitization phase. After OVA/alum sensitization, neutrophils and inflammatory monocytes were recruited into peritoneal cavity and up-regulated TLRs expression. However, adoptive transfer of inflammatory monocytes but not peritoneal macrophages or neutrophils induced allergic symptoms in recipient mice after OVA challenge even without OVA/alum sensitization, and treating the inflammatory monocytes with TLR agonist in vitro before transfer could abolish this effect, indicating that recruited inflammatory monocytes played a determinant role in OVA-induced allergic asthma, and activation of TLR signaling in them could attenuate allergic symptoms. Finally, we found that activation of TLR signaling could increase the expression of T-helper (Th) 1-associated cytokines in inflammatory monocytes. Our results suggest that activation of TLR signaling in sensitization-recruited inflammatory monocytes attenuates OVA-induced allergic asthma by promoting the expression of Th1-associated cytokines.
Collapse
Affiliation(s)
- Chao Huang
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China
| | - Jian Wang
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China
| | - Xiaodong Zheng
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China
| | - Yongyan Chen
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|