1
|
Xu Y, Ding Q, Xie Y, Zhang Q, Zhou Y, Sun H, Qian R, Wang L, Chen X, Gao Y, Yao Y, Xiao Y. Green tea polyphenol alleviates silica particle-induced lung injury by suppressing IL-17/NF-κB p65 signaling-driven inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156238. [PMID: 39550922 DOI: 10.1016/j.phymed.2024.156238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Silicosis, an interstitial lung disease caused by inhalation of silica particles, poses a significant health concern globally. Green tea polyphenol (TP) stands out as a promising therapeutic candidate, yet its specific protective effects and in-depth mechanisms against silicosis have not been thoroughly investigated. PURPOSE This study aimed to systematically assess the protective potential of TP against silicosis and to elucidate the underlying mechanisms of its action. METHODS A combination of physiological, transcriptomic, molecular, and computational techniques was employed. HPLC was used to identify the components of TP, and its antioxidant properties were tested with DPPH and ABTS assays. The effects of TP on lung injury were assessed in silicosis mice using histopathology, qRT-PCR, and western blot. Transcriptomic analysis was applied to explore the differentially expressed genes and pathways in response to TP intervention. In vitro studies with mouse alveolar macrophages (MH-S) examined TP's effects on cell viability, proliferation, apoptosis, and inflammation responses. Integrated qRT-PCR, western blot, immunohistochemistry, and molecular docking were performed to confirm the molecular mechanism underlying the protective effects of TP against silicosis. RESULTS TP effectively attenuated pulmonary inflammation and fibrosis in silicosis mice, as evidenced by significant reductions in inflammation and fibrotic markers. Moreover, TP's therapeutic benefits were linked to its cytoprotective effects on alveolar macrophages, notably its ability to protect MH-S cells from silica particle-induced apoptosis, inhibition of proliferation, and inflammatory response, underscoring its targeted protective effects at the cellular level. Mechanistically, TP exerted its anti-silicosis activity by targeting key pathways implicated in inflammatory responses, notably through the inhibition of the IL-17/NF-κB p65 signaling cascade. Molecular docking simulations corroborated these findings, demonstrating favorable binding affinities between TP's bioactive components (EGC, ECG, and EGCG) and crucial proteins (IL-17A, IL-17F, p65, TNF-α, IL-6, and IL-1β) involved in the IL-17/NF-κB p65 signaling pathway. This pathway inhibition led to a significant decrease in the production of pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, thus mitigated silicosis. CONCLUSION TP demonstrates efficacy in alleviating silica particle-induced lung injury by suppressing inflammation through the IL-17/NF-κB p65 signaling pathway, underscoring its potential as a valuable natural compound for silicosis management.
Collapse
Affiliation(s)
- Yunyi Xu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qionghua Ding
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yuhuan Xie
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qingqing Zhang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yangyu Zhou
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Hairui Sun
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Rui Qian
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Liqun Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xuxi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Gao
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yuqin Yao
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Xiao
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Du YJ, Lu ZW, Li KD, Wang YY, Wu H, Huang RG, Jin X, Wang YY, Wang J, Geng AY, Li BZ. No causal association between pneumoconiosis and three inflammatory immune diseases: a Mendelian randomization study. Front Public Health 2024; 12:1373044. [PMID: 38601492 PMCID: PMC11004292 DOI: 10.3389/fpubh.2024.1373044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Objectives To investigate the causal relationships between pneumoconiosis and rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and gout. Methods The random-effects inverse variance weighted (IVW) approach was utilized to explore the causal effects of the instrumental variables (IVs). Sensitivity analyses using the MR-Egger and weighted median (WM) methods were did to investigate horizontal pleiotropy. A leave-one-out analysis was used to avoid the bias resulting from single-nucleotide polymorphisms (SNPs). Results There was no causal association between pneumoconiosis and SLE, RA or gout in the European population [OR = 1.01, 95% CI: 0.94-1.10, p = 0.74; OR = 1.00, 95% CI: 0.999-1.000, p = 0.50; OR = 1.00, 95% CI: 1.000-1.001, p = 0.55]. Causal relationships were also not found in pneumoconiosis due to asbestos and other mineral fibers and SLE, RA and gout [OR = 1.01, 95% CI: 0.96-1.07, p = 0.66; OR = 1.00, 95% CI: 1.00-1.00, p = 0.68; OR = 1.00, 95% CI: 1.00-1.00, p = 0.20]. Conclusion Our study suggests that pneumoconiosis may have no causal relationship with the three inflammatory immune diseases.
Collapse
Affiliation(s)
- Yu-Jie Du
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Zhang-Wei Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Kai-Di Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Yi-Yu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Hong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Rong-Gui Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Xue Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Yi-Yuan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - An-Yi Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Diseases, Hefei, China
- Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Liu TT, Sun HF, Han YX, Zhan Y, Jiang JD. The role of inflammation in silicosis. Front Pharmacol 2024; 15:1362509. [PMID: 38515835 PMCID: PMC10955140 DOI: 10.3389/fphar.2024.1362509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Silicosis is a chronic illness marked by diffuse fibrosis in lung tissue resulting from continuous exposure to SiO2-rich dust in the workplace. The onset and progression of silicosis is a complicated and poorly understood pathological process involving numerous cells and molecules. However, silicosis poses a severe threat to public health in developing countries, where it is the most prevalent occupational disease. There is convincing evidence supporting that innate and adaptive immune cells, as well as their cytokines, play a significant role in the development of silicosis. In this review, we describe the roles of immune cells and cytokines in silicosis, and summarize current knowledge on several important inflammatory signaling pathways associated with the disease, aiming to provide novel targets and strategies for the treatment of silicosis-related inflammation.
Collapse
Affiliation(s)
| | | | | | - Yun Zhan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW There has been a rapid increase in silicosis cases, particularly related to artificial stone. The key to management is avoidance of silica exposure. Despite this, many develop progressive disease and there are no routinely recommended treatments. This review provides a summary of the literature pertaining to pharmacological therapies for silicosis and examines the plausibility of success of such treatments given the disease pathogenesis. RECENT FINDINGS In-vitro and in-vivo models demonstrate potential efficacy for drugs, which target inflammasomes, cytokines, effector cells, fibrosis, autophagy, and oxidation. SUMMARY There is some evidence for potential therapeutic targets in silicosis but limited translation into human studies. Treatment of silicosis likely requires a multimodal approach, and there is considerable cross-talk between pathways; agents that modulate both inflammation, fibrosis, autophagy, and ROS production are likely to be most efficacious.
Collapse
Affiliation(s)
- Hayley Barnes
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
- Central Clinical School, Monash University, Melbourne
| | - Maggie Lam
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Ryan Hoy
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
| |
Collapse
|
5
|
Morin L, Lecureur V, Lescoat A. Results from omic approaches in rat or mouse models exposed to inhaled crystalline silica: a systematic review. Part Fibre Toxicol 2024; 21:10. [PMID: 38429797 PMCID: PMC10905840 DOI: 10.1186/s12989-024-00573-x] [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: 11/23/2022] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Crystalline silica (cSiO2) is a mineral found in rocks; workers from the construction or denim industries are particularly exposed to cSiO2 through inhalation. cSiO2 inhalation increases the risk of silicosis and systemic autoimmune diseases. Inhaled cSiO2 microparticles can reach the alveoli where they induce inflammation, cell death, auto-immunity and fibrosis but the specific molecular pathways involved in these cSiO2 effects remain unclear. This systematic review aims to provide a comprehensive state of the art on omic approaches and exposure models used to study the effects of inhaled cSiO2 in mice and rats and to highlight key results from omic data in rodents also validated in human. METHODS The protocol of systematic review follows PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Eligible articles were identified in PubMed, Embase and Web of Science. The search strategy included original articles published after 1990 and written in English which included mouse or rat models exposed to cSiO2 and utilized omic approaches to identify pathways modulated by cSiO2. Data were extracted and quality assessment was based on the SYRCLE's Risk of Bias tool for animal studies. RESULTS Rats and male rodents were the more used models while female rodents and autoimmune prone models were less studied. Exposure of animals were both acute and chronic and the timing of outcome measurement through omics approaches were homogeneously distributed. Transcriptomic techniques were more commonly performed while proteomic, metabolomic and single-cell omic methods were less utilized. Immunity and inflammation were the main domains modified by cSiO2 exposure in lungs of mice and rats. Less than 20% of the results obtained in rodents were finally verified in humans. CONCLUSION Omic technics offer new insights on the effects of cSiO2 exposure in mice and rats although the majority of data still need to be validated in humans. Autoimmune prone model should be better characterised and systemic effects of cSiO2 need to be further studied to better understand cSiO2-induced autoimmunity. Single-cell omics should be performed to inform on pathological processes induced by cSiO2 exposure.
Collapse
Affiliation(s)
- Laura Morin
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France
| | - Valérie Lecureur
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France.
| | - Alain Lescoat
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France
- Department of Internal Medicine, Rennes University Hospital, 35000, Rennes, France
| |
Collapse
|
6
|
Martínez-López A, Candel S, Tyrkalska SD. Animal models of silicosis: fishing for new therapeutic targets and treatments. Eur Respir Rev 2023; 32:230078. [PMID: 37558264 PMCID: PMC10424253 DOI: 10.1183/16000617.0078-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023] Open
Abstract
Silicosis as an occupational lung disease has been present in our lives for centuries. Research studies have already developed and implemented many animal models to study the pathogenesis and molecular basis of the disease and enabled the search for treatments. As all experimental animal models used to date have their advantages and disadvantages, there is a continuous search for a better model, which will not only accelerate basic research, but also contribute to clinical aspects and drug development. We review here, for the first time, the main animal models developed to date to study silicosis and the unique advantages of the zebrafish model that make it an optimal complement to other models. Among the main advantages of zebrafish for modelling human diseases are its ease of husbandry, low maintenance cost, external fertilisation and development, its transparency from early life, and its amenability to chemical and genetic screening. We discuss the use of zebrafish as a model of silicosis, its similarities to other animal models and the characteristics of patients at molecular and clinical levels, and show the current state of the art of inflammatory and fibrotic zebrafish models that could be used in silicosis research.
