1
|
He X, Yao D, Yuan X, Ban J, Gou Y, You M. Occupational agents-mediated asthma: From the perspective of autophagy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175880. [PMID: 39216756 DOI: 10.1016/j.scitotenv.2024.175880] [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: 07/05/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Occupational asthma (OA) is a common occupational pulmonary disease that is frequently underdiagnosed and underreported. The complexity of diagnosing and treating OA creates a significant social and economic burden, making it an important public health issue. In addition to avoiding allergens, patients with OA require pharmacotherapy; however, new therapeutic targets and strategies need further investigation. Autophagy may be a promising intervention target, but there is a lack of relevant studies summarizing the role of autophagy in OA. In this review consolidates the current understanding of OA, detailing principal and novel agents responsible for its onset. Additionally, we summarize the mechanisms of autophagy in HMW and LMW agents induced OA, revealing that occupational allergens can induce autophagy disorders in lung epithelial cells, smooth muscle cells, and dendritic cells, ultimately leading to OA through involving inflammatory responses, oxidative stress, and cell death. Finally, we discuss the prospects of targeting autophagy as an effective strategy for managing OA and even steroid-resistant asthma, encompassing autophagy interventions focused on organoids, organ-on-a-chip systems, nanomaterials vehicle, and nanobubbles; developing combined exposure models, and the role of non-classical autophagy in occupational asthma. In briefly, this review summarizes the role of autophagy in occupational asthma, offers a theoretical foundation for OA interventions based on autophagy, and identifies directions and challenges for future research.
Collapse
Affiliation(s)
- Xiu He
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Dengxiang Yao
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Xiaoli Yuan
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Jiaqi Ban
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Yuxuan Gou
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Clinical Medical School, Guizhou Medical University, Guiyang 561113, China
| | - Mingdan You
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
2
|
Wu J, Wang P, Xie X, Yang X, Tang S, Zhao J, Liu T, Wang J, Zhang J, Xia T, Feng X. Gasdermin D silencing alleviates airway inflammation and remodeling in an ovalbumin-induced asthmatic mouse model. Cell Death Dis 2024; 15:400. [PMID: 38849380 PMCID: PMC11161474 DOI: 10.1038/s41419-024-06777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024]
Abstract
Emerging evidence demonstrates that pyroptosis has been implicated in the pathogenesis of asthma. Gasdermin D (GSDMD) is the pyroptosis executioner. The mechanism of GSDMD in asthma remains unclear. The aim of this study was to elucidate the potential role of GSDMD in asthmatic airway inflammation and remodeling. Immunofluorescence staining was conducted on airway epithelial tissues obtained from both asthma patients and healthy controls (HCs) to evaluate the expression level of N-GSDMD. ELISA was used to measure concentrations of cytokines (IL-1β, IL-18, IL-17A, and IL-10) in serum samples collected from asthma patients and healthy individuals. We demonstrated that N-GSDMD, IL-18, and IL-1β were significantly increased in samples with mild asthma compared with those from the controls. Then, wild type and Gsdmd-knockout (Gsdmd-/-) mice were used to establish asthma model. We performed histopathological staining, ELISA, and flow cytometry to explore the function of GSDMD in allergic airway inflammation and tissue remodeling in vivo. We observed that the expression of N-GSDMD, IL-18, and IL-1β was enhanced in OVA-induced asthma mouse model. Gsdmd knockout resulted in attenuated IL-18, and IL-1β production in both bronchoalveolar lavage fluid (BALF) and lung tissue in asthmatic mice. In addition, Gsdmd-/- mice exhibit a significant reduction in airway inflammation and remodeling, which might be associated with reduced Th17 inflammatory response and M2 polarization of macrophages. Further, we found that GSDMD knockout may improve asthmatic airway inflammation and remodeling through regulating macrophage adhesion, migration, and macrophage M2 polarization by targeting Notch signaling pathway. These findings demonstrate that GSDMD deficiency profoundly alleviates allergic inflammation and tissue remodeling. Therefore, GSDMD may serve as a potential therapeutic target against asthma.
Collapse
Affiliation(s)
- Jinxiang Wu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Pin Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Xinyu Xie
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Xiaoqi Yang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Shuangmei Tang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Jiping Zhao
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Tian Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Junfei Wang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tongliang Xia
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China
| | - Xin Feng
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, China.
| |
Collapse
|
3
|
Zhang Y, Tian T, Liang C, Wang J, Zhang J, Tian S, Xie R, Yang T, Han B. Lysine specific demethylase 1 inhibits sodium arsenite activation of HSCs by regulating SESN2/AMPK/ULK1 signaling pathway activity. ENVIRONMENTAL TOXICOLOGY 2024; 39:3563-3577. [PMID: 38477077 DOI: 10.1002/tox.24184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/10/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024]
Abstract
Lysine specific demethylase 1 (LSD1) is a histone demethylase that specifically catalyzes the demethylation of histone H3K4 (H3K4me1/2) and regulates gene expression. In addition, it can mediate the process of autophagy through its demethylase activity. Sestrin2 (SESN2) is a stress-induced protein and a positive regulator of autophagy. In NaAsO2-induced mouse fibrotic livers and activated hepatic stellate cells (HSCs), LSD1 expression is decreased, SESN2 expression is increased, and autophagy levels are also increased. Overexpression of LSD1 and silencing of SESN2 decreased the level of autophagy and attenuated the activation of HSCs induced by NaAsO2. LSD1 promoted SESN2 gene transcription by increasing H3K4me1/2 in the SESN2 promoter region. 3-methyladenine (3-MA) and chloroquine were used to inhibit autophagy of HSCs, and the degree of activation was also alleviated. Taken together, LSD1 positively regulates SESN2 by increasing H3K4me1/2 enrichment in the SESN2 promoter region, which in turn increases the level of autophagy and promotes the activation of HSCs. Our results may provide new evidence for the importance of LSD1 in the process of autophagy and activation of HSCs induced by arsenic poisoning. Increasing the expression and activity of LSD1 is expected to be an effective way to reverse the autophagy and activation of HSCs induced by arsenic poisoning.
Collapse
Affiliation(s)
- Yingwan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tian Tian
- Department of Eugenic Genetics, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, China
| | - Cai Liang
- Southwest Hospital, Army Medical University, Chongqing, China
| | - Junli Wang
- The Second People's Hospital of Guiyang, Guizhou, China
| | - Jiayuan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shanshan Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
4
|
Baglivo I, Quaranta VN, Dragonieri S, Colantuono S, Menzella F, Selvaggio D, Carpagnano GE, Caruso C. The New Paradigm: The Role of Proteins and Triggers in the Evolution of Allergic Asthma. Int J Mol Sci 2024; 25:5747. [PMID: 38891935 PMCID: PMC11171572 DOI: 10.3390/ijms25115747] [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: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Epithelial barrier damage plays a central role in the development and maintenance of allergic inflammation. Rises in the epithelial barrier permeability of airways alter tissue homeostasis and allow the penetration of allergens and other external agents. Different factors contribute to barrier impairment, such as eosinophilic infiltration and allergen protease action-eosinophilic cationic proteins' effects and allergens' proteolytic activity both contribute significantly to epithelial damage. In the airways, allergen proteases degrade the epithelial junctional proteins, allowing allergen penetration and its uptake by dendritic cells. This increase in allergen-immune system interaction induces the release of alarmins and the activation of type 2 inflammatory pathways, causing or worsening the main symptoms at the skin, bowel, and respiratory levels. We aim to highlight the molecular mechanisms underlying allergenic protease-induced epithelial barrier damage and the role of immune response in allergic asthma onset, maintenance, and progression. Moreover, we will explore potential clinical and radiological biomarkers of airway remodeling in allergic asthma patients.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Silvano Dragonieri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital-AULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - David Selvaggio
- UOS di Malattie dell’Apparato Respiratorio Ospedale Cristo Re, 00167 Roma, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
5
|
Liu P, Wang Y, Chen C, Liu H, Ye J, Zhang X, Ma C, Zhao D. Research trends on airway remodeling: A bibliometrics analysis. Heliyon 2024; 10:e24824. [PMID: 38333835 PMCID: PMC10850909 DOI: 10.1016/j.heliyon.2024.e24824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Background Airway remodeling is an essential pathological basis of respiratory diseases such as asthma and COPD, which is significantly related to pulmonary function and clinical symptoms. And pulmonary disease can be improved by regulating airway remodeling. This study aimed to establish a knowledge map of airway remodeling to clarify current research hotspots and future research trends. Methods A comprehensive search was performed to analyze all relevant articles on airway remodeling using the Web of Science Core Collection Database from January 01, 2004 to June 03, 2023.2 reviewers screened the retrieved literature. Besides, the CiteSpace (6.2. R3) and VOSviewer (1.6.19) were utilized to visualize the research focus and trend regarding the effect of airway remodeling. Results A total of 4077 articles about airway remodeling were retrieved. The United States is the country with the most published literature, underscoring the country's role in airway remodeling. In recent years, China has been the country with the fastest growth in the number of published literature, suggesting that China will play a more critical role in airway remodeling in the future. From the perspective of co-operation among countries, European co-operation was closer than Asian co-operation. The co-citation analysis showed that 98,313 citations were recorded in 3594 articles, and 25 clusters could be realized. In recent years, Burst detection shows that oxidative stress and epithelial-mesenchymal transition are hot words. Conclusions Based on the bibliometric analysis of airway remodeling studies in the past 20 years, a multi-level knowledge structure map was drawn, it mainly includes countries, institutions, research fields, authors, journals, keywords and so on. The research directions represented by obstructive airway disease, PDGF-BB treatment of airway smooth muscle, allergen-induced airway remodeling, extracellular matrix, and non-coding RNA are the research hotspots in the field of airway remodeling. While the risk factors for airway remodeling, the application of new noninvasively assessing tools, biomarkers as well as The molecular mechanism represented by EMT and autophagy had been frontiers in recent years.
Collapse
Affiliation(s)
- Pengcheng Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Chen Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Hui Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Jing Ye
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Xiaoming Zhang
- School of Basic Medicine, Anhui Medical University, Hefei, 230000, China
| | - Changxiu Ma
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Dahai Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| |
Collapse
|
6
|
Pan Y, Zhang D, Zhang J, Liu X, Xu J, Zeng R, Cui W, Liu T, Wang J, Dong L. Suppression of SPARC Ameliorates Ovalbumin-induced Airway Remodeling via TGFβ1/Smad2 in Chronic Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:91-108. [PMID: 38262393 PMCID: PMC10823139 DOI: 10.4168/aair.2024.16.1.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/10/2023] [Accepted: 07/07/2023] [Indexed: 01/25/2024]
Abstract
PURPOSE Airway remodeling is a critical feature of asthma. Secreted protein acidic and rich in cysteine (SPARC), which plays a cardinal role in regulating cell-matrix interactions, has been implicated in various fibrotic diseases. However, the effect of SPARC in asthma remains unknown. METHODS We studied the expression of SPARC in human bronchial epithelial cells and serum of asthmatics as well as in the lung tissues of chronic asthma mice. The role of SPARC was examined by using a Lentivirus-mediated SPARC knockdown method in the ovalbumin (OVA)-induced asthma mice. The biological processes regulated by SPARC were identified using RNA sequencing. The function of SPARC in the remodeling process induced by transforming growth factor β1 (TGFβ1) was conducted by using SPARC small interfering RNA (siRNA) or recombinant human SPARC protein in 16HBE cells. RESULTS We observed that SPARC was up-regulated in human bronchial epithelia of asthmatics and the asthmatic mice. The levels of serum SPARC in asthmatics were also elevated and negatively correlated with the forced expiratory volume in one second (FEV1) to forced vital capacity ratio (FVC) (r = -0.485, P < 0.01) and FEV1 (%predicted) (r = -0.425, P = 0.001). In the chronic asthmatic mice, Lentivirus-mediated SPARC knockdown significantly decreased airway remodeling and airway hyper-responsiveness. According to gene set enrichment analysis, negatively enriched pathways found in the OVA + short hairpin-SPARC group included ECM organization and collagen formation. In the lung function studies, knockdown of SPARC by siRNA reduced the expression of remodeling-associated biomarkers, cell migration, and contraction by blocking the TGFβ1/Smad2 pathway. Addition of human recombinant SPARC protein promoted the TGFβ1-induced remodeling process, cell migration, and contraction in 16HBE cells via the TGFβ1/Smad2 pathway. CONCLUSIONS Our studies provided evidence for the involvement of SPARC in the airway remodeling of asthma via the TGFβ1/Smad2 pathway.
Collapse
Affiliation(s)
- Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xiaofei Liu
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jiawei Xu
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Rong Zeng
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Wenjing Cui
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Tian Liu
- Department of Pulmonary and Critical Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Junfei Wang
- Department of Pulmonary and Critical Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China.
| |
Collapse
|
7
|
Hou J, Yang Y, Han X. Machine Learning and Single-Cell Analysis Identify Molecular Features of IPF-Associated Fibroblast Subtypes and Their Implications on IPF Prognosis. Int J Mol Sci 2023; 25:94. [PMID: 38203265 PMCID: PMC10778894 DOI: 10.3390/ijms25010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease of unknown cause, and the involvement of fibroblasts in its pathogenesis is well recognized. However, a comprehensive understanding of fibroblasts' heterogeneity, their molecular characteristics, and their clinical relevance in IPF is lacking. In this study, we aimed to systematically classify fibroblast populations, uncover the molecular and biological features of fibroblast subtypes in fibrotic lung tissue, and establish an IPF-associated, fibroblast-related predictive model for IPF. Herein, a meticulous analysis of scRNA-seq data obtained from lung tissues of both normal and IPF patients was conducted to identify fibroblast subpopulations in fibrotic lung tissues. In addition, hdWGCNA was utilized to identify co-expressed gene modules associated with IPF-related fibroblasts. Furthermore, we explored the prognostic utility of signature genes for these IPF-related fibroblast subtypes using a machine learning-based approach. Two predominant fibroblast subpopulations, termed IPF-related fibroblasts, were identified in fibrotic lung tissues. Additionally, we identified co-expressed gene modules that are closely associated with IPF-fibroblasts by utilizing hdWGCNA. We identified gene signatures that hold promise as prognostic markers in IPF. Moreover, we constructed a predictive model specifically focused on IPF-fibroblasts which can be utilized to assess disease prognosis in IPF patients. These findings have the potential to improve disease prediction and facilitate targeted interventions for patients with IPF.
Collapse
Affiliation(s)
- Jiwei Hou
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Canter of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | | | - Xin Han
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Canter of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| |
Collapse
|
8
|
Xu J, Yu Z, Liu X. Angiotensin-(1-7) suppresses airway inflammation and airway remodeling via inhibiting ATG5 in allergic asthma. BMC Pulm Med 2023; 23:422. [PMID: 37919667 PMCID: PMC10623740 DOI: 10.1186/s12890-023-02719-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Angiotensin (Ang)-(1-7) can reduce airway inflammation and airway remodeling in allergic asthma. Autophagy-related 5 (ATG5) has attracted wide attentions in asthma. However, the effects of Ang-(1-7) on ATG5-mediated autophagy in allergic asthma are unclear. METHODS In this study, human bronchial epithelial cell (BEAS-2B) and human bronchial smooth muscle cell (HBSMC) were treated with different dose of Ang-(1-7) to observe changes of cell viability. Changes of ATG5 protein expression were measured in 10 ng/mL of interleukin (IL)-13-treated cells. Transfection of ATG5 small interference RNA (siRNA) or ATG5 cDNA in cells was used to analyze the effects of ATG5 on secretion of cytokines in the IL-13-treated cells. The effects of Ang-(1-7) were compared to the effects of ATG5 siRNA transfection or ATG5 cDNA transfection in the IL-13-treated cells. In wild-type (WT) mice and ATG5 knockout (ATG5-/-) mice, ovalbumin (OVA)-induced airway inflammation, fibrosis and autophagy were observed. In the OVA-induced WT mice, Ang-(1-7) treatment was performed to observe its effects on airway inflammation, fibrosis and autophagy. RESULTS The results showed that ATG5 protein level was decreased with Ang-(1-7) dose administration in the IL-13-treated BEAS-2B and IL13-treated HBSMC. Ang-(1-7) played similar results to ATG5 siRNA that it suppressed the secretion of IL-25 and IL-13 in the IL-13-treated BEAS-2B cells, and inhibited the expression of transforming growth factor (TGF)-β1 and α-smooth muscle actin (α-SMA) protein in the IL-13-treated HBSMC cells. ATG5 cDNA treatment significantly increased the secretion of IL-25 and IL-13 and expression of TGF-β1 and α-SMA protein in IL-13-treated cells. Ang-(1-7) treatment suppressed the effects of ATG5 cDNA in the IL-13-treated cells. In OVA-induced WT mice, Ang-(1-7) treatment suppressed airway inflammation, remodeling and autophagy. ATG5 knockout also suppressed the airway inflammation, remodeling and autophagy. CONCLUSIONS Ang-(1-7) treatment suppressed airway inflammation and remodeling in allergic asthma through inhibiting ATG5, providing an underlying mechanism of Ang-(1-7) for allergic asthma treatment.
Collapse
Affiliation(s)
- Jianfeng Xu
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264001, China
| | - Zhenyu Yu
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Yantai, 246001, China
| | - Xueping Liu
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264001, China.