Collapse
Affiliation(s)
- Alicia Martínez-López
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- These authors contributed equally to this work
| | - Sergio Candel
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
- These authors contributed equally to this work
| | - Sylwia D Tyrkalska
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
7
|
Senoo S, Higo H, Taniguchi A, Kiura K, Maeda Y, Miyahara N. Pulmonary fibrosis and type-17 immunity. Respir Investig 2023; 61:553-562. [PMID: 37356133 DOI: 10.1016/j.resinv.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/17/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023]
Abstract
Fibrosis of the lung can occur in idiopathic pulmonary fibrosis, collagen vascular diseases, and hypersensitivity pneumonitis, among other diseases. Transforming growth factor (TGF)-β, vascular epithelial growth factor, fibroblast growth factor, and platelet-derived growth factor contribute to the pathophysiology of fibrosis. TGF-β and other cytokines, including interleukin (IL)-1β, IL-6, and IL-23, activate type-17 immunity, which is involved in pulmonary fibrosis. The components of type-17 immunity include type-17 helper T cells, γδT cells, IL-17A-producing CD8-positive T cells, invariant NKT cells, and group 3 innate lymphoid cells. IL-17A, the main cytokine of type-17 immunity, is able to induce the epithelial-mesenchymal transition in epithelial cells via a production of TGF-β, directly stimulate fibroblasts and fibrocytes, and inhibit autophagy, which otherwise protects against pulmonary fibrosis. IL-23 induces type-17 immunity and plays an important role in the acute exacerbation of pulmonary fibrosis. Clinical studies have also linked type-17 immunity to the pathogenesis of pulmonary fibrosis. Consequently, targeting type-17 immunity may serve as a new therapeutic strategy to prevent the development or exacerbation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Satoru Senoo
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hisao Higo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Akihiko Taniguchi
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Nobuaki Miyahara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan; Department of Medical Technology, Okayama University Academic Field of Health Sciences, Okayama, Japan.
| |
Collapse
|
8
|
Effects of Green Tea Polyphenol Epigallocatechin-3-Gallate on Markers of Inflammation and Fibrosis in a Rat Model of Pulmonary Silicosis. Int J Mol Sci 2023; 24:ijms24031857. [PMID: 36768179 PMCID: PMC9916388 DOI: 10.3390/ijms24031857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/08/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Inhalation of silica particles causes inflammatory changes leading to fibrotizing silicosis. Considering a lack of effective therapy, and a growing information on the wide actions of green tea polyphenols, particularly epigallocatechin-3-gallate (EGCG), the aim of this study was to evaluate the early effects of EGCG on markers of inflammation and lung fibrosis in silicotic rats. The silicosis model was induced by a single transoral intratracheal instillation of silica (50 mg/mL/animal), while controls received an equivalent volume of saline. The treatment with intraperitoneal EGCG (20 mg/kg, or saline in controls) was initiated the next day after silica instillation and was given twice a week. Animals were euthanized 14 or 28 days after the treatment onset, and the total and differential counts of leukocytes in the blood and bronchoalveolar lavage fluid (BALF), wet/dry lung weight ratio, and markers of inflammation, oxidative stress, and fibrosis in the lung were determined. The presence of collagen and smooth muscle mass in the walls of bronchioles and lung vessels was investigated immunohistochemically. Early treatment with EGCG showed some potential to alleviate inflammation, and a trend to decrease oxidative stress-induced changes, including apoptosis, and a prevention of fibrotic changes in the bronchioles and pulmonary vessels. However, further investigations should be undertaken to elucidate the effects of EGCG in the lung silicosis model in more detail. In addition, because of insufficient data from EGCG delivery in silicosis, the positive and eventual adverse effects of this herbal compound should be carefully studied before any preventive use or therapy with EGCG may be recommended.
Collapse
|
9
|
Tyrkalska SD, Pedoto A, Martínez-López A, Ros-Lucas JA, Mesa-Del-Castillo P, Candel S, Mulero V. Silica crystals activate toll-like receptors and inflammasomes to promote local and systemic immune responses in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104523. [PMID: 36055417 DOI: 10.1016/j.dci.2022.104523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Silica crystals are potent activators of the inflammasome that cause a fibrotic lung disease, called silicosis, with no effective treatment available. We report here that injection of silica crystals into the hindbrain ventricle of zebrafish embryos led to the initiation of local and systemic immune responses driven through both Toll-like receptors (TLR)- and inflammasome-dependent signaling pathways, followed by induction of pro-fibrotic markers. Genetic and pharmacological analysis revealed that the Nlrp3 inflammasome regulated silica crystal-induced inflammation and pyroptotic cell death, but not emergency myelopoiesis. In addition, Cxcl8a/Cxcr2-dependent recruitment of myeloid cells to silica crystals was required to promote emergency myelopoiesis and systemic inflammation. The zebrafish model of silicosis developed here shed light onto the molecular mechanisms involved in the activation of the immune system by silica crystals.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Annamaria Pedoto
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Alicia Martínez-López
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - José A Ros-Lucas
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Servicio de Neumología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Pablo Mesa-Del-Castillo
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain; Servicio de Reumatología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Sergio Candel
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
10
|
Liu Y, Zhao N, Xu Q, Deng F, Wang P, Dong L, Lu X, Xia L, Wang M, Chen Z, Zhou J, Zuo D. MBL Binding with AhR Controls Th17 Immunity in Silicosis-Associated Lung Inflammation and Fibrosis. J Inflamm Res 2022; 15:4315-4329. [PMID: 35923908 PMCID: PMC9342710 DOI: 10.2147/jir.s357453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
Objective Mannan-binding lectin (MBL), a soluble pattern recognition molecule of the innate immune system, is primarily synthesized in the liver and secreted into the circulation. Low serum level of MBL has been reported to be related to an increased risk of lung diseases. Herein, we aimed to investigate the function of MBL in silicosis-associated pulmonary inflammation. Methods Serum collected from silicosis patients was tested for correlation between serum MBL levels and Th17 immunity. In vitro studies were performed to further demonstrated the effect of MBL on Th17 polarization. Silica was intratracheally injected in wild type (WT) or MBL-deficient (MBL–/–) mice to induce silicosis-associated lung inflammation and fibrosis. Th17 response was evaluated to explore the effect of MBL on silicosis in vivo. Results Silicosis patients with high serum MBL levels displayed ameliorative lung function. We demonstrated that serum MBL levels negatively correlated to Th17 cell frequency in silicosis patients. MBL protein markedly reduced expression of IL-17 but enhanced expression of Foxp3 in CD4+ T cells in vitro when subjected to Th17 or Treg polarizing conditions, respectively. The presence of MBL during Th17 cell polarization significantly limited aryl hydrocarbon receptor (AhR) expression and suppressed the signal transducer and activator of transcription 3 (STAT3) phosphorylation. Treatment with the AhR antagonist abolished the effect of MBL on Th17 response. Strikingly, MBL directly bound to AhR and affected its nuclear translocation. Furthermore, MBL–/– mice displayed elevated Th17 cell levels compared with WT mice in response to the silica challenge. The CD4+ T lymphocytes from silica-administrated MBL–/– mice exhibited more AhR expression than the wild-type counterparts. Conclusion Our study suggested that MBL limited the Th17 immunity via controlling the AhR/STAT3 pathway, thus providing new insight into silicosis and other inflammatory diseases in patients with MBL deficiency.
Collapse
Affiliation(s)
- Yunzhi Liu
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Na Zhao
- Department of Medical Laboratory, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong, 510399, People’s Republic of China
| | - Qishan Xu
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Fan Deng
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Ping Wang
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Lijun Dong
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Xiao Lu
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Lihua Xia
- Department of Medical Laboratory, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong, 510399, People’s Republic of China
| | - Mingyong Wang
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang, 453003, People’s Republic of China
| | - Zhengliang Chen
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Jia Zhou
- Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Correspondence: Jia Zhou, Guangdong Province Key Laboratory of Proteomics, Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China, Tel +86-20-61648220, Fax +86-20-61648221, Email
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
- Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People’s Republic of China
- Daming Zuo, Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China, Tel +86-20-61648552, Fax + 86-20-61648221, Email
| |
Collapse
|
11
|
Li C, He YY, Zhang YT, You YC, Yuan HY, Wei YG, Chen X, Chen J. Tauroursodeoxycholic acid (TUDCA) disparate pharmacological effects to lung tissue-resident memory T cells contribute to alleviated silicosis. Biomed Pharmacother 2022; 151:113173. [PMID: 35623165 DOI: 10.1016/j.biopha.2022.113173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2022] [Accepted: 05/22/2022] [Indexed: 11/02/2022] Open
Abstract
Exposure to crystalline silica (CS) results in a persistent pulmonary inflammatory response, which results in abnormal tissue repair and excessive matrix deposition. Due to vague pathogenesis, there is virtually no practical therapeutic approach. Here we showed the pharmacological effects of TUDCA on CS-induced pulmonary inflammation and fibrosis. It also helped a faster recovery of CS-impaired pulmonary function. Mechanistically, TUDCA suppressed interferon (IFN)-γ and interleukin (IL)-17A productions by pulmonary helper T (Th) cells. We demonstrated that CS-boosted cytokine-producing Th cells were effector memory (TEM) phenotype. TUDCA decreased the pathogenic TEM cells expansion in the lung. Using in vivo labeling method, we discovered the TEM cells were lung tissue residency with CD103 expression. TUDCA's anti-fibrotic effects were linked to decreasing IFN-γ producing CD103- TEM-like and IL-17A producing CD103+ TRM-like T cells as well as restricting TRM-like Treg cells in the lung. Specifically, TUDCA could restrain CD103+ TRM-like Treg cell proliferation but not limit the CD103- ones. Further characterization study proved that though the Tregs originally came from the thymus, the expressing levels of ST-2 were different, which provides insights into TUDCA's various effects on cell proliferation. Collectively, our data paved the way to understanding the pathogenesis of silicosis and may provide new treatments for this pulmonary fibrotic disease.