| |
Collapse
|
9
|
Peng H, Sun F, Jiang Y, Guo Z, Liu X, Zuo A, Lu D. Semaphorin 7a aggravates TGF-β1-induced airway EMT through the FAK/ERK1/2 signaling pathway in asthma. Front Immunol 2023; 14:1167605. [PMID: 38022556 PMCID: PMC10646317 DOI: 10.3389/fimmu.2023.1167605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Background TGF-β1 can induce epithelial-mesenchymal transition (EMT) in primary airway epithelial cells (AECs). Semaphorin7A (Sema7a) plays a crucial role in regulating immune responses and initiating and maintaining transforming growth factor β1 TGF-β1-induced fibrosis. Objective To determine the expression of Sema7a, in serum isolated from asthmatics and non-asthmatics, the role of Sema7a in TGF-β1 induced proliferation, migration and airway EMT in human bronchial epithelial cells (HBECs) in vitro. Methods The concentrations of Sema7a in serum of asthmatic patients was detected by enzyme-linked immunosorbent assay (ELISA). The expressions of Sema7a and integrin-β1 were examined using conventional western blotting and real-time quantitative PCR (RT-PCR). Interaction between the Sema7a and Integrin-β1 was detected using the Integrin-β1 blocking antibody (GLPG0187). The changes in EMT indicators were performed by western blotting and immunofluorescence, as well as the expression levels of phosphorylated Focal-adhesion kinase (FAK) and Extracellular-signal-regulated kinase1/2 (ERK1/2) were analyzed by western blot and their mRNA expression was determined by RT-PCR. Results We described the first differentially expressed protein of sema7a, in patients with diagnosed bronchial asthma were significantly higher than those of healthy persons (P<0.05). Western blotting and RT-PCR showed that Sema7a and Integrin-β1 expression were significantly increased in lung tissue from the ovalbumin (OVA)-induced asthma model. GLPG0187 inhibited TGF-β1-mediated HBECs EMT, proliferation and migration, which was associated with Focal-adhesion kinase (FAK) and Extracellular-signal-regulated kinase1/2 (ERK1/2) phosphorylation. Conclusion Sema7a may play an important role in asthma airway remodeling by inducing EMT. Therefore, new therapeutic approaches for the treatment of chronic asthma, could be aided by the development of agents that target the Sema7a.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Degan Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| |
Collapse
|
10
|
Liu S, Liu M, Zhong J, Chen S, Wang Z, Gao X, Li F. Anti-S100A4 antibody administration alleviates bronchial epithelial-mesenchymal transition in asthmatic mice. Open Med (Wars) 2023; 18:20220622. [PMID: 37873538 PMCID: PMC10590613 DOI: 10.1515/med-2022-0622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 10/25/2023] Open
Abstract
We elucidated the effect of S100A4 on airway remodeling by regulating airway inflammation and epithelial-mesenchymal transition (EMT) in mouse models of asthma. Asthmatic mouse models were established by sensitization and challenged with ovalbumin (OVA). Anti-S100A4 antibody or control IgG antibody was administered daily before the OVA challenge. After the last challenge, airway inflammation and airway hyperresponsiveness were measured; lung tissues and bronchoalveolar lavage fluid (BALF) were harvested. Lung tissue sections were stained and evaluated for pathological changes. Levels of inflammatory cytokines were measured using ELISA. Levels of S100A4 and EMT markers were determined via western blotting analysis. Human bronchial epithelial cells were stimulated with 100 mg/mL house dust mites (HDMs) to evaluate the effect of S100A4 downregulation on EMT in vitro. S100A4 was increased in lung tissues and BALF from asthmatic mice. The asthmatic mice presented airway hyperresponsiveness, airway inflammation, and airway remodeling. After anti-S100A4 antibody administration, pathophysiological signs, including airway hyperresponsiveness and increased infiltration of inflammatory cells, were attenuated. Additionally, anti-S100A4 administration downregulated vimentin and α-SMA expression and upregulated E-cadherin expression in OVA-challenged mice. S100A4 downregulation also inhibited EMT process in HDM-stimulated 16HBE cells. Anti-S100A4 antibody administration alters airway remodeling by preventing EMT in mouse models of asthma.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Min Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Jinnan Zhong
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Shi Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Ziming Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Xiaoyan Gao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, Wuhan430000, Hubei, China
| | - Fajiu Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jianghan University, No. 168, Hong Kong Road, Jiang’an District, Wuhan430000, Hubei, China
| |
Collapse
|
11
|
Zhou JP, Wang Y, Li SQ, Zhang JQ, Lin YN, Sun XW, Zhou LN, Zhang L, Lu FY, Ding YJ, Li QY. Exogenous Ang-(1-7) inhibits autophagy via HIF-1α/THBS1/BECN1 axis to alleviate chronic intermittent hypoxia-enhanced airway remodelling of asthma. Cell Death Discov 2023; 9:366. [PMID: 37783703 PMCID: PMC10545676 DOI: 10.1038/s41420-023-01662-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023] Open
Abstract
Obstructive sleep apnoea (OSA)-induced chronic intermittent hypoxia (CIH) has been considered a risk factor for severe asthma. Airway remodelling, which could be modulated by autophagy, plays a key role in severe asthma. However, the extent of autophagy's involvement in CIH-potentiated airway remodelling remains largely unexplored. Furthermore, we had found that angiotensin-(1-7) [Ang-(1-7)] has therapeutic effects on airway remodelling in asthma, but the underlying mechanism is either unclear. This study aimed to explore how CIH aggravates asthma and mechanism of protective effects of Ang-(1-7) on airway remodelling, with a focus on autophagy. We observed that CIH promoted epithelial-to-mesenchymal transition (EMT), indicated by elevated EMT and fibrotic markers such as Snail and Collagen IV, both in vitro and in vivo. CIH intensified cell autophagy, evident from increased LC3B expression and reduced p62 levels. Ang-(1-7) reversed the CIH-enhanced expression of Snail, Collagen IV, and LC3B. To explore how CIH enhanced autophagy in cellular and animal model of asthma, overexpression of hypoxia-inducible factor 1-alpha (HIF-1α) and Thrombospondin 1 (THBS1) were identified in CIH-exposure mice lung compared with normal mice lung tissues from the GEO database. Finally, through chromatin immunoprecipitation and immunoprecipitation assays, we verified that Ang-(1-7) inhibits CIH-induced binding of HIF-1α to the promoter of THBS1, and also disrupts the protein-protein interaction between THBS1 and the autophagy-associated protein Beclin 1 (BECN1), ultimately leading to autophagy inhibition. Our findings suggest that exogenous Ang-(1-7) can inhibit autophagy via HIF-1α/THBS1/BECN1 axis, thereby alleviating CIH-enhanced airway remodelling in asthma. These findings imply the potential therapeutic effect of Ang-(1-7) in asthma with OSA.
Collapse
Affiliation(s)
- Jian Ping Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Shi Qi Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Jia Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Ying Ni Lin
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Xian Wen Sun
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Li Na Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Liu Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Fang Ying Lu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Yong Jie Ding
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China.
| |
Collapse
|
12
|
Siddiqui S, Bachert C, Bjermer L, Buchheit KM, Castro M, Qin Y, Rupani H, Sagara H, Howarth P, Taillé C. Eosinophils and tissue remodeling: Relevance to airway disease. J Allergy Clin Immunol 2023; 152:841-857. [PMID: 37343842 DOI: 10.1016/j.jaci.2023.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/15/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023]
Abstract
The ability of human tissue to reorganize and restore its existing structure underlies tissue homeostasis in the healthy airways, but in disease can persist without normal resolution, leading to an altered airway structure. Eosinophils play a cardinal role in airway remodeling both in health and disease, driving epithelial homeostasis and extracellular matrix turnover. Physiological consequences associated with eosinophil-driven remodeling include impaired lung function and reduced bronchodilator reversibility in asthma, and obstructed airflow in chronic rhinosinusitis with nasal polyps. Given the contribution of airway remodeling to the development and persistence of symptoms in airways disease, targeting remodeling is an important therapeutic consideration. Indeed, there is early evidence that eosinophil attenuation may reduce remodeling and disease progression in asthma. This review provides an overview of tissue remodeling in both health and airway disease with a particular focus on eosinophilic asthma and chronic rhinosinusitis with nasal polyps, as well as the role of eosinophils in these processes and the implications for therapeutic interventions. Areas for future research are also noted, to help improve our understanding of the homeostatic and pathological roles of eosinophils in tissue remodeling, which should aid the development of targeted and effective treatments for eosinophilic diseases of the airways.
Collapse
Affiliation(s)
- Salman Siddiqui
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Claus Bachert
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospital of Münster, Münster, Germany; First Affiliated Hospital, Sun Yat-Sen University, International Airway Research Center, Guangzhou, China; Division of Ear, Nose, and Throat Diseases, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden; Upper Airways Research Laboratory, Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Leif Bjermer
- Department of Clinical Sciences, Respiratory Medicine, and Allergology, Lund University, Lund, Sweden
| | - Kathleen M Buchheit
- Jeff and Penny Vinik Center for Allergic Diseases Research, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Mario Castro
- Division of Pulmonary, Critical Care Medicine, University of Kansas School of Medicine, Kansas City, NC
| | - Yimin Qin
- Global Medical Affairs, Global Specialty and Primary Care, GlaxoSmithKline, Research Triangle Park, NC
| | - Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Hironori Sagara
- Department of Medicine, Division of Respiratory Medicine and Allergology, Showa University, School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Peter Howarth
- Global Medical, Global Specialty and Primary Care, GlaxoSmithKline, Brentford, Middlesex, United Kingdom
| | - Camille Taillé
- Pneumology Department, Reference Center for Rare Pulmonary Diseases, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unit 1152, University of Paris Cité, Paris, France
| |
Collapse
|
13
|
Dai D, Xie J, Zheng Y, Chen F, Zhao B, Miao L. H3K27 acetylation-induced FSTL1 upregulation by P300/RUNX1 co-activation exacerbated autophagy-mediated neuronal damage and NF-κB-stimulated inflammation in Alzheimer's disease. Cytotechnology 2023; 75:449-460. [PMID: 37655275 PMCID: PMC10465437 DOI: 10.1007/s10616-023-00589-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/02/2023] [Indexed: 09/02/2023] Open
Abstract
Follistatin-like protein 1 (FSTL1) has been demonstrated to participate in the pathogenesis of several neurological diseases. The current study informed the role of H3K27 acetylation-induced FSTL1 upregulation in Alzheimer's disease (AD). Our investigation discovered the upregulated FSTL1 expression and enhanced autophagy activity in AD. FSTL1 knockdown successfully attenuated the injuries of Aβ1-42-challenged SH-SY5Y cells through the inhibition of autophagy activity. Besides, FSTL1 deficiency suppresses the inflammatory response and NF-κB signaling in AD. Moreover, it was found that p300 was recruited by transcriptional factor RUNX1 to stimulate the H3K27 acetylation in FSTL1 promoter region, which caused the upregulation of FSTL1 in AD. To summarize, p300 acted as a co-activator of RUNX1 to trigger the activation of FSTL1 in AD, resulting in the exacerbated injuries and inflammatory responses of Aβ1-42-induced SH-SY5Y cells.