Collapse
Affiliation(s)
- Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| | - Yang-Yang He
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Yu-Ting Zhang
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Yi-Chuan You
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Hao-Yang Yuan
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Yun-Geng Wei
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Xi Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| |
Collapse
|
12
|
Patel D, Challagundla N, Mandaliya D, Yadav S, Naik O, Dalai P, Shah D, Vora H, Agrawal-Rajput R. Caspase-1 inhibition by YVAD generates tregs pivoting IL-17 to IL-22 response in β-glucan induced airway inflammation. Immunopharmacol Immunotoxicol 2022; 44:316-325. [PMID: 35225131 DOI: 10.1080/08923973.2022.2043899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 02/13/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND During Aspergillus fumigatus mediated lung inflammation, NLRP3 inflammasome is rapidly activated that aggravates IL-1β production contributing to lung inflammation. Previously, we have shown the protective role of SYK-1 inhibition in inhibiting inflammasome activation during lung inflammation. In the current manuscript, we explored the protective role of direct caspase-1 inhibition during β-glucan-induced lung inflammation. METHODS We have mimicked the lung inflammation by administering intranasal β-glucan in mice model. YVAD was used for caspase-1 inhibition. RESULTS We have shown that caspase-1 inhibition by YVAD did not alter inflammasome independent inflammatory cytokines, while it significantly reduced inflammasome activation and IL-1β secretion. Caspase-1 inhibited bone marrow derived dendritic cells (BMDCs), co-cultured with T cells showed decreased T-cell proliferation and direct them to secrete high TGF-β and IL-10 compared to the T cells co-cultured with β-glucan primed dendritic cells. Caspase-1 inhibition in BMDCs also induced IL-22 secretion from CD4+T cells. Caspase-1 inhibition in intranasal β-glucan administered mice showed decreased tissue damage, immune cell infiltration and IgA secretion compared to control mice. Further, splenocytes challenged with β-glucan show high IL-10 secretion and increased FOXp3 and Ahr indicating an increase in regulatory T cells on caspase-1 inhibition. CONCLUSION Caspase-1 inhibition can thus be an attractive target to prevent inflammation mediated tissue damage during Aspergillus fumigatus mouse model and can be explored as an attractive therapeutic strategy.HIGHLIGHTSCaspase-1 inhibition protects lung damage from inflammation during β-glucan exposureCaspase-1 inhibition in dendritic cells decreases IL-1β production resulting in decreased pathogenic Th17Caspase-1 inhibition promotes regulatory T cells thereby inhibiting lung inflammation.
Collapse
Affiliation(s)
- Divyesh Patel
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Naveen Challagundla
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Dipeeka Mandaliya
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Omkar Naik
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Parameswar Dalai
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Dhruvi Shah
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Hima Vora
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| |
Collapse
|
13
|
Lam M, Mansell A, Tate MD. Another One Fights the Dust - Targeting the NLRP3 Inflammasome for the Treatment of Silicosis. Am J Respir Cell Mol Biol 2022; 66:601-611. [PMID: 35290170 DOI: 10.1165/rcmb.2021-0545tr] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Silicosis is a multifaceted lung disease, characterised by persistent inflammation and structural remodelling. Despite its poor prognosis, there are no treatments currently available for patients with silicosis. Recent pre-clinical findings in models of lung fibrosis have suggested a major role for the nucleotide binding domain and leucine-rich repeat pyrin domain containing 3 (NLRP3) inflammasome in silica-driven inflammation and fibrosis. This review outlines the beneficial effects of targeting the NLRP3 inflammasome in in vitro cell experiments and in in vivo animal models, whereby inflammation and fibrosis are abrogated following NLRP3 inflammasome inhibition. While preclinical evidence is promising, studies which explore NLRP3 inflammasomes in the clinical setting are warranted. In particular, there is still a need to identify biomarkers which may be helpful for the early detection of silicosis and to fully elucidate mechanisms underlying these beneficial effects to further develop or repurpose existing anti-NLRP3 drugs as novel treatments that limit disease progression.
Collapse
Affiliation(s)
- Maggie Lam
- Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases, 366840, Clayton, Victoria, Australia.,Monash University , Department of Molecular and Translational Sciences, Clayton, Victoria, Australia
| | - Ashley Mansell
- Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases, 366840, Clayton, Victoria, Australia.,Monash Univerisity, Department of Molecular and Translational Sciences, Clayton, Victoria, Australia.,Adiso Therapeutics Inc, Concord, Massachusetts, United States
| | - Michelle D Tate
- Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases, 366840, Clayton, Victoria, Australia.,Monash University, Department of Molecular and Translational Sciences, Clayton, Victoria, Australia;
| |
Collapse
|
14
|
Hasan M, Paul NC, Paul SK, Saikat ASM, Akter H, Mandal M, Lee SS. Natural Product-Based Potential Therapeutic Interventions of Pulmonary Fibrosis. Molecules 2022; 27:1481. [PMID: 35268581 PMCID: PMC8911636 DOI: 10.3390/molecules27051481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary fibrosis (PF) is a disease-refractive lung condition with an increased rate of mortality. The potential factors causing PF include viral infections, radiation exposure, and toxic airborne chemicals. Idiopathic PF (IPF) is related to pneumonia affecting the elderly and is characterized by recurring scar formation in the lungs. An impaired wound healing process, defined by the dysregulated aggregation of extracellular matrix components, triggers fibrotic scar formation in the lungs. The potential pathogenesis includes oxidative stress, altered cell signaling, inflammation, etc. Nintedanib and pirfenidone have been approved with a conditional endorsement for the management of IPF. In addition, natural product-based treatment strategies have shown promising results in treating PF. In this study, we reviewed the recently published literature and discussed the potential uses of natural products, classified into three types-isolated active compounds, crude extracts of plants, and traditional medicine, consisting of mixtures of different plant products-in treating PF. These natural products are promising in the treatment of PF via inhibiting inflammation, oxidative stress, and endothelial mesenchymal transition, as well as affecting TGF-β-mediated cell signaling, etc. Based on the current review, we have revealed the signaling mechanisms of PF pathogenesis and the potential opportunities offered by natural product-based medicine in treating PF.
Collapse
Affiliation(s)
- Mahbub Hasan
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
- Department of Oriental Biomedical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Korea
| | - Nidhan Chandra Paul
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Shamrat Kumar Paul
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Abu Saim Mohammad Saikat
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Hafeza Akter
- Pharmacology and Toxicology Research Division, Health Medical Science Research Foundation, Dhaka 1207, Bangladesh;
| | - Manoj Mandal
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Sang-Suk Lee
- Department of Oriental Biomedical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Korea
| |
Collapse
|
15
|
Liu T, Bao R, Wang Q, Hao W, Liu Y, Chang S, Wang M, Li Y, Liu Z, Sun Y. SiO 2-induced ferroptosis in macrophages promotes the development of pulmonary fibrosis in silicosis models. Toxicol Res (Camb) 2022; 11:42-51. [PMID: 35237410 PMCID: PMC8882780 DOI: 10.1093/toxres/tfab105] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 09/06/2023] Open
Abstract
Silicosis is a devastating disease that, without effective treatment, endangers the health of miners. Therefore, studies exploring the pathogenesis of SiO2-induced pulmonary fibrosis are necessary to develop treatments for silicosis. Although macrophages are known to play a pivotal role in SiO2-induced pulmonary fibrosis, the underlying mechanism remains unknown. Here, we explored whether ferroptosis was involved in SiO2-induced pulmonary fibrosis. To this end, C57BL/6 mice and mouse macrophage (RAW264.7) cells and mouse lung fibroblast (MLF) cells were subjected to iron content, cell viability, enzyme-linked immunosorbent assay, immunofluorescence staining, histological, western blotting, quantitative reverse transcription-PCR, reactive oxygen species, and lipid peroxidation analysis. In vivo, SiO2 was found to damage the lung alveolar structure, cause infiltration of inflammatory cells, and facilitate fibrosis. Additionally, it increased the iron concentration and lipid peroxidation as well as altered the expression of ferroptosis-related genes and the mitochondrial morphology in macrophages. In vitro, ferroptosis occurred in SiO2-treated RAW264.7 cells, which showed iron overload, lipid peroxidation, and gene alterations. Furthermore, ferrostatin-1 (Fer-1) attenuated ferroptosis in SiO2-treated RAW264.7 cells by inhibiting lipid peroxidation and cell death and regulating ferroptosis-related genes expression, in addition to attenuating the secretion of pro-fibrotic cytokines and fibrosis. Collectively, SiO2 induces ferroptosis in macrophages, which leads to the secretion of pro-fibrotic cytokines and fibrosis.
Collapse
Affiliation(s)
- Taiyang Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Rui Bao
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Qiushi Wang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Wei Hao
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yaoyang Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Sirong Chang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Meng Wang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yuanyuan Li
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Zhihong Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yue Sun
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| |
Collapse
|
16
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J Adamcakova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | |
Collapse
|
17
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J ADAMCAKOVA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - D MOKRA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| |
Collapse
|
18
|
Extracellular cathepsin Z signals through the α 5 integrin and augments NLRP3 inflammasome activation. J Biol Chem 2021; 298:101459. [PMID: 34864055 PMCID: PMC8753182 DOI: 10.1016/j.jbc.2021.101459] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 12/31/2022] Open
Abstract
Respiratory silicosis is a preventable occupational disease that develops secondary to the aspiration of crystalline silicon dioxide (silica) into the lungs, activation of the NLRP3 inflammasome, and IL-1β production. Cathepsin Z has been associated with the development of inflammation and IL-1β production; however, the mechanism of how cathepsin Z leads to IL-1β production is unknown. Here, the requirement for cathepsin Z in silicosis was determined using WT mice and mice deficient in cathepsin Z. The activation of the NLRP3 inflammasome in macrophages was studied using WT and cathepsin Z-deficient bone marrow-derived murine dendritic cells and the human monocytic cell line THP-1. The cells were activated with silica, and IL-1β release was determined using enzyme-linked immunosorbent assay or IL-1β bioassays. The relative contribution of the active domain or integrin-binding domain of cathepsin Z was studied using recombinant cathepsin Z constructs and the α5 integrin neutralizing antibody. We report that the lysosomal cysteine protease cathepsin Z potentiates the development of inflammation associated with respiratory silicosis by augmenting NLRP3 inflammasome-derived IL-1β expression in response to silica. The secreted cathepsin Z functions nonproteolytically via the internal integrin-binding domain to impact caspase-1 activation and the production of active IL-1β through integrin α5 without affecting the transcription levels of NLRP3 inflammasome components. This work reveals a regulatory pathway for the NLRP3 inflammasome that occurs in an outside-in fashion and provides a link between extracellular cathepsin Z and inflammation. Furthermore, it reveals a level of NLRP3 inflammasome regulation that has previously only been found downstream of extracellular pathogens.
Collapse
|
19
|
Melo EM, Oliveira VLS, Boff D, Galvão I. Pulmonary macrophages and their different roles in health and disease. Int J Biochem Cell Biol 2021; 141:106095. [PMID: 34653619 DOI: 10.1016/j.biocel.2021.106095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 12/20/2022]
Abstract
Macrophages are a heterogeneous population of myeloid cells with phenotype and function modulated according to the microenvironment in which they are found. The lung resident macrophages known as Alveolar Macrophages (AM) and Interstitial Macrophages (IM) are localized in two different compartments. During lung homeostasis, macrophages can remove inhaled particulates, cellular debris and contribute to some metabolic processes. Macrophages may assume a pro-inflammatory phenotype after being classically activated (M1) or anti-inflammatory when being alternatively activated (M2). M1 and M2 have different transcription profiles and act by eliminating bacteria, viruses and fungi from the host or repairing the damage triggered by inflammation, respectively. Nevertheless, macrophages also may contribute to lung damage during persistent inflammation or continuous exposure to antigens. In this review, we discuss the origin and function of pulmonary macrophages in the context of homeostasis, infectious and non-infectious lung diseases.
Collapse
Affiliation(s)
- Eliza Mathias Melo
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vivian Louise Soares Oliveira
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Daiane Boff
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Izabela Galvão
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
20
|
Zhang Y, Huang S, Tan S, Chen M, Yang S, Chen S. 3-methyadenine inhibits lipopolysaccharides-induced pulmonary inflammation at the early stage of silicosis via blocking NF-κB signaling pathway. Toxicol Ind Health 2021; 37:662-673. [PMID: 34565256 DOI: 10.1177/07482337211039426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Occupational exposure to silica dust is related to pulmonary inflammation and silicosis. Lipopolysaccharides (LPSs) could aggravate apoptosis in alveolar macrophages (AMs) of human silicosis through autophagy, yet how the reduction of autophagy attenuated LPS-induced lung injury and the related mechanisms need to be investigated. In the study, we aim to understand the role of 3-methyladenine (3-MA), an inhibitor of autophagy, in LPS-mediated inflammatory responses and fibrosis. We collected AMs from observers/silicosis patients. The results showed that LPS induced NF-κB-related pulmonary inflammation in observers and silicosis patients, as confirmed by an increase in the expression of IL-1β, IL-6, TNF-α, and p65, which could be inhibited by 3-MA treatment. In mice models, at the early stage (7d) of silicosis, but not the late (28d) stage, blocking autophagy reversed the increased levels of IL-1β, IL-6, TNF-α, and p65 caused by LPS. Mechanism study revealed that LPS triggered the expression of LC3 II, p62, and cleaved caspase-3 at the early stage exposed to silica, which could be restored by 3-MA, while there was no difference in the expression of LAMP1 either at the early or late stage of silicosis in different groups. Similarly, 3-MA treatment did not prevent fibrosis characterized by destroyed alveoli, collagen deposition, and increased expression of α-SMA and Col-1 induced by LPS at the late stage of silicosis. The results suggested that 3-MA has a role in the protection of lung injury at the early stage of silicosis and provided an experimental basis for preventive strategies of pulmonary inflammation and silicosis.
Collapse
Affiliation(s)
- Yujing Zhang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shuai Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shiyi Tan
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Mingke Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shang Yang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shi Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| |
Collapse
|
21
|
Extracellular Heat Shock Proteins as Therapeutic Targets and Biomarkers in Fibrosing Interstitial Lung Diseases. Int J Mol Sci 2021; 22:ijms22179316. [PMID: 34502225 PMCID: PMC8430559 DOI: 10.3390/ijms22179316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Interstitial lung diseases (ILDs) include a large number of diseases and causes with variable outcomes often associated with progressive fibrosis. Although each of the individual fibrosing ILDs are rare, collectively, they affect a considerable number of patients, representing a significant burden of disease. Idiopathic pulmonary fibrosis (IPF) is the typical chronic fibrosing ILD associated with progressive decline in lung. Other fibrosing ILDs are often associated with connective tissues diseases, including rheumatoid arthritis-ILD (RA-ILD) and systemic sclerosis-associated ILD (SSc-ILD), or environmental/drug exposure. Given the vast number of progressive fibrosing ILDs and the disparities in clinical patterns and disease features, the course of these diseases is heterogeneous and cannot accurately be predicted for an individual patient. As a consequence, the discovery of novel biomarkers for these types of diseases is a major clinical challenge. Heat shock proteins (HSPs) are molecular chaperons that have been extensively described to be involved in fibrogenesis. Their extracellular forms (eHSPs) have been recently and successfully used as therapeutic targets or circulating biomarkers in cancer. The current review will describe the role of eHSPs in fibrosing ILDs, highlighting the importance of these particular stress proteins to develop new therapeutic strategies and discover potential biomarkers in these diseases.
Collapse
|
22
|
Adamcakova J, Mokra D. New Insights into Pathomechanisms and Treatment Possibilities for Lung Silicosis. Int J Mol Sci 2021; 22:ijms22084162. [PMID: 33920534 PMCID: PMC8072896 DOI: 10.3390/ijms22084162] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Inhalation of silica particles is an environmental and occupational cause of silicosis, a type of pneumoconiosis. Development of the lung silicosis is a unique process in which the vicious cycle of ingestion of inhaled silica particles by alveolar macrophages and their release triggers inflammation, generation of nodular lesions, and irreversible fibrosis. The pathophysiology of silicosis is complex, and interactions between the pathomechanisms have not been completely understood. However, elucidation of silica-induced inflammation cascades and inflammation-fibrosis relations has uncovered several novel possibilities of therapeutic targeting. This article reviews new information on the pathophysiology of silicosis and points out several promising treatment approaches targeting silicosis-related pathways.
Collapse
|
23
|
Salum KCR, Castro MCS, Nani ÂSF, Kohlrausch FB. Is individual genetic susceptibility a link between silica exposure and development or severity of silicosis? A systematic review. Inhal Toxicol 2020; 32:375-387. [PMID: 33006295 DOI: 10.1080/08958378.2020.1825569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Silicosis is a lung disease of fibrotic nature resulting from the inhalation and deposition of dust containing crystalline silica. Subjects exposed to the same environmental factors may show distinct radiological manifestations, and since silicosis is known as a multifactorial disease, it is plausible that individual genetic susceptibility may play a role in the pathology. This review of the literature aims to provide an assessment of the present data on the genetic association studies in silicosis and describe the genes that potentially might influence silicosis susceptibility in silica-exposed individuals. METHODS We accessed the database of PubMed for articles published in English about interindividual genetic susceptibility to silicosis using terms related to the subject matter. RESULTS Following the evaluation process, 28 studies were included in this systematic review, including 23 original studies and 5 meta-analyses. CONCLUSIONS Regardless of the advances in the knowledge of the importance of gene variations in silicosis, more studies need to be performed, in particular, special polygenic and genome-wide investigations.
Collapse
Affiliation(s)
- Kaio Cezar Rodrigues Salum
- Programa de Pós-Graduação em Ciências e Biotecnologia, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Marcos Cesar Santos Castro
- Programa de Pós-Graduação em Ciências e Biotecnologia, Universidade Federal Fluminense (UFF), Niterói, Brazil.,Hospital Universitário Antônio Pedro, Universidade Federal Fluminense (UFF), Niterói, Brazil.,Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | | | - Fabiana Barzotto Kohlrausch
- Programa de Pós-Graduação em Ciências e Biotecnologia, Universidade Federal Fluminense (UFF), Niterói, Brazil
| |
Collapse
|
24
|
Tan S, Yang S, Chen M, Wang Y, Zhu L, Sun Z, Chen S. Lipopolysaccharides promote pulmonary fibrosis in silicosis through the aggravation of apoptosis and inflammation in alveolar macrophages. Open Life Sci 2020; 15:598-605. [PMID: 33817248 PMCID: PMC7874552 DOI: 10.1515/biol-2020-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Alveolar macrophages (AMs) play an important defensive role by removing dust and bacteria from alveoli. Apoptosis of AMs is associated with lung fibrosis; however, the relationship between this apoptotic event and environmental factors, such as the presence of lipopolysaccharides (LPSs) in the workplace, has not yet been addressed. To investigate whether exposure to LPS can exacerbate fibrosis, we collected AMs from 12 male workers exposed to silica and incubated them in the presence and absence of LPS for 24 h. We show that the levels of cleaved caspase-3 and pro-inflammatory cytokines interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha were increased in these AMs following LPS treatment. Moreover, we demonstrate that LPS exposure aggravated apoptosis and the release of inflammatory factors in AMs in a mouse model of silicosis, which eventually promoted pulmonary fibrosis. These results suggest that exposure to LPS may accelerate the progression of pulmonary fibrosis in silicosis by increasing apoptosis and inflammation in AMs.