Collapse
Affiliation(s)
- Dongmei Dai
- Department of Psychiatry, 904th Hospital of Joint Logistic Support Force of PLA, No. 55, Heping North Rd., Tianning District, Changzhou, 213000 Jiangsu China
| | - Junzheng Xie
- Department of Psychiatry, 904th Hospital of Joint Logistic Support Force of PLA, No. 55, Heping North Rd., Tianning District, Changzhou, 213000 Jiangsu China
| | - Yun Zheng
- Department of Psychiatry, 904th Hospital of Joint Logistic Support Force of PLA, No. 55, Heping North Rd., Tianning District, Changzhou, 213000 Jiangsu China
| | - Fangbin Chen
- Department of Psychiatry, 904th Hospital of Joint Logistic Support Force of PLA, No. 55, Heping North Rd., Tianning District, Changzhou, 213000 Jiangsu China
| | - Bin Zhao
- Department of Material Dependency, 904th Hospital of Joint Logistic Support Force of PLA, Changzhou, China
| | - Li Miao
- Department of Material Dependency, 904th Hospital of Joint Logistic Support Force of PLA, Changzhou, China
| |
Collapse
|
14
|
Zhou R, Jin C, Jiao L, Zhang S, Tian M, Liu J, Yang S, Yao W, Zhou F. Geranylgeranylacetone, an inducer of heat shock protein 70, attenuates pulmonary fibrosis via inhibiting NF-κB/NOX4/ROS signalling pathway in vitro and in vivo. Chem Biol Interact 2023; 382:110603. [PMID: 37307957 DOI: 10.1016/j.cbi.2023.110603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/21/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating and progressive pulmonary disease which is characterized by epithelial cell damage and extracellular collagen deposition. To date, the therapeutic options for IPF are still very limited, so the relevant mechanisms need to be explored. Heat shock protein 70 (HSP70), which has protective versus antitumor effects on cells under stress, is a member of the heat shock protein family. In the current study, qRT-PCR, western blotting, immunofluorescence staining, and migration assays were used to explore the Epithelial-mesenchymal transition (EMT) process in BEAS-2B cells. Moreover, the role of GGA in the process of pulmonary fibrosis was detected by HE, Masson staining, pulmonary function test and immunohistochemistry in C57BL/6 mice. Our results indicated that GGA, as an inducer of HSP70, enhanced the transformation of BEAS-2B cells from epithelial to mesenchymal cells through the NF-κB/NOX4/ROS (reactive oxygen species) signalling pathway and could significantly reduce apoptosis of BEAS-2B cells induced by TGF-β1(Transforming growth factor β1) in vitro. In vivo studies demonstrated that HSP70-inducing drugs, such as GGA, attenuated pulmonary fibrosis progression induced by bleomycin (BLM). Collectively, these results suggested that overexpression of HSP70 attenuated pulmonary fibrosis induced by BLM in C57BL/6 mice and EMT process induced by TGF-β1 through NF-κB/NOX4/ROS pathway in vitro. Thus, HSP70 might be a potential therapeutic strategy for human lung fibrosis.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Chaomei Jin
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Linlin Jiao
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Siyu Zhang
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Mei Tian
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Jiamin Liu
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Songtai Yang
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Wu Yao
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| | - Fang Zhou
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, China.
| |
Collapse
|
15
|
Hu J, Ding R, Liu S, Wang J, Li J, Shang Y. Hypermethylation of RNF125 promotes autophagy-induced oxidative stress in asthma by increasing HMGB1 stability. iScience 2023; 26:107503. [PMID: 37599832 PMCID: PMC10432822 DOI: 10.1016/j.isci.2023.107503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/08/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
Asthma is a global chronic airway disease. The expression and role of RNF125, an E3 ubiquitin ligase, in asthma remain uncertain. In this study, we revealed that RNF125 was downregulated in the bronchial epithelium of mice and patients with asthma. Rnf125 hypermethylation was responsible for the low expression of RNF125 in primary airway epithelial cells of mice treated with OVA. Moreover, we demonstrated that RNF125 could attenuate autophagy, oxidative stress, and protect epithelial barrier in vivo and in vitro. Additionally, we identified HMGB1 as a substrate of RNF125, which interacted with the HMG B-box domain of HMGB1 and induced degradation via the ubiquitin proteasome system, reducing autophagy and oxidative stress. Overall, our findings elucidated that hypermethylation of Rnf125 reduced its expression, which promoted autophagy-induced oxidative stress in asthma by increasing HMGB1 stability. These findings offer a theoretical and experimental basis for the pathogenesis of asthma.
Collapse
Affiliation(s)
- Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ruiwei Ding
- Pediatric Department, Qingdao Women and Children’s Hospital, Qingdao 266000, China
| | - Shaozhuang Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jia Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianjun Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
16
|
Liu L, Zhou L, Wang LL, Zheng PD, Zhang FQ, Mao ZY, Zhang HJ, Liu HG. Programmed Cell Death in Asthma: Apoptosis, Autophagy, Pyroptosis, Ferroptosis, and Necroptosis. J Inflamm Res 2023; 16:2727-2754. [PMID: 37415620 PMCID: PMC10321329 DOI: 10.2147/jir.s417801] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
Bronchial asthma is a complex heterogeneous airway disease, which has emerged as a global health issue. A comprehensive understanding of the different molecular mechanisms of bronchial asthma may be an efficient means to improve its clinical efficacy in the future. Increasing research evidence indicates that some types of programmed cell death (PCD), including apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, contributed to asthma pathogenesis, and may become new targets for future asthma treatment. This review briefly discusses the molecular mechanism and signaling pathway of these forms of PCD focuses on summarizing their roles in the pathogenesis and treatment strategies of asthma and offers some efficient means to improve clinical efficacy of therapeutics for asthma in the near future.
Collapse
Affiliation(s)
- Lu Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ling-Ling Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Peng-Dou Zheng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Feng-Qin Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhen-Yu Mao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Huo-Jun Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Hui-Guo Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
17
|
Chen SJ, Huang Y, Yu F, Feng X, Zheng YY, Li Q, Niu Q, Jiang YH, Zhao LQ, Wang M, Cheng PP, Song LJ, Liang LM, He XL, Xiong L, Xiang F, Wang X, Ma WL, Ye H. BMAL1/p53 mediating bronchial epithelial cell autophagy contributes to PM2.5-aggravated asthma. Cell Commun Signal 2023; 21:39. [PMID: 36803515 PMCID: PMC9940367 DOI: 10.1186/s12964-023-01057-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) is associated with increased incidence and severity of asthma. PM2.5 exposure disrupts airway epithelial cells, which elicits and sustains PM2.5-induced airway inflammation and remodeling. However, the mechanisms underlying development and exacerbation of PM2.5-induced asthma were still poorly understood. The aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) is a major circadian clock transcriptional activator that is also extensively expressed in peripheral tissues and plays a crucial role in organ and tissue metabolism. RESULTS In this study, we found PM2.5 aggravated airway remodeling in mouse chronic asthma, and exacerbated asthma manifestation in mouse acute asthma. Next, low BMAL1 expression was found to be crucial for airway remodeling in PM2.5-challenged asthmatic mice. Subsequently, we confirmed that BMAL1 could bind and promote ubiquitination of p53, which can regulate p53 degradation and block its increase under normal conditions. However, PM2.5-induced BMAL1 inhibition resulted in up-regulation of p53 protein in bronchial epithelial cells, then increased-p53 promoted autophagy. Autophagy in bronchial epithelial cells mediated collagen-I synthesis as well as airway remodeling in asthma. CONCLUSIONS Taken together, our results suggest that BMAL1/p53-mediated bronchial epithelial cell autophagy contributes to PM2.5-aggravated asthma. This study highlights the functional importance of BMAL1-dependent p53 regulation during asthma, and provides a novel mechanistic insight into the therapeutic mechanisms of BMAL1. Video Abstract.
Collapse
Affiliation(s)
- Shuai-Jun Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Yi Huang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Yuan-Yi Zheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Qian Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Ye-Han Jiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Li-Qin Zhao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xiaorong Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China. .,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China.
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China. .,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China.
| |
Collapse
|
18
|
Liu M, Liu S, Li F, Li C, Chen S, Gao X, Wang X. The miR-124-3p regulates the allergic airway inflammation and remodeling in an ovalbumin-asthmatic mouse model by inhibiting S100A4. Immun Inflamm Dis 2023; 11:e730. [PMID: 36799806 PMCID: PMC9896513 DOI: 10.1002/iid3.730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/27/2022] [Accepted: 10/13/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Asthma is a chronic respiratory disease with an increasing incidence every year. microRNAs (miRNAs) have been demonstrated to have implications for asthma. However, limited information is available regarding the effect of miR-124-3p on this disease. Therefore, this study aimed to explore the possible effects of miR-124-3p and S100A4 on inflammation and epithelial-mesenchymal transition (EMT) in asthma using mouse models. METHOD Ovalbumin was used to induce asthmatic mouse models. Lung injury in mouse models was assessed, and the bronchoalveolar lavage fluid of mice was collected to determine the number of eosinophilic granulocytes and assess inflammation. The expression levels of miR-124-3p, S100A4, E-cadherin, N-cadherin, Snail1, vimentin, and TGF-β1/Smad2 signaling pathway-related proteins were measured using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. In vitro experiments, cells were transfected with miR-124-3p mimics or inhibitors to test the expression of S100A4 by RT-qPCR and western blot analysis, and the mutual binding of miR-124-3p and S100A4 was validated by dual-luciferase reporter gene assay. RESULTS Overexpression of miR-124-3p or inhibition of S100A4 expression attenuated bronchial mucus secretion and collagenous fibers and suppressed inflammatory cell infiltration. Additionally, upon miR-124-3p overexpression or S100A4 suppression, eosinophilic granulocytes were decreased, interleukin-4 (IL-4) and IL-13 expression levels were reduced in the bronchoalveolar lavage fluid, serum total IgE level was reduced, and the TGF-β1/Smad2 signaling pathway was suppressed. Mechanically, a dual-luciferase reporter gene assay verified the binding relationship between miR-124-3p and S100A4. CONCLUSION miR-124-3p can negatively target S100A4 to attenuate inflammation in asthmatic mouse models by suppressing the EMT process and the TGF-β/smad2 signaling pathway.