Collapse
Affiliation(s)
- Shiyi Tan
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Shang Yang
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Mingke Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Yurun Wang
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Li Zhu
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Beidaihe, 066100, Hebei, China
| | - Zhiqian Sun
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Beidaihe, 066100, Hebei, China
| | - Shi Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, No. 371 Tongzipo Road, Changsha, 410013, Hunan, China
| |
Collapse
|
25
|
Ma J, Xu Y, Li W, Zhou Y, Wang D, Yang M, Wang B, Chen W. High-mobility group box 1 promotes epithelial-to-mesenchymal transition in crystalline silica induced pulmonary inflammation and fibrosis. Toxicol Lett 2020; 330:134-143. [PMID: 32428545 DOI: 10.1016/j.toxlet.2020.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/02/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022]
Abstract
Silicosis is an inflammatory and fibrotic lung disease caused by prolonged inhalation of silica. The potential role of high-mobility group box-1 (HMGB-1) and its underlying mechanisms in silicosis remain unclear. In this study, intratracheal instillation of a silica suspension was used to establish silicosis in male C57BL/6 mice. To elucidate the effects of HMGB-1 on the pathogenesis of silicosis, we used HMGB-1 neutralizing antibody (anti-HMGB-1) and recombinant HMGB-1 (rmHMGB-1) to abrogate or increase the HMGB-1 levels, respectively. At days 7, 28, and 84, the accumulation of macrophages and neutrophils decreased by anti-HMGB-1 treatment. The expression levels of interleukin-6 and tumor necrosis factor-α in lung increased in response to silica exposure across three time points; anti-HMGB-1 could alleviate those expressions at day 28 and 84. In contrast, rmHMGB-1 aggravated this process. At days 28 and 84, the protein expression of fibronectin and col1a1 decreased in the silica + anti-HMGB-1 groups but increased in silica + rmHMGB-1 groups compared to mice with silica alone. Further study suggested that HMGB-1-mediated epithelial-mesenchymal transition participated in the development of silicosis. In conclusion, the findings demonstrate that HMGB-1 participates in the pathogenesis of silicosis and may represent a potential therapeutic target for the treatment of silicosis.
Collapse
Affiliation(s)
- Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yiju Xu
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Li
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yun Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dongming Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Meng Yang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
26
|
Zhao Y, Hao C, Bao L, Wang D, Li Y, Qu Y, Ding M, Zhao A, Yao W. Silica particles disorganize the polarization of pulmonary macrophages in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 193:110364. [PMID: 32114243 DOI: 10.1016/j.ecoenv.2020.110364] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 06/10/2023]
Abstract
Silicosis is a fatal fibrotic lung disease caused by long-term silica particle exposure, in which pulmonary macrophages play an important role. However, the relationship between macrophage polarization and silicosis remains unclear. We established an experimental silicosis mouse model to investigate macrophage polarization during silicosis development. C57BL/c mice were exposed to silica by intra-tracheal instillation and sacrificed at different time points. Lung tissues and bronchoalveolar lavage fluid were collected for flow cytometry, quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assays, western blotting, and histology examinations. The polarization of pulmonary macrophages was dysregulated during silicosis development. In the early stage of silicosis, M1 macrophages were induced and played a leading role in eliciting inflammatory; in the late stage, M2 macrophages were induced to promote tissue repair. Levels of several cytokines in lung tissue microenvironment changed with macrophage polarization. Inflammatory cytokines such as tumor necrosis factor-α and interleukin (IL)-1β and IL-6 were upregulated in the inflammation stage, while the anti-inflammatory cytokine IL-10 was upregulated in the fibrosis stage. Furthermore, we found that STAT (signal transducer and activator of transcription) and IRF (interferon regulatory factor) signaling pathway were involved in the regulation of macrophage polarization in silicosis. In summary, macrophage polarization is closely related to the occurrence and development of silicosis and may be a key point for further elucidating silicosis pathogenesis.
Collapse
Affiliation(s)
- Youliang Zhao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Changfu Hao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Lei Bao
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Di Wang
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yiping Li
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yaqian Qu
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Mingcui Ding
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Ahui Zhao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Wu Yao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| |
Collapse
|
27
|
Mulay SR, Steiger S, Shi C, Anders HJ. A guide to crystal-related and nano- or microparticle-related tissue responses. FEBS J 2020; 287:818-832. [PMID: 31829497 DOI: 10.1111/febs.15174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
Crystals and nano- and microparticles form inside the human body from intrinsic proteins, minerals, or metabolites or enter the body as particulate matter from occupational and environmental sources. Associated tissue injuries and diseases mostly develop from cellular responses to such crystal deposits and include inflammation, cell necrosis, granuloma formation, tissue fibrosis, and stone-related obstruction of excretory organs. But how do crystals and nano- and microparticles trigger these biological processes? Which pathomechanisms are identical across different particle types, sizes, and shapes? In addition, which mechanisms are specific to the atomic or molecular structure of crystals or to specific sizes or shapes? Do specific cellular or molecular mechanisms qualify as target for therapeutic interventions? Here, we provide a guide to approach this diverse and multidisciplinary research domain. We give an overview about the clinical spectrum of crystallopathies, about shared and specific pathomechanisms as a conceptual overview before digging deeper into the specialty field of interest.
Collapse
Affiliation(s)
- Shrikant R Mulay
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Stefanie Steiger
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Germany
| | - Chongxu Shi
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Germany
| |
Collapse
|
28
|
Salum KCR, Castro MCS, Moreira VB, Nani ASF, Kohlrausch FB. Interleukin 1α and 1β gene variations are associated with tuberculosis in silica exposed subjects. Am J Ind Med 2020; 63:74-84. [PMID: 31692000 DOI: 10.1002/ajim.23066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Silicosis is a fibrotic lung disease resulting from the inhalation of crystalline silica and can be classified as simple or complicated according to the International Labour Organization criteria. Furthermore, individuals exposed to crystalline silica also have a higher risk for the development of tuberculosis (Tb). The contribution of inflammatory cytokines to the risk of silicosis and Tb in different populations has previously been reported. Since genetic background might be related to susceptibility to silicosis and Tb, the study of polymorphisms within IL-1α, IL-1β, and tumor necrosis factor protein-coding genes may contribute to elucidating the genetic basis of these diseases. METHODS Single nucleotide polymorphisms (SNPs) were genotyped by polymerase chain reaction using restriction fragment length polymorphism or by Taqman methodology, in a sample of 102 silica-exposed patients from Brazil. RESULTS No significant associations were observed between the SNPs studied and the severity of silicosis. However, significant associations were found between Tb and the C allele (odds ratio [OR] = 1.93, 95% confidence interval [CI], 1.01-3.73) and the CC genotype (OR = 2.34, 95% CI, 1.04-5.31) of IL1A -899C>T. The IL1B +3954C>T polymorphism also showed an association with Tb (T allele dominant model OR = 2.38, 95% CI, 1.04-5.41). CONCLUSION These preliminary results demonstrate that the IL1A and IL1B gene variations may contribute to some extent to susceptibility to Tb, but not silicosis. However, additional studies are still needed to confirm these results.
Collapse
Affiliation(s)
| | - Marcos Cesar Santos Castro
- Departamento de Medicina Clínica, Hospital Universitário Antônio PedroUniversidade Federal FluminenseNiterói Brazil
- Ambulatório de Pneumologia, Hospital Universitário Pedro ErnestoUniversidade do Estado do Rio de JaneiroRio de Janeiro Brazil
| | - Valéria Barbosa Moreira
- Departamento de Medicina Clínica, Hospital Universitário Antônio PedroUniversidade Federal FluminenseNiterói Brazil
| | - Angela Santos Ferreira Nani
- Departamento de Medicina Clínica, Hospital Universitário Antônio PedroUniversidade Federal FluminenseNiterói Brazil
| | | |
Collapse
|
29
|
Pfau JC, McNew T, Hanley K, Swan L, Black B. Autoimmune markers for progression of Libby amphibole lamellar pleural thickening. Inhal Toxicol 2019; 31:409-419. [PMID: 31814459 DOI: 10.1080/08958378.2019.1699616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Exposure to Libby Asbestiform Amphibole (LAA) is associated with asbestos-related diseases, including mesothelioma, pulmonary carcinoma, pleural fibrosis, and systemic autoimmune diseases. The pleural fibrosis can manifest as a rapidly progressing lamellar pleural thickening (LPT), which causes thoracic pain, dyspnea, and worsening pulmonary function tests (PFT). It is refractory to treatment and frequently fatal.Objective: Because of the immune dysfunction that has been described in the LAA-exposed population and the association of pleural manifestations with the presence of autoantibodies, this study tested whether specific immunological factors were associated with progressive LPT and whether they could be used as markers of progressive disease.Methods: Subjects were placed into three study groups defined as (1) progressive LPT, (2) stable LPT, (3) no LPT. Serum samples were tested for antinuclear autoantibodies, mesothelial cell autoantibodies, anti-plasminogen antibodies, IL1 beta, and IL17; which have all been shown to be elevated in mice and/or humans exposed to LAA.Results: Group 1 had significantly higher mean values for all of the autoantibodies, but not IL1 or IL-17, compared to the control Group 3. All three autoantibody tests had high specificity but low sensitivity, but ROC area-under-the-curve values for all three antibodies were over 0.7, statistically higher than a test with no value. When all LPT subjects were combined (Progressive plus Stable), no marker had predictive value for disease.Conclusion: The data support the hypothesis that progressive LPT is associated with immunological findings that may serve as an initial screen for progressive LPT.
Collapse
Affiliation(s)
- Jean C Pfau
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Tracy McNew
- Center for Asbestos Related Diseases, Libby, MT, USA
| | | | - Lindsay Swan
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Brad Black
- Center for Asbestos Related Diseases, Libby, MT, USA
| |
Collapse
|
30
|
Zhang S, Zhang W, Zeng X, Zhao W, Wang Z, Dong X, Jia Y, Shen J, Chen R, Lin X. Inhibition of Rac1 activity alleviates PM2.5-induced pulmonary inflammation via the AKT signaling pathway. Toxicol Lett 2019; 310:61-69. [DOI: 10.1016/j.toxlet.2019.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/05/2019] [Accepted: 04/11/2019] [Indexed: 01/09/2023]
|
31
|
Dihydrotanshinone I Alleviates Crystalline Silica-Induced Pulmonary Inflammation by Regulation of the Th Immune Response and Inhibition of STAT1/STAT3. Mediators Inflamm 2019; 2019:3427053. [PMID: 31379467 PMCID: PMC6652093 DOI: 10.1155/2019/3427053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
Occupational exposure to crystalline silica (CS) results in a persistent pulmonary inflammatory response that eventually leads to abnormal tissue repair, disability, and death. The inflammatory-immune responses occur in the early stages of CS exposure, and both innate and adaptive immunity are involved. CD4+ T cells play a pivotal role in the pathogenesis of CS-induced pulmonary disease, which has no proven curative therapy. Dihydrotanshinone I (DHI), a natural product isolated from Salvia miltiorrhiza Bunge (Danshen), has anti-inflammatory and immunomodulatory properties. However, whether DHI has a protective effect on CS-induced lung disease, how it influences the Th immune response, and the potential underlying molecular mechanism(s) have not been fully clarified. In this study, DHI treatment of CS-exposed mice reduced the expression of proinflammatory cytokines and the infiltration of immune cells. It significantly ameliorated CS-induced pulmonary inflammation by attenuating T helper (Th)1 and Th17 responses, which were tightly related to the inhibition of STAT1 and STAT3. DHI significantly altered Th2 cytokines but not the Th2 nuclear transcription factor. Furthermore, our study found that DHI treatment also affected regulatory T cell activity in CS-injured mice. Taken together, our findings indicated that DHI could modulate Th responses and alleviate CS-induced pulmonary inflammation, suggesting a novel application of DHI in CS-induced pulmonary disease.
Collapse
|
32
|
Role of Nephronectin in Pathophysiology of Silicosis. Int J Mol Sci 2019; 20:ijms20102581. [PMID: 31130697 PMCID: PMC6566895 DOI: 10.3390/ijms20102581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/16/2019] [Accepted: 05/25/2019] [Indexed: 01/11/2023] Open
Abstract
Silicosis is a typical form of pneumoconiosis and is characterized as a type of lung fibrosis. Silica particles are captured and recognized upon by alveolar macrophages via the macrophage receptor with collagenous structure (MARCO) scavenger receptor, and thereafter the inflammasome is activated. Thereafter, various chemokines/cytokines play their roles to eventually form fibrosis. Additionally, silica particles chronically activate T helper cells which sets the background for the formation of silicosis-associated autoimmune disturbances. The occurrence and progression of lung fibrosis, the extracellular matrix-related molecules such as integrins and their ligands including fibronectin, vitronectin, laminin, and collagens, all play important roles. Here, the roles of these molecules in silicosis-related lung fibrosis are reviewed from the literature. Additionally, the measurement of serum nephronectin (Npnt), a new member of the integrin family of ligands, is discussed, together with investigations attempting to delineate the role of Npnt in silica-induced lung fibrosis. Serum Npnt was found to be higher in silicosis patients compared to healthy volunteers and seems to play a role in the progression of fibrosis with other cytokines. Therefore, serum Npnt levels may be employed as a suitable marker to monitor the progression of fibrosis in silicosis patients.
Collapse
|
33
|
Huaux F. Emerging Role of Immunosuppression in Diseases Induced by Micro- and Nano-Particles: Time to Revisit the Exclusive Inflammatory Scenario. Front Immunol 2018; 9:2364. [PMID: 30510551 PMCID: PMC6252316 DOI: 10.3389/fimmu.2018.02364] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022] Open
Abstract
Fibrosis, cancer, and autoimmunity developing upon particle exposure have been exclusively linked with uncontrolled inflammatory processes. The critical role of inflammation is now challenged by several contradictory observations indicating that the emergence of these chronic disorders may result from non-inflammatory events. A growing number of studies reveals that micro- and nano-particles can cause exaggerated and persistent immunosuppression characterized by the release of potent anti-inflammatory cytokines (IL-10 and TGF-β), and the recruitment of major regulatory immune cells (M2 macrophages, T and B regs, and MDSC). This persistent immunosuppressive environment is initially established to limit early inflammation but contributes later to fibrosis, cancer, and infection. Immunosuppression promotes fibroblast proliferation and matrix element synthesis and subverts innate and adaptive immune surveillance against tumor cells and microorganisms. This review details the contribution of immunosuppressive cells and their derived immunoregulatory mediators and delineates the mutual role of inflammatory vs. immunosuppressive mechanisms in the pathogenesis of chronic diseases induced by particles. The consideration of these new results explains how particle-related diseases can develop independently of chronic inflammation, enriches current bioassays predicting particle toxicity and suggests new clinical strategies for treating patients affected by particle-associated diseases.
Collapse
Affiliation(s)
- François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology, Institut de Recherche Experimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
34
|
Bronchoalveolar Lavage Fluid microRNA-146a: A Biomarker of Disease Severity and Pulmonary Function in Patients With Silicosis. J Occup Environ Med 2018; 58:e177-82. [PMID: 27158964 DOI: 10.1097/jom.0000000000000719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To examine the impact of microRNA-146a (miR-146a) on pulmonary function and disease severity in silicosis patients. METHODS Twenty-nine silicosis patients and six observation subjects were recruited. MiR-146a expression level was detected by qRT-PCR, and pulmonary function was assessed with a spirometer. RESULTS MiR-146a expression level was higher in silicosis patients than in observation subjects, and the probability of suffering from silicosis increased with increasing miR-146a level. MiR-146a was associated with the severity of silicosis. As the miR-146a increased, the probability of suffering from silicosis increased for stage I patients, and for stage II & III patients, the probability first increased and then decreased. MiR-146a was also associated with decreased pulmonary function measures, pulmonary function impairment, and restrictive ventilator dysfunction. CONCLUSIONS miR-146a was significantly associated with the disease severity and pulmonary function of silicosis.
Collapse
|
35
|
Gurczynski SJ, Moore BB. IL-17 in the lung: the good, the bad, and the ugly. Am J Physiol Lung Cell Mol Physiol 2017; 314:L6-L16. [PMID: 28860146 DOI: 10.1152/ajplung.00344.2017] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The IL-17 family of cytokines has emerged over the last two decades as a pleiotropic group of molecules that function in a wide variety of both beneficial and detrimental (pathological) processes, mainly in mucosal barrier tissue. The beneficial effects of IL-17 expression are especially important in the lung, where exposure to foreign agents is abundant. IL-17A plays an important role in protection from both extracellular bacteria and fungi, as well as viruses that infect cells of the mucosal tracts. IL-17 coregulated cytokines, such as IL-22, are involved in maintaining epithelial cell homeostasis and participate in epithelial cell repair/regeneration following inflammatory insults. Thus, the IL-17/IL-22 axis is important in both responding to, and recovering from, pathogens. However, aberrant expression or overexpression of IL-17 cytokines contributes to a number of pathological outcomes, including asthma, pneumonitis, and generation or exacerbation of pulmonary fibrosis. This review covers the good, bad, and ugly aspects of IL-17 in the lung.
Collapse
Affiliation(s)
- Stephen J Gurczynski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
36
|
IL-10-Producing B Cells Suppress Effector T Cells Activation and Promote Regulatory T Cells in Crystalline Silica-Induced Inflammatory Response In Vitro. Mediators Inflamm 2017; 2017:8415094. [PMID: 28831210 PMCID: PMC5558645 DOI: 10.1155/2017/8415094] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/11/2017] [Indexed: 12/11/2022] Open
Abstract
Long-term exposure to crystalline silica leads to silicosis, which is characterized by persistent lung inflammation and lung fibrosis. Multiple immune cells have been demonstrated to participate in crystalline silica-induced immune responses. Our previous study indicated that B10 could control lung inflammation through modulating the Th balance in experimental silicosis in mice. However, the regulatory mechanism of B10 on CD4+ T cells is still unclear. MACS-sorted CD19+ B cells from the three different groups were cultured with CD4+ T cells either with or without transwell insert plates to evaluate the effects of B10 on CD4+ T cells, including Teff and Treg. B10 was eliminated by anti-CD22 application in vivo. Flow cytometry was used to test the frequencies of CD4+ T cells, and the expressions of the related cytokines were detected by real-time PCR and CBA. Insufficient B10 elevated the levels of proinflammatory cytokines and promoted Th responses in a way independent upon cell-cell contact in the Teff and B cell coculture system. B10 could both increase Treg activity and enhance conversion of Teff into Treg. Our findings demonstrated that B10 could affect Th responses by the release of IL-10, enhancing Treg functions and converting Teff into Treg.
Collapse
|
37
|
Hassani E, Bagheri M, Rad IA, Mohebbi I. Association between SNPs at IL-17A and IL-17F and susceptibility to accelerated silicosis. Toxicol Ind Health 2017; 33:673-680. [DOI: 10.1177/0748233717695431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The association between single nucleotide polymorphisms (SNPs) in the interleukin (IL)-17 gene and silicosis has been evaluated in different populations. The aim of the present study was to analyze the association between SNPs at IL-17A (−832A/G) and IL-17F (+7488A/G) and susceptibility to accelerated silicosis in the Iranian Kurdish population. We studied 48 patients with accelerated silicosis and 62 controls. Genomic DNA was isolated using the “salting out” method. PCR-RFLP was performed for all SNPs typing. The frequencies of A/A, A/G, and G/G genotypes at IL-17A (−832A/G) were 4 (8.33%), 23 (47.92%), and 21 (43.75%) in patients and 5 (8.06%), 35 (56.45%), and 22 (35.48%) in controls, respectively. The frequencies of A and G alleles at IL-17 (−832A/G) were 31 (32.29%) and 65 (67.71%) in patients, and 45 (36.29%) and 79 (63.71%) in the controls, respectively. The frequencies of A/A, A/G, and G/G genotypes at IL-17F (+7488A/G) were 1 (2.08%), 47 (97.92%), and 0 (0%) in patients, and 11 (17.74%), 51 (82.26%), and 0 (0%) in the controls, respectively. The frequencies of A and G alleles at IL-17F (+7488A/G) were 49 (51.04%) and 47 (48.96%) in patients, and 73 (58.87%) and 51 (41.13%) in the controls, respectively. IL-17F (+7488A/G) genotype was more frequent among the cases compared with controls (97.92% vs. 82.26%). The frequency of the IL-17F (+7488A/G) genotype was significantly greater in patients with accelerated silicosis (odds ratio = 10.13 95%; confidence interval = 1.2–81.5; p = 0.008). The IL-17F (+7488A/G) genotype revealed a significantly increased risk of accelerated silicosis ( p < 0.05). The IL-17F (+7488 G) allele was associated with an increased risk of accelerated silicosis, but in the case of the IL-17A (−832A/G) polymorphism, a significant association was not observed.