Collapse
Affiliation(s)
- Min Liu
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Shuang Liu
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Fajiu Li
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Chenghong Li
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Shi Chen
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Xiaoyan Gao
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| | - Xiaojiang Wang
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Jianghan UniversityWuhanHubeiP.R. China
| |
Collapse
|
19
|
She W, Sun T, Long C, Chen M, Chen X, Liao Q, Wang M. Linc00511 Knockdown Inhibited TGF-β1-Induced Epithelial-Mesenchymal Transition of Bronchial Epithelial Cells by Targeting miR-16-5p/Smad3. Am J Rhinol Allergy 2023; 37:313-323. [PMID: 36594176 DOI: 10.1177/19458924221144853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Airway remodeling in patients with asthma was correlated with induced epithelial-mesenchymal transition (EMT) of bronchial epithelial cells. OBJECTIVE This study examined the mechanism of Linc00511 on induced EMT of bronchial epithelial cells after transforming growth factor-β1 (TGF-β1) induction. METHODS The human bronchial epithelial cell 16HBE was treated with 10 ng/mL TGF-β1 for 12 h, 24 h, or 48 h to induce EMT. Cell proliferation and migration rate were detected using CCK8 and wound healing assays, respectively. The expression of key markers of EMT (E-cadherin, N-cadherin, Small mothers against decapentaplegic family member 3 [Smad3], and slug) was tested by Western blot. RESULTS We found that Linc00511 was time dependently increased in TGF-β-treated 16HBE cells. Silencing Linc00511 reduced 16HBE cell proliferation, migration, and EMT progress. In addition, the dual-luciferase reporter assay showed Linc00511 was a molecular sponge for miR-16-5p. MiR-16-5p decreased the expression of Smad3 by targeting its 3'-untranslated region (3'UTR). After TGF-β1 exposure, miR-16-5p silencing counteracted the decreases of 16HBE cell proliferation, migration, and EMT induced by Linc00511 knockdown. And Smad3 overexpression also reversed the inhibitory effect of Linc00511 knockdown on proliferation, migration, and EMT progression in TGF-β1-induced human bronchial epithelial cells. CONCLUSION Linc00511 may be a valuable biomarker for asthma therapy.
Collapse
Affiliation(s)
- Weiwei She
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China.,Department of Respiratory and Critical Care Medicine, Nanxishan Hospital affiliated to Guilin Medical College, Guilin, China
| | - Tianshou Sun
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Chengfeng Long
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Meiyu Chen
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Xu Chen
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Qinxue Liao
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Mingdong Wang
- Department of Respiratory and Critical Care Medicine, 477248Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| |
Collapse
|
20
|
Dong H, Yang W, Li W, Zhu S, Zhu L, Gao P, Hao Y. New insights into autophagy in inflammatory subtypes of asthma. Front Immunol 2023; 14:1156086. [PMID: 37090692 PMCID: PMC10117973 DOI: 10.3389/fimmu.2023.1156086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Asthma is a heterogeneous airway disease characterized by airway inflammation and hyperresponsiveness. Autophagy is a self-degrading process that helps maintain cellular homeostasis. Dysregulation of autophagy is involved in the pathogenesis of many diseases. In the context of asthma, autophagy has been shown to be associated with inflammation, airway remodeling, and responsiveness to drug therapy. In-depth characterization of the role of autophagy in asthma can enhance the understanding of the pathogenesis, and provide a theoretical basis for the development of new biomarkers and targeted therapy for asthma. In this article, we focus on the relationship of autophagy and asthma, and discuss its implications for asthma pathogenesis and treatment.
Collapse
Affiliation(s)
- Hongna Dong
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Simin Zhu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Zhu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Peng Gao, ; Yuqiu Hao,
| | - Yuqiu Hao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Peng Gao, ; Yuqiu Hao,
| |
Collapse
|
21
|
Xia J, Tian Y, Shao Z, Li C, Ding M, Qi Y, Xu X, Dai K, Wu C, Yao W, Hao C. MALAT1-miR-30c-5p-CTGF/ATG5 axis regulates silica-induced experimental silicosis by mediating EMT in alveolar epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114392. [PMID: 36508811 DOI: 10.1016/j.ecoenv.2022.114392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/15/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Epithelial-mesenchymal transdifferentiation of alveolar type Ⅱ epithelial cells is a vital source of pulmonary myofibroblasts, and myofibroblasts formation is recognized as an important phase in the pathological process of silicosis. miR-30c-5p has been determined to be relevant in the activation of the epithelial-mesenchymal transition (EMT) in numerous disease processes. However, elucidating the role played by miR-30c-5p in the silicosis-associated EMT process remains a great challenge. In this work, based on the establishment of mouse silicosis and A549 cells EMT models, miR-30c-5p was interfered with in vivo and in vitro models to reveal its effects on EMT and autophagy. Moreover, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), connective tissue growth factor (CTGF), autophagy-related gene 5 (ATG5), and autophagy were further interfered with in the A549 cells models to uncover the possible molecular mechanism through which miR-30c-5p inhibits silicosis associated EMT. The results demonstrated the targeted binding of miR-30c-5p to CTGF, ATG5, and MALAT1, and showed that miR-30c-5p could prevent EMT in lung epithelial cells by acting on CTGF and ATG5-associated autophagy, thereby inhibiting the silicosis fibrosis process. Furthermore, we also found that lncRNA MALAT1 might competitively absorb miR-30c-5p and affect the EMT of lung epithelial cells. In a word, interfering with miR-30c-5p and its related molecules (MALAT1, CTGF, and ATG5-associated autophagy) may provide a reference point for the application of silicosis intervention-related targets.
Collapse
Affiliation(s)
- Jiarui Xia
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Yangyang Tian
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Zheng Shao
- The Third Affiliated Hospital of Zhengzhou University, Henan, PR China
| | - Chao Li
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Mingcui Ding
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Yuanmeng Qi
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Xiao Xu
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Kai Dai
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Chenchen Wu
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China
| | - Wu Yao
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China.
| | - Changfu Hao
- Department of Occupational and Environment Health, School of Public Health, Zhengzhou University, No. 100 Science Avenue 5, Zhengzhou 450001, Henan Province, PR China.
| |
Collapse
|
22
|
Rao J, Wang H, Ni M, Wang Z, Wang Z, Wei S, Liu M, Wang P, Qiu J, Zhang L, Wu C, Shen H, Wang X, Cheng F, Lu L. FSTL1 promotes liver fibrosis by reprogramming macrophage function through modulating the intracellular function of PKM2. Gut 2022; 71:2539-2550. [PMID: 35140065 PMCID: PMC9664121 DOI: 10.1136/gutjnl-2021-325150] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/23/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Follistatin-like protein 1 (FSTL1) is widely recognised as a secreted glycoprotein, but its role in modulating macrophage-related inflammation during liver fibrosis has not been documented. Herein, we aimed to characterise the roles of macrophage FSTL1 in the development of liver fibrosis. DESIGN Expression analysis was conducted with human liver samples obtained from 33 patients with liver fibrosis and 18 individuals without fibrosis serving as controls. Myeloid-specific FSTL1-knockout (FSTL1M-KO) mice were constructed to explore the function and mechanism of macrophage FSTL1 in 3 murine models of liver fibrosis induced by carbon tetrachloride injection, bile duct ligation or a methionine-deficient and choline-deficient diet. RESULTS FSTL1 expression was significantly elevated in macrophages from fibrotic livers of both humans and mice. Myeloid-specific FSTL1 deficiency effectively attenuated the progression of liver fibrosis. In FSTL1M-KO mice, the microenvironment that developed during liver fibrosis showed relatively less inflammation, as demonstrated by attenuated infiltration of monocytes/macrophages and neutrophils and decreased expression of proinflammatory factors. FSTL1M-KO macrophages exhibited suppressed proinflammatory M1 polarisation and nuclear factor kappa B pathway activation in vivo and in vitro. Furthermore, this study showed that, through its FK domain, FSTL1 bound directly to the pyruvate kinase M2 (PKM2). Interestingly, FSTL1 promoted PKM2 phosphorylation and nuclear translocation, reduced PKM2 ubiquitination to enhance PKM2-dependent glycolysis and increased M1 polarisation. Pharmacological activation of PKM2 (DASA-58) partially countered FSTL1-mediated glycolysis and inflammation. CONCLUSION Macrophage FSTL1 promotes the progression of liver fibrosis by inducing M1 polarisation and inflammation based on the intracellular PKM2 reprogramming function of macrophages.
Collapse
Affiliation(s)
- Jianhua Rao
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Hao Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Ni
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zeng Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyi Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Song Wei
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mu Liu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiannan Qiu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Wu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongbing Shen
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuehao Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Cheng
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Ling Lu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, Jiangsu, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Wang Y, Dong X, Pan C, Zhu C, Qi H, Wang Y, Wei H, Xie Q, Wu L, Shen H, Li S, Xie Y. Single-cell transcriptomic characterization reveals the landscape of airway remodeling and inflammation in a cynomolgus monkey model of asthma. Front Immunol 2022; 13:1040442. [PMID: 36439114 PMCID: PMC9685410 DOI: 10.3389/fimmu.2022.1040442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/20/2022] [Indexed: 06/22/2024] Open
Abstract
Monkey disease models, which are comparable to humans in terms of genetic, anatomical, and physiological characteristics, are important for understanding disease mechanisms and evaluating the efficiency of biological treatments. Here, we established an A.suum-induced model of asthma in cynomolgus monkeys to profile airway inflammation and remodeling in the lungs by single-cell RNA sequencing (scRNA-seq). The asthma model results in airway hyperresponsiveness and remodeling, demonstrated by pulmonary function test and histological characterization. scRNA-seq reveals that the model elevates the numbers of stromal, epithelial and mesenchymal cells (MCs). Particularly, the model increases the numbers of endothelial cells (ECs), fibroblasts (Fibs) and smooth muscle cells (SMCs) in the lungs, with upregulated gene expression associated with cell functions enriched in cell migration and angiogenesis in ECs and Fibs, and VEGF-driven cell proliferation, apoptotic process and complement activation in SMCs. Interestingly, we discover a novel Fib subtype that mediates type I inflammation in the asthmatic lungs. Moreover, MCs in the asthmatic lungs are found to regulate airway remodeling and immunological responses, with elevated gene expression enriched in cell migration, proliferation, angiogenesis and innate immunological responses. Not only the numbers of epithelial cells in the asthmatic lungs change at the time of lung tissue collection, but also their gene expressions are significantly altered, with an enrichment in the biological processes of IL-17 signaling pathway and apoptosis in the majority of subtypes of epithelial cells. Moreover, the ubiquitin process and DNA repair are more prevalent in ciliated epithelial cells. Last, cell-to-cell interaction analysis reveals a complex network among stromal cells, MCs and macrophages that contribute to the development of asthma and airway remodeling. Our findings provide a critical resource for understanding the principle underlying airway remodeling and inflammation in a monkey model of asthma, as well as valuable hints for the future treatment of asthma, especially the airway remodeling-characterized refractory asthma.