Collapse
Affiliation(s)
- Ebrahim Hassani
- Department of Anesthesiology, Urmia University of Medical Sciences, Urmia, Iran
| | - Morteza Bagheri
- Cellular and Molecular Research Center, Department of Genetics, Urmia University of Medical Sciences, Urmia, Iran
| | - Isa Abdi Rad
- Cellular and Molecular Research Center, Department of Genetics, Urmia University of Medical Sciences, Urmia, Iran
| | - Iraj Mohebbi
- Social Determinants of Health Research Center, Department of Occupational Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
38
|
|
39
|
Zachariassen LF, Krych L, Engkilde K, Nielsen DS, Kot W, Hansen CHF, Hansen AK. Sensitivity to oxazolone induced dermatitis is transferable with gut microbiota in mice. Sci Rep 2017; 7:44385. [PMID: 28290517 PMCID: PMC5349591 DOI: 10.1038/srep44385] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/07/2017] [Indexed: 12/15/2022] Open
Abstract
Atopic Dermatitis (AD) has been associated with gut microbiota (GM) dysbiosis in humans, indicating a causative role of GM in AD etiology. Furthermore, the GM strongly correlates to essential disease parameters in the well-known oxazolone-induced mouse model of AD. Here, we demonstrate that it is possible to transfer both a high-responding and a low-responding AD phenotype with GM from conventional mice to germ-free mice. The mice inoculated with the high-responding GM had significantly higher clinical score, increased ear thickness, and increased levels of IL-1β, TNFα, IL-4, IL-5, and IL-6 compared to the mice inoculated with the low-responding GM. The inter-individual variation was in general not affected by this increase in effect size. Germ-free mice induced with AD revealed a high disease response as well as high inter-individual variation indicating protective properties of certain microbial taxa in this model. This study underlines that the GM has a strong impact on AD in mouse models, and that the power of studies may be increased by the application of mice inoculated with a specific GM from high responders to increase the effect size.
Collapse
Affiliation(s)
- Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Denmark
| | - Kåre Engkilde
- The Bartholin Institute, Rigshospitalet, Copenhagen, Denmark
| | | | - Witold Kot
- Department of Environmental Science, Faculty of Science and Technology, Aarhus University, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
40
|
Engelmann R, Müller-Hilke B. Experimental silicosis does not aggravate collagen-induced arthritis in mice. J Negat Results Biomed 2017; 16:5. [PMID: 28285600 PMCID: PMC5346855 DOI: 10.1186/s12952-017-0071-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 01/22/2023] Open
Abstract
Objectives To investigate the effect of chronic lung inflammation on the incidence and severity of collagen-induced arthritis in mice. Methods Chronic lung inflammation in the form of silicosis was induced via intranasal application of silica particles. Immunization with collagen Type II commenced one week later and mice were sacrificed six weeks after booster immunization. Thereafter, silicosis was confirmed via flow cytometry and arthritis was evaluated performing knee and paw histology. Results Pronounced lung inflammation in the silica-treated compared to PBS-treated control mice was demonstrated by significantly elevated broncho-alveolar lavage (BAL) cell count, attributable to increased numbers of macrophages and granulocytes. Inflammation in the lungs was not associated with elevated PAD2 and PAD4 expression, yet silica treated animals had significantly higher aCCP serum titers. However, lung inflammation did not lead to an increase in the incidence of arthritis, nor did it exacerbate the macroscopic or histologic joint scores. Conclusions Chronic lung inflammation resulting from silicosis does not aggravate collagen-induced arthritis in mice. Electronic supplementary material The online version of this article (doi:10.1186/s12952-017-0071-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robby Engelmann
- Institute of Immunology & Core Facility for Cell Sorting & Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany.
| | - Brigitte Müller-Hilke
- Institute of Immunology & Core Facility for Cell Sorting & Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| |
Collapse
|
41
|
Wang H, Song L, Ju W, Wang X, Dong L, Zhang Y, Ya P, Yang C, Li F. The acute airway inflammation induced by PM 2.5 exposure and the treatment of essential oils in Balb/c mice. Sci Rep 2017; 7:44256. [PMID: 28276511 PMCID: PMC5343586 DOI: 10.1038/srep44256] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/03/2017] [Indexed: 01/17/2023] Open
Abstract
PM2.5 is the main particulate air pollutant whose aerodynamic diameter is less than 2.5 micron. The inflammation of various respiratory diseases are associated with PM2.5 inhalation. Pro-inflammatory cytokine IL-1β generated from effected cells usually plays a crucial role in many kinds of lung inflammatory reactions. The exacerbation of Th immune responses are identified in some PM2.5 related diseases. To elucidate the underlying mechanism of PM2.5-induced acute lung inflammation, we exposed Balb/c mice to PM2.5 intratracheally and established a mice model. Acute lung inflammation and increased IL-1β expression was observed after PM2.5 instillation. Regulatory factors of IL-1β (TLR4/MyD88 signaling pathway and NLRP3 inflammasome) participated in this lung inflammatory response as well. Treatment with compound essential oils (CEOs) substantially attenuated PM2.5-induced acute lung inflammation. The decreased IL-1β and Th immune responses after CEOs treatment were significant. PM2.5 may increase the secretion of IL-1β through TLR4/MyD88 and NLRP3 pathway resulting in murine airway inflammation. CEOs could attenuate the lung inflammation by reducing IL-1β and Th immune responses in this model. This study describes a potentially important mechanism of PM2.5-induced acute lung inflammation and that may bring about novel therapies for the inflammatory diseases associated with PM2.5 inhalation.
Collapse
Affiliation(s)
- Hetong Wang
- Dept of Chemistry, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| | - Laiyu Song
- Dept of Immunological and Microbiological Laboratory, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| | - Wenhui Ju
- Atmospheric Environment Research Institute, China Research Academy of Environmental Sciences, Beijing 100012, People’s Republic of China
| | - Xuguang Wang
- Environmental Monitoring Station of Langfan, Langfang Environmental Protection Bureau, Langfang 065000, Hebei Province, People’s Republic of China
| | - Lu Dong
- Dept of Chemistry, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| | - Yining Zhang
- Dept of Chemistry, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| | - Ping Ya
- Dept of Chemistry, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| | - Chun Yang
- Dept of Nuclear Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, LiaoNing Province, People’s Republic of China
| | - Fasheng Li
- Dept of Chemistry, Dalian Medical University, Dalian 116044, Liaoning Province, People’s Republic of China
| |
Collapse
|
42
|
Chen Y, Li C, Lu Y, Zhuang H, Gu W, Liu B, Liu F, Sun J, Yan B, Weng D, Chen J. IL-10-Producing CD1d hiCD5 + Regulatory B Cells May Play a Critical Role in Modulating Immune Homeostasis in Silicosis Patients. Front Immunol 2017; 8:110. [PMID: 28243231 PMCID: PMC5303715 DOI: 10.3389/fimmu.2017.00110] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/24/2017] [Indexed: 12/28/2022] Open
Abstract
Silicosis is characterized by chronic lung inflammation and fibrosis, which are extremely harmful to human health. The pathogenesis of silicosis involves uncontrolled immune processes. Evidence supports that regulatory B cells (Bregs) produce negative regulatory cytokines, such as IL-10, which can negatively regulate immune responses in inflammation and autoimmune diseases. Our previous study found that IL-10-producing B cells were involved in the development of silica-induced lung inflammation and fibrosis of mice. However, little is known about the role of Bregs in silicosis patients (SP). In this study, we found that serum concentrations of IL-10 were significantly increased in SP by using protein array screening. We further determined that the frequency of IL-10-producing CD1dhiCD5+ Bregs, not IL-10-producing non-B lymphocytes, was significantly higher in SP compared to subjects under surveillance (SS) and healthy workers (HW) by flow cytometry. We also found that regulatory T cells (Tregs) and Th2 cytokines (IL-4, IL-5, and IL-13) were significantly increased in SP. Th1 cytokines (IFN-γ, IL-2, and IL-12) and inflammatory cytokines (IL-1β, IL-6, and TNF-α) were not significantly different between SP, SS, and HW. Our study indicated that IL-10-producing CD1dhiCD5+ Bregs might maintain Tregs and regulate Th1/Th2 polarization in SP, suggesting that IL-10-producing Bregs may play a critical role in modulating immune homeostasis in SP.
Collapse
Affiliation(s)
- Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Yiping Lu
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Huiying Zhuang
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Weijia Gu
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Bo Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| | - Jinkai Sun
- Department of Respiratory Medicine, Shenyang No. 9 Hospital , Shenyang , China
| | - Bo Yan
- Department of Respiratory Medicine, Shenyang No. 9 Hospital , Shenyang , China
| | - Dong Weng
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, China; Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University , Shenyang , China
| |
Collapse
|
43
|
Abdelaziz RR, Elkashef WF, Said E. Tadalafil reduces airway hyperactivity and protects against lung and respiratory airways dysfunction in a rat model of silicosis. Int Immunopharmacol 2016; 40:530-541. [DOI: 10.1016/j.intimp.2016.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/17/2016] [Accepted: 10/11/2016] [Indexed: 11/30/2022]
|
44
|
Marshall EA, Ng KW, Kung SHY, Conway EM, Martinez VD, Halvorsen EC, Rowbotham DA, Vucic EA, Plumb AW, Becker-Santos DD, Enfield KSS, Kennett JY, Bennewith KL, Lockwood WW, Lam S, English JC, Abraham N, Lam WL. Emerging roles of T helper 17 and regulatory T cells in lung cancer progression and metastasis. Mol Cancer 2016; 15:67. [PMID: 27784305 PMCID: PMC5082389 DOI: 10.1186/s12943-016-0551-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths worldwide. Lung cancer risk factors, including smoking and exposure to environmental carcinogens, have been linked to chronic inflammation. An integral feature of inflammation is the activation, expansion and infiltration of diverse immune cell types, including CD4+ T cells. Within this T cell subset are immunosuppressive regulatory T (Treg) cells and pro-inflammatory T helper 17 (Th17) cells that act in a fine balance to regulate appropriate adaptive immune responses.In the context of lung cancer, evidence suggests that Tregs promote metastasis and metastatic tumor foci development. Additionally, Th17 cells have been shown to be an integral component of the inflammatory milieu in the tumor microenvironment, and potentially involved in promoting distinct lung tumor phenotypes. Studies have shown that the composition of Tregs and Th17 cells are altered in the tumor microenvironment, and that these two CD4+ T cell subsets play active roles in promoting lung cancer progression and metastasis.We review current knowledge on the influence of Treg and Th17 cells on lung cancer tumorigenesis, progression, metastasis and prognosis. Furthermore, we discuss the potential biological and clinical implications of the balance among Treg/Th17 cells in the context of the lung tumor microenvironment and highlight the potential prognostic function and relationship to metastasis in lung cancer.