Collapse
Affiliation(s)
- Yingshuo Wang
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyan Dong
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Caizhe Pan
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Cihang Zhu
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hantao Qi
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Wang
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wei
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiangmin Xie
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Respiratory Drugs Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Wu
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijuan Shen
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuxian Li
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicheng Xie
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Sisto M, Ribatti D, Lisi S. Molecular Mechanisms Linking Inflammation to Autoimmunity in Sjögren's Syndrome: Identification of New Targets. Int J Mol Sci 2022; 23:13229. [PMID: 36362017 PMCID: PMC9658723 DOI: 10.3390/ijms232113229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 10/15/2023] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune rheumatic disorder characterized by the lymphocytic infiltration of exocrine glands and the production of autoantibodies to self-antigens. The involvement of the exocrine glands drives the pathognomonic manifestations of dry eyes (keratoconjunctivitis sicca) and dry mouth (xerostomia) that define sicca syndrome. To date, the molecular mechanisms mediating pathological salivary gland dysfunction in SS remain to be elucidated, despite extensive studies investigating the underlying cause of this disease, hampering the development of novel therapeutic strategies. Many researchers have identified a multifactorial pathogenesis of SS, including environmental, genetic, neuroendocrine, and immune factors. In this review, we explore the latest developments in understanding the molecular mechanisms involved in the pathogenesis of SS, which have attracted increasing interest in recent years.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 1, I-70124 Bari, Italy
| | | | | |
Collapse
|
25
|
Hu H, Guo L, Overholser J, Wang X. Mitochondrial VDAC1: A Potential Therapeutic Target of Inflammation-Related Diseases and Clinical Opportunities. Cells 2022; 11:cells11193174. [PMID: 36231136 PMCID: PMC9562648 DOI: 10.3390/cells11193174] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022] Open
Abstract
The multifunctional protein, voltage-dependent anion channel 1 (VDAC1), is located on the mitochondrial outer membrane. It is a pivotal protein that maintains mitochondrial function to power cellular bioactivities via energy generation. VDAC1 is involved in regulating energy production, mitochondrial oxidase stress, Ca2+ transportation, substance metabolism, apoptosis, mitochondrial autophagy (mitophagy), and many other functions. VDAC1 malfunction is associated with mitochondrial disorders that affect inflammatory responses, resulting in an up-regulation of the body’s defensive response to stress stimulation. Overresponses to inflammation may cause chronic diseases. Mitochondrial DNA (mtDNA) acts as a danger signal that can further trigger native immune system activities after its secretion. VDAC1 mediates the release of mtDNA into the cytoplasm to enhance cytokine levels by activating immune responses. VDAC1 regulates mitochondrial Ca2+ transportation, lipid metabolism and mitophagy, which are involved in inflammation-related disease pathogenesis. Many scientists have suggested approaches to deal with inflammation overresponse issues via specific targeting therapies. Due to the broad functionality of VDAC1, it may become a useful target for therapy in inflammation-related diseases. The mechanisms of VDAC1 and its role in inflammation require further exploration. We comprehensively and systematically summarized the role of VDAC1 in the inflammatory response, and hope that our research will lead to novel therapeutic strategies that target VDAC1 in order to treat inflammation-related disorders.
Collapse
Affiliation(s)
- Hang Hu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Linlin Guo
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (L.G.); (X.W.)
| | - Jay Overholser
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Correspondence: (L.G.); (X.W.)
| |
Collapse
|
26
|
Veraar C, Kirschner E, Schwarz S, Jaksch P, Hoetzenecker K, Tschernko E, Dworschak M, Ankersmit HJ, Moser B. Follistatin-like 1 and Biomarkers of Neutrophil Activation Are Associated with Poor Short-Term Outcome after Lung Transplantation on VA-ECMO. BIOLOGY 2022; 11:biology11101475. [PMID: 36290379 PMCID: PMC9598172 DOI: 10.3390/biology11101475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/17/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
The investigation of biomarkers associated with undesired outcome following lung transplantation (LuTX) is essential for a better understanding of the underlying pathophysiology, an earlier identification of susceptible recipients and the development of targeted therapeutic options. We therefore determined the longitudinal perioperative course of putative cytokines related to neutrophil activation (chemokine CC motif ligand 4 (CCL-4), interleukin (IL)-23 and Lipocalin 2 (LCN2)) and a cytokine that has been implicated in graft-versus-host disease (Follistatin-like 1 (FSTL1)) in 42 consecutive patients undergoing LuTX. We plotted receiver-operating curves (ROC) to assess the predictive power of the measured cytokines for short-term outcomes namely primary graft dysfunction (PGD), early complications requiring extracorporeal membrane oxygenation (ECMO), and a high postoperative sequential organ failure assessment (SOFA). All cytokines increased immediately after surgery. ROC analyses determined significant associations between CCL4 and a high SOFA score (area under the curve (AUC) 0.74 (95%CI:0.5−0.9; p < 0.05), between LCN2 and postoperative ECMO support (AUC 0.73 (95%CI:0.5−0.9; p < 0.05), and between FSTL1 and PGD (AUC 0.70 (95%CI:0.5−0.9; p < 0.05). The serum concentrations of the neutrophil-derived cytokines LCN2 and CCL4 as well as FSTL1 were all related to poor outcome after LuTX. The specific predictive power, however, still has to be assessed in larger trials. The potential role of FSTL1 as a biomarker in the development of PGD could be of great interest particularly since this protein appears to play a crucial role in allograft tolerance.
Collapse
Affiliation(s)
- Cecilia Veraar
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence:
| | - Enzo Kirschner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Schwarz
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Edda Tschernko
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Dworschak
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik J. Ankersmit
- Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
27
|
Sisto M, Ribatti D, Ingravallo G, Lisi S. The Expression of Follistatin-like 1 Protein Is Associated with the Activation of the EMT Program in Sjögren’s Syndrome. J Clin Med 2022; 11:jcm11185368. [PMID: 36143013 PMCID: PMC9503234 DOI: 10.3390/jcm11185368] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 12/11/2022] Open
Abstract
Background: The activation of the epithelial to mesenchymal transition (EMT) program is a pathological response of the Sjögren’s syndrome (SS) salivary glands epithelial cells (SGEC) to chronic inflammation. Follistatin-like 1 protein (FSTL1) is a secreted glycoprotein induced by transforming growth factor-β1 (TGF-β1), actively involved in the modulation of EMT. However, the role of FSTL1 in the EMT program activation in SS has not yet been investigated. Methods: TGF-β1-stimulated healthy human SGEC, SS SGEC, and SS salivary glands (SGs) biopsies were used to assess the effect of FSTL1 on the activation of the EMT program. FSTL1 gene activity was inhibited by the siRNA gene knockdown technique. Results: Here we reported that FSTL1 is up-regulated in SS SGs tissue in a correlated manner with the inflammatory grade. Blockage of FSTL1 gene expression by siRNA negatively modulates the TGF-β1-induced EMT program in vitro. We discovered that these actions were mediated through the modulation of the SMAD2/3-dependent EMT signaling pathway. Conclusions: Our data suggest that the TGF-β1-FSTL1-SMAD2/3 regulatory circuit plays a key role in the regulation of EMT in SS and targeting FSTL1 may be a strategy for the treatment of SGs EMT-dependent fibrosis.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs (SMBNOS), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: ; Tel.:+39-080-547-8315; Fax: +39-080-547-8327
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs (SMBNOS), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Sabrina Lisi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs (SMBNOS), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, 70121 Bari, Italy
| |
Collapse
|
28
|
Branched-chain amino acid transaminase 1 inhibition attenuates childhood asthma in mice by effecting airway remodeling and autophagy. Respir Physiol Neurobiol 2022; 306:103961. [PMID: 35961527 DOI: 10.1016/j.resp.2022.103961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/23/2022]
Abstract
Childhood asthma is a common chronic childhood disease. Branched-chain amino acid transaminase 1 (BCAT1) was reported to be upregulated in chronic airway diseases, while its role in childhood asthma is unclear. Asthma mouse models were established in neonatal mice by 10 µg ovalbumin (OVA) intraperitoneal injection and 3% OVA inhalational challenge. In OVA-challenged mice, BCAT1 levels were upregulated. BCAT1 inhibitor alleviated airway structure and inflammation by suppressing IgE, OVA-specific IgE and inflammatory cytokine release and inflammatory cell infiltration. BCAT1 inhibitor alleviated airway remodeling by inhibiting goblet cell hyperplasia, mucus secretion and the expression of α-SMA and collagen I/III. The BCAT1 inhibitor prevented OVA-enhanced autophagy by decreasing Beclin-1, Atg5 and LC3I/II and increasing p65 levels. In IL-13-stimulated BEAS-2B cells, rapamycin promoted inflammatory cytokine release and autophagy after BCAT1 inhibitor administration. Our research revealed that BCAT1 was upregulated in neonatal asthmatic mice and that a BCAT1 inhibitor might restrain airway inflammation and remodeling by decreasing autophagy, which offered a novel mechanistic understanding of childhood asthma.