Collapse
Affiliation(s)
- Erin A Marshall
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Kevin W Ng
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Sonia H Y Kung
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada. .,British Columbia Cancer Research Centre Centre, Vancouver, Canada.
| | - Emma M Conway
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Victor D Martinez
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Elizabeth C Halvorsen
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - David A Rowbotham
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Emily A Vucic
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Adam W Plumb
- Departments of Microbiology and Immunology, University of British Columbia, Vancouver, Canada.,Department of Zoology, University of British Columbia, Vancouver, Canada
| | | | - Katey S S Enfield
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Jennifer Y Kennett
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Kevin L Bennewith
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - William W Lockwood
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - John C English
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ninan Abraham
- Departments of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Wan L Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada. .,British Columbia Cancer Research Centre Centre, Vancouver, Canada.
| |
Collapse
|
45
|
Li C, Du S, Lu Y, Lu X, Liu F, Chen Y, Weng D, Chen J. Blocking the 4-1BB Pathway Ameliorates Crystalline Silica-induced Lung Inflammation and Fibrosis in Mice. Am J Cancer Res 2016; 6:2052-2067. [PMID: 27698940 PMCID: PMC5039680 DOI: 10.7150/thno.16180] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/30/2016] [Indexed: 12/30/2022] Open
Abstract
Long term pulmonary exposure to crystalline silica leads to silicosis that manifests progressive interstitial fibrosis, eventually leading to respiratory failure and death. Despite efforts to eliminate silicosis, clinical cases continue to occur in both developing and developed countries. The exact mechanisms of crystalline silica-induced pulmonary fibrosis remain elusive. Herein, we find that 4-1BB is induced in response to crystalline silica injury in lungs and that it is highly expressed during development of experimental silicosis. Therefore, we explore the role of 4-1BB pathway during crystalline silica-induced lung injury and find that a specific inhibitor blocking the pathway could effectively alleviate crystalline silica-induced lung inflammation and subsequent pulmonary fibrosis in vivo. Compared to controls, the treated mice exhibited reduced Th1 and Th17 responses. The concentrations of pro-inflammatory cytokines in bronchoalveolar lavage fluid (BALF), including tumor necrosis factor (TNF)-α, interferon (IFN)-γ and interleukin (IL)-17A following crystalline silica challenge were also reduced in inhibitor-treated mice. Although there was no significant alteration in Th2 cytokines of IL-4 and IL-13, another type of pro-fibrogenic cell, regulatory T cell (Treg) was significantly affected. In addition, one of the major participants in fibrogenesis, fibrocyte recruited less due to the blockade. Furthermore, we demonstrated the decreased fibrocyte recruitment was associated with chemokine reductions in lung. Our study discovers the 4-1BB pathway signaling enhances inflammatory response and promotes pulmonary fibrosis induced by crystalline silica. The findings here provide novel insights into the molecular events that control crystalline silica-induced lung inflammation and fibrosis through regulating Th responses and the recruitment of fibrocytes in crystalline silica-exposed lung.
Collapse
|
46
|
Liu F, Dai W, Li C, Lu X, Chen Y, Weng D, Chen J. Role of IL-10-producing regulatory B cells in modulating T-helper cell immune responses during silica-induced lung inflammation and fibrosis. Sci Rep 2016; 6:28911. [PMID: 27354007 PMCID: PMC4926212 DOI: 10.1038/srep28911] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/13/2016] [Indexed: 12/18/2022] Open
Abstract
Silicosis is characterized by chronic lung inflammation and fibrosis, which are seriously harmful to human health. Previous research demonstrated that uncontrolled T-helper (Th) cell immune responses were involved in the pathogenesis of silicosis. Lymphocytes also are reported to have important roles. Existing studies on lymphocyte regulation of Th immune responses were limited to T cells, such as the regulatory T (Treg) cell, which could negatively regulate inflammation and promote the process of silicosis. However, other regulatory subsets in silicosis have not been investigated in detail, and the mechanism of immune homeostasis modulation needs further exploration. Another regulatory lymphocyte, the regulatory B cell, has recently drawn increasing attention. In this study, we comprehensively showed the role of IL-10-producing regulatory B cell (B10) in a silicosis model of mice. B10 was inducible by silica instillation. Insufficient B10 amplified inflammation and attenuated lung fibrosis by promoting the Th1 immune response. Insufficient B10 clearly inhibited Treg and decreased the level of IL-10. Our study indicated that B10 could control lung inflammation and exacerbate lung fibrosis by inhibiting Th1 response and modulating the Th balance. The regulatory function of B10 could be associated with Treg induction and IL-10 secretion.
Collapse
Affiliation(s)
- Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Wujing Dai
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Xiaowei Lu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Dong Weng
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P. R. China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| |
Collapse
|
47
|
Liu SJ, Wang P, Jiao J, Han L, Lu YM. Differential gene expression associated with inflammation in peripheral blood cells of patients with pneumoconiosis. J Occup Health 2016; 58:373-80. [PMID: 27265534 PMCID: PMC5356945 DOI: 10.1539/joh.16-0021-oa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objectives: To study expression changes in inflammation-related genes in peripheral blood of patients with pneumoconiosis and to explore the possibility of these genes as pneumoconiosis biomarkers. Methods: Peripheral blood samples of patients with pneumoconiosis patients and controls were collected, and total RNA of the blood cells were extracted and reverse transcribed to cDNA. Screenings of deferentially expressed genes associated with inflammation between patients with pneumoconiosis and controls were performed using real-time quantitative PCR array and the expressions of the three most upregulated genes were confirmed by real-time PCR. Results: The expression of 11 genes was significantly altered in patients with pneumoconiosis compared with those of the control. Among these 11 genes, 8 genes were upregulated and 3 were downregulated. Preliminary results indicated that interleukin 6 (IL-6) mRNA expression in patients with pneumoconiosis was higher than that in controls (P=0.019). The level of IL6 mRNA expression in the patients was higher than that in non-smoking controls, but it was neither affected by type and stage of pneumoconiosis nor by time of contact with dust. Conclusions: IL6 was possibly involved in the development of pneumoconiosis.
Collapse
Affiliation(s)
- Shu-Jun Liu
- The Second Clinical Medical College, Nanchang University
| | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- Bernardo S. Franklin
- Institute of Innate Immunity, University Hospitals, University of Bonn, Bonn 53127, Germany; , ,
| | - Matthew S. Mangan
- Institute of Innate Immunity, University Hospitals, University of Bonn, Bonn 53127, Germany; , ,
- German Center for Neurodegenerative Diseases, Bonn 53175, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals, University of Bonn, Bonn 53127, Germany; , ,
- German Center for Neurodegenerative Diseases, Bonn 53175, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| |
Collapse
|
49
|
Blockade of Wnt/β-Catenin Pathway Aggravated Silica-Induced Lung Inflammation through Tregs Regulation on Th Immune Responses. Mediators Inflamm 2016; 2016:6235614. [PMID: 27069316 PMCID: PMC4812397 DOI: 10.1155/2016/6235614] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 12/31/2022] Open
Abstract
CD4+ T cells play an important role in regulating silica-induced inflammation and fibrosis. Recent studies showed that Wnt/β-catenin pathway could modulate the function and the differentiation of CD4+ T cells. Therefore, Wnt/β-catenin pathway may participate in the development and progress of silicosis. To investigate the role of Wnt/β-catenin pathway, we used lentivirus expressing β-catenin shRNA to block the Wnt/β-catenin pathway by intratracheal instillation to the mice model of silicosis. Treatment of lentivirus could significantly aggravate the silica-induced lung inflammation and attenuated the fibrosis at the late stage. By analyzing CD4+ T cells, we found that blockade of Wnt/β-catenin pathway suppressed regulatory T cells (Tregs). Reciprocally, enhanced Th17 response was responsible for the further accumulation of neutrophils and production of proinflammatory cytokines. In addition, blockade of Wnt/β-catenin pathway delayed the Th1/Th2 polarization by inhibiting Tregs and Th2 response. These results indicated that Wnt/β-catenin pathway could regulate Tregs to modulate Th immune response, which finally altered the pathological character of silicosis. Our study suggested that Wnt/β-catenin pathway might be a potential target to treat the silica-induced inflammation and fibrosis.
Collapse
|
50
|
Liu T, Dai W, Li C, Liu F, Chen Y, Weng D, Chen J. Baicalin Alleviates Silica-Induced Lung Inflammation and Fibrosis by Inhibiting the Th17 Response in C57BL/6 Mice. JOURNAL OF NATURAL PRODUCTS 2015; 78:3049-3057. [PMID: 26605988 DOI: 10.1021/acs.jnatprod.5b00868] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Silicosis is an inflammatory and fibrotic lung disease caused by inhalation of silica. Th17 cells play a key role in causing silica-induced lung inflammation and fibrosis. Baicalin, a compound isolated from the Chinese herb Huangqin, could suppress the differentiation of Th17 cells and alleviate inflammation. However, there are very few reports of the immunoregulatory mechanisms of baicalin in experimental silica-induced lung inflammation and fibrosis. In our study, mice were exposed to silica by intratracheal instillation, and in this way we established an experimental silicosis model. To elucidate the effects and mechanisms of baicalin in silica-induced inflammation and fibrosis, we used baicalin to treat the developed mouse model of silicosis. Treatment with baicalin attenuated the accumulation of inflammatory cells and led to milder pathological inflammatory and fibrotic changes in lung tissues. Baicalin affected the immunological balance between Th17 and Treg responses. Therefore, baicalin caused a decrease in Th17 cells by stimulating Treg cells and by inhibiting IL-6 and IL-23. We further demonstrated that silica-induced Th1 and Th2 immune responses were both inhibited by increased Treg activation, which was promoted by baicalin. Our findings confirmed the potential functions of baicalin in inhibiting the Th17 response and reducing silica-induced inflammation and fibrosis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Wujing Dai
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Chao Li
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Fangwei Liu
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Ying Chen
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Dong Weng
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| |
Collapse
|