Collapse
|
29
|
Guo F, Hao Y, Zhang L, Croteau-Chonka DC, Thibault D, Kothari P, Li L, Levy BD, Zhou X, Raby BA. Asthma Susceptibility Gene ORMDL3 Promotes Autophagy in Human Bronchial Epithelium. Am J Respir Cell Mol Biol 2022; 66:661-670. [PMID: 35353673 PMCID: PMC9163638 DOI: 10.1165/rcmb.2021-0305oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/03/2022] [Indexed: 02/05/2023] Open
Abstract
The genome-wide association study (GWAS)-identified asthma susceptibility risk alleles on chromosome 17q21 increase the expression of ORMDL3 (ORMDL sphingolipid biosynthesis regulator 3) in lung tissue. Given the importance of epithelial integrity in asthma, we hypothesized that ORMDL3 directly impacted bronchial epithelial function. To determine whether and how ORMDL3 expression impacts the bronchial epithelium, in studies using both primary human bronchial epithelial cells and human bronchial epithelial cell line, 16HBE (16HBE14o-), we assessed the impact of ORMDL3 on autophagy. Studies included: autophagosome detection by electron microscopy, RFP-GFP-LC3B to assess autophagic activity, and Western blot analysis of autophagy-related proteins. Mechanistic assessments included immunoprecipitation assays, intracellular calcium mobilization assessments, and cell viability assays. Coexpression of ORMDL3 and autophagy-related genes was measured in primary human bronchial epithelial cells derived from 44 subjects. Overexpressing ORMDL3 demonstrated increased numbers of autophagosomes and increased levels of autophagy-related proteins LC3B, ATG3, ATG7, and ATG16L1. ORMDL3 overexpression promotes autophagy and subsequent cell death by impairing intracellular calcium mobilization through interacting with SERCA2. Strong correlation was observed between expression of ORMDL3 and autophagy-related genes in patient-derived bronchial epithelial cells. Increased ORMDL3 expression induces autophagy, possibly through interacting with SERCA2, thereby inhibiting intracellular calcium influx, and induces cell death, impairing bronchial epithelial function in asthma.
Collapse
Affiliation(s)
- Feng Guo
- Channing Division of Network Medicine and
| | - Yuan Hao
- Channing Division of Network Medicine and
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts; and
| | - Li Zhang
- Channing Division of Network Medicine and
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | | | | | | | - Lijia Li
- Channing Division of Network Medicine and
| | - Bruce D. Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Xiaobo Zhou
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Benjamin A. Raby
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
30
|
Liu F, Zhang J, Zhang D, Qi Q, Cui W, Pan Y, Liu X, Xu J, Qiao X, Wang Z, Dong L. Follistatin-related protein 1 in asthma: miR-200b-3p interactions affect airway remodeling and inflammation phenotype. Int Immunopharmacol 2022; 109:108793. [PMID: 35483234 DOI: 10.1016/j.intimp.2022.108793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/05/2022]
Abstract
Follistatin-related protein 1 (FSTL1) is significantly associated with the asthma severity and outcome in humans and diverse mouse models of asthma. Previous studies have also suggested that FSTL1 could activate autophagy and NLRP3, thus playing as a causative agent in the asthma progression. However, mechanisms that regulate airway epithelial cell-specific FSTL1 expression and function in asthma are unknown. Here, we further evaluated the spatiotemporal relationships between the FSTL1 and asthma development through ovalbumin (OVA) -induced asthma models. Integrative analysis in asthmatics airway epithelium identifies microRNA (miR)-200b-3p as a novel upstream of FSTL1. Next, we collected airway biopsies, induced sputum, and blood samples isolated from asthmatics patients and the OVA-induced mouse model. We revealed that miR-200b-3p expression is downregulated in asthmatics airway epithelium, while its expression was negatively correlated with FSTL1. On this basis, the function and expression pattern analysis of miR-200b-3p were performed using miRNA-target prediction databases and long non-coding RNA (lncRNA) microarray assay. It is illustrated that miR-200b-3p, which is downregulated with pro-fibrotic stimulation of TGF-β1, could also be sponged by lncRNA PCAT19 and regulate FSTL1 expression in asthma progression. In vivo, miR-200b-3p overexpression in mice prevents OVA-induced airway remodeling and inflammation. Lastly, protective roles of miR-200b-3p are partly attributed to the direct and functional repression of FSTL1. Our findings suggest a crucial role for the miR-200b-3p/FSTL1 axis in regulating asthmatic's airway remodeling and inflammation phenotype.
Collapse
Affiliation(s)
- Fen Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qian Qi
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Wenjing Cui
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jiawei Xu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Xinrui Qiao
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zihan Wang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China; Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
31
|
Liu H, Guan Q, Zhao P, Li J. TGF-β-induced CCR8 promoted macrophage transdifferentiation into myofibroblast-like cells. Exp Lung Res 2022:1-14. [PMID: 35377281 DOI: 10.1080/01902148.2022.2055227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/18/2022] [Accepted: 03/13/2022] [Indexed: 11/04/2022]
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is an interstitial disease of unknown origin, characterized by tissue fibrosis, for which currently there is no effective treatment. Macrophages, the main immune cells in lung tissue, are involved in the whole process of pulmonary fibrosis. In recent years, intercellular transformation has led to wide spread concern among pulmonary fibrosis researchers. Macrophages with flexible heterogeneity and plasticity participate in different physiological processes in the body. Cell chemokine receptor 8 (CCR8) is expressed in a variety of cells and plays a significant chemotactic role in the induction of cell activation and migration. It can also promote the differentiation of macrophages under certain environmental conditions. The current study is intended to explore the role of CCR8 in macrophage to myofibroblast transdifferentiation (MMT) in IPF. Methods: We conducted experiments using CCR8-specific small interfering RNA (siRNA), an autophagy inhibitor (3-methyladenine, 3-MA), and an agonist (rapamycin) to explore the underlying mechanisms of macrophage transdifferentiation into myofibroblast cells in transforming growth factor-beta (TGF-β)-induced pulmonary fibrosis. Results: TGF-β treatment increased the CCR8 protein level in a time- and dose-dependent manner in mouse alveolar macrophages, as well as macrophage transdifferentiation-related markers, including vimentin, collagen 1, and a-SMA, and cell migration. In addition, the levels of autophagy were enhanced in macrophages treated with TGF-β. We found that 3-MA, an autophagy inhibitor, decreased the expression levels of macrophage transdifferentiation-related markers and attenuated cell migration. Furthermore, the inhibition of CCR8 via CCR8-specific siRNA reduced the levels of autophagy and macrophage transdifferentiation-related markers, and inhibited the cell migration. Enhancing autophagy with rapamycin attenuated the inhibition effect of CCR8-specific siRNA on macrophage migration and the increase in myofibroblast marker proteins. Conclusions: Our findings showed that the macrophages exposed to TGF-β had the potential to transdifferentiate into myofibroblasts and CCR8 was involved in the process. The effect of CCR8 on TGF-β-induced macrophage transdifferentiation occurs mainly through autophagy. Targeting CCR8 may be a novel therapeutic strategy for the treatment of IPF.
Collapse
Affiliation(s)
- Haijun Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructedby Henan province & Education Ministry of People's Republic of China, Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qingzhou Guan
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructedby Henan province & Education Ministry of People's Republic of China, Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Peng Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructedby Henan province & Education Ministry of People's Republic of China, Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructedby Henan province & Education Ministry of People's Republic of China, Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
32
|
Zhang D, Yang H, Dong XL, Zhang JT, Liu XF, Pan Y, Zhang J, Xu JW, Wang ZH, Cui WJ, Dong L. TL1A/DR3 Axis, A Key Target of TNF-a, Augments the Epithelial–Mesenchymal Transformation of Epithelial Cells in OVA-Induced Asthma. Front Immunol 2022; 13:854995. [PMID: 35359966 PMCID: PMC8963920 DOI: 10.3389/fimmu.2022.854995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/16/2022] [Indexed: 01/16/2023] Open
Abstract
Tumor necrosis factor (TNF)-like cytokine 1A (TL1A), a member of the TNF family, exists in the form of membrane-bound (mTL1A) and soluble protein (sTL1A). TL1A binding its only known functional receptor death domain receptor 3 (DR3) affects the transmission of various signals. This study first proposed that the TL1A/DR3 axis was significantly upregulated in patients and mice with both asthma and high TNF-a expression and in TNF-a-stimulated epithelial Beas-2B cells. Two independent approaches were used to demonstrate that the TL1A/DR3 axis of mice was strongly correlated with TNF-a in terms of exacerbating asthmatic epithelial–mesenchymal transformation (EMT). First, high expression levels of EMT proteins (e.g., collagen I, fibronectin, N-cadherin, and vimentin) and TL1A/DR3 axis were observed when mice airways were stimulated by recombinant mouse TNF-a protein. Moreover, EMT protein and TL1A/DR3 axis expression synchronously decreased after mice with OVA-induced asthma were treated with infliximab by neutralizing TNF-a activity. Furthermore, the OVA-induced EMT of asthmatic mice was remarkably improved upon the deletion of the TL1A/DR3 axis by knocking out the TL1A gene. TL1A siRNA remarkably intervened EMT formation induced by TNF-a in the Beas-2B cells. In addition, EMT was induced by the addition of high concentrations of recombinant human sTL1A with the cell medium. The TL1A overexpression via pc-mTL1A in vitro remarkably increased the EMT formation induced by TNF-a. Overall, these findings indicate that the TL1A/DR3 axis may have a therapeutic role for asthmatic with high TNF-a level.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Yang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue-Li Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jin-Tao Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Fei Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Zhang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jia-Wei Xu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Zi-Han Wang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen-Jing Cui
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
- *Correspondence: Liang Dong,
| |
Collapse
|
33
|
Zhang J, Zhang D, Pan Y, Liu X, Xu J, Qiao X, Cui W, Dong L. The TL1A-DR3 Axis in Asthma: Membrane-Bound and Secreted TL1A Co-Determined the Development of Airway Remodeling. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:233-253. [PMID: 35255540 PMCID: PMC8914606 DOI: 10.4168/aair.2022.14.2.233] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/17/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022]
Abstract
Purpose Tumor necrosis factor-like ligand 1A (TL1A), especially its secreted form, has been shown to contribute to eosinophilic inflammation and mucus production, cardinal features of asthma, through its receptor, death receptor 3 (DR3). However, the role of the TL1A-DR3 axis in asthma, especially in terms of airway remodeling, has not yet been fully understood. Methods The present study investigated the expression and secretion of TL1A in the lung and human bronchial epithelial cells. DR3 small interfering RNA (siRNA), TL1A siRNA, and truncated plasmids were used respectively to identify the function of the TL1A-DR3 axis in vitro. To further validate the roles of the TL1A-DR3 axis in asthma, we collected airway biopsies and sputa from asthmatic patients and constructed a mouse model following rTL1A administration, DR3 knockdown, and TL1A knockout, the asthma-related inflammatory response and the pathological changes in airways were analyzed using various experimental methods. Associated signaling pathways downstream of TL1A knockout in the mouse model were analyzed using RNA sequencing. Results TL1A, especially its non-secreted form (nsTL1A) was involved in the remodeling process in asthmatics’ airways. Knockdown of TL1A or its receptor DR3 decreased the expression of fibrosis-associated protein in BEAS-2B cells. Reversely, overexpression of nsTL1A in airway epithelial cells facilitated the transforming growth factor-β-induced remodeling progress. In the asthma mouse model, activating the TL1A-DR3 axis contributes to airway inflammation, remodeling, and tissue destruction. Reciprocally, DR3 knockdown or TL1A knockout partly reverses airway remodeling in the asthma model induced by ovalbumin. Conclusions Our results confirm differential TL1A expression (including its secreted and non-secreted form) in asthma, which modulates remodeling. The shared mechanism of action by which nsTL1A and secreted TL1A exert their effects on asthma development might be mediated via the nuclear factor-κB pathway. The TL1A-DR3 axis presents a promising therapeutic target in asthma.
Collapse
Affiliation(s)
- Jintao Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Liu
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiawei Xu
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinrui Qiao
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjing Cui
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China.
| |
Collapse
|
34
|
Wolf J, Hajdu RI, Boneva S, Schlecht A, Lapp T, Wacker K, Agostini H, Reinhard T, Auw-Hädrich C, Schlunck G, Lange C. Characterization of the Cellular Microenvironment and Novel Specific Biomarkers in Pterygia Using RNA Sequencing. Front Med (Lausanne) 2022; 8:714458. [PMID: 35174178 PMCID: PMC8841401 DOI: 10.3389/fmed.2021.714458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/24/2021] [Indexed: 01/04/2023] Open
Abstract
With a worldwide prevalence of ~12%, pterygium is a common degenerative and environmentally triggered ocular surface disorder characterized by wing-shaped growth of conjunctival tissue onto the cornea that can lead to blindness if left untreated. This study characterizes the transcriptional profile and the cellular microenvironment of conjunctival pterygia and identifies novel pterygia-specific biomarkers. Formalin-fixed and paraffin-embedded pterygia as well as healthy conjunctival specimens were analyzed using MACE RNA sequencing (n = 8 each) and immunohistochemistry (pterygia n = 7, control n = 3). According to the bioinformatic cell type enrichment analysis using xCell, the cellular microenvironment of pterygia was characterized by an enrichment of myofibroblasts, T-lymphocytes and various antigen-presenting cells, including dendritic cells and macrophages. Differentially expressed genes that were increased in pterygia compared to control tissue were mainly involved in autophagy (including DCN, TMBIM6), cellular response to stress (including TPT1, DDX5) as well as fibroblast proliferation and epithelial to mesenchymal transition (including CTNNB1, TGFBR1, and FN1). Immunohistochemical analysis confirmed a significantly increased FN1 stromal immunoreactivity in pterygia when compared to control tissue. In addition, a variety of factors involved in apoptosis were significantly downregulated in pterygia, including LCN2, CTSD, and NISCH. Furthermore, 450 pterygia-specific biomarkers were identified by including transcriptional data of different ocular surface pathologies serving as controls (training group), which were then validated using transcriptional data of cultured human pterygium cells. Among the most pterygia-specific factors were transcripts such as AHNAK, RTN4, TPT1, FSTL1, and SPARC. Immunohistochemical validation of SPARC revealed a significantly increased stromal immunoreactivity in pterygia when compared to controls, most notably in vessels and intravascular vessel wall-adherent mononuclear cells. Taken together, the present study provides new insights into the cellular microenvironment and the transcriptional profile of pterygia, identifies new and specific biomarkers and in addition to fibrosis-related genes, uncovers autophagy, stress response and apoptosis modulation as pterygium-associated processes. These findings expand our understanding of the pathophysiology of pterygia, provide new diagnostic tools, and may enable new targeted therapeutic options for this common and sight-threatening ocular surface disease.
Collapse
Affiliation(s)
- Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Rozina Ida Hajdu
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anja Schlecht
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Institute of Anatomy and Cell Biology, Wuerzburg University, Wuerzburg, Germany
| | - Thabo Lapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Katrin Wacker
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Claudia Auw-Hädrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, Münster, Germany
- *Correspondence: Clemens Lange
| |
Collapse
|
35
|
Suzuki Y, Aono Y, Akiyama N, Horiike Y, Naoi H, Horiguchi R, Shibata K, Hozumi H, Karayama M, Furuhashi K, Enomoto N, Fujisawa T, Nakamura Y, Inui N, Suda T. Involvement of autophagy in exacerbation of eosinophilic airway inflammation in a murine model of obese asthma. Autophagy 2022; 18:2216-2228. [PMID: 35098856 PMCID: PMC9397451 DOI: 10.1080/15548627.2022.2025571] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Obesity is a common comorbidity in patients with asthma, and obese asthma patients present the most refractory phenotype among patients with severe asthma. Similar to the observations in non-obese asthma patients, clinical studies have revealed heterogeneity in obese asthma patients, including the occurrences of T helper (Th)2-high and Th2-low phenotypes. However, the mechanisms underlying obesity-related asthma are not completely understood. Though macroautophagy/autophagy is involved in asthma and obesity, its role in obesity-associated asthma is unknown. We hypothesized that autophagy is involved in the pathogenesis of obese asthma. For our investigations, we used high-fat diet-induced Atg5 (autophagy related 5)-deficient mice and epithelial cell-specific atg5−/− (Scgb1a1/CCSP-atg5−/−) obesity-induced mice. House dust mite (HDM)-sensitized atg5−/− obese mice exhibited marked eosinophilic inflammation and airway hyper-reactivity (AHR), compared to wild-type (WT) obese mice. Analyses of atg5−/− obese mice showed increased levels of Th2 cells but not ILC2s together with elevated expression of Th2 cytokines in the lung. In response to the HDM challenge, activated epithelial autophagy was observed in lean but not obese WT mice. Epithelium-specific deletion of Atg5 induced eosinophilic inflammation in Scgb1a1/CCSP-atg5−/− obese mice, and genetic analyses of epithelial cells from HDM-immunized atg5−/− obesity-induced mice showed an elevated expression of thymic stromal lymphopoietin (TSLP) and IL33. Notably, HDM-sensitized atg5−/− mice developed TSLP- and IL33-dependent eosinophilic inflammation and AHR. Our results suggest that autophagy contributes to the exacerbation of eosinophilic inflammation in obese asthma. Modulations of autophagy may be a therapeutic target in obesity-associated asthma. Abbreviations: AHR: airway hyper-reactivity; BAL: bronchoalveolar lavage; Cdyn: dynamic compliance; BM: bone marrow; HDM: house dust mite; HFD: high-fat diet; ILC2s: type 2 innate lymphocyte cells; ROS: reactive oxygen species; RL: lung resistance; TSLP: thymic stromal lymphopoietin; TCC: total cell count; WT: wild type.
Collapse
Affiliation(s)
- Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuya Aono
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Norimichi Akiyama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuoki Horiike
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hyogo Naoi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryo Horiguchi
- Advanced Research Facilities and Services, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kiyoshi Shibata
- Advanced Research Facilities and Services, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
36
|
Song D, Jiang Y, Zhao Q, Li J, Zhao Y. lncRNA-NEAT1 Sponges miR-128 to Promote Inflammatory Reaction and Phenotypic Transformation of Airway Smooth Muscle Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7499911. [PMID: 35082915 PMCID: PMC8786537 DOI: 10.1155/2022/7499911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Pediatric asthma is still a health threat to the children. Long noncoding RNA-NEAT1 (lncRNA-NEAT1) was reported to be positively correlated with the severity of asthma. We aimed to study the effects and mechanism of lncRNA-NEAT1on inflammatory reaction and phenotypic transformation of airway smooth muscle cells (ASMCs) in the bronchial asthma. METHOD The degree of lncRNA-NEAT1 and miR-128 mRNA in children with bronchial asthma and healthy individuals was tested by qRT-PCR. After the inflammatory reaction and phenotypic transformation of PDGF-BB-induced ASMCs, the expression of lncRNA-NEAT1 or miR-128 in the AMSC was disturbed in the AMSC. Subsequently, the expression of lncRNA-NEAT1 and miR-128 was detected by the way of qRT-PCR, and western blot was applied to measure the expression of MMP-2, MMP-9, α-SMA, calponin, NF-κB, and so on in the cells. The content of TNF-α, IL-1β, IL-6, and IL-8 in the cell culture supernatant was checked by ELISA. MTT, Transwell, and flow cytometry were used to detect cell proliferation, migration, and apoptosis. Further, the targeting relations between lncRNA-NEAT1 and miR-128 were evaluated by the dual-luciferase reporter assay. RESULT In the sputum of children with bronchial asthma, lncRNA-NEAT1 was significantly upregulated while miR-128 was rapidly downregulated. Besides, lncRNA-NEAT1 and miR-128 were competitively combined and, for their expression, negatively correlated. CONCLUSION lncRNA-NEAT1 sponges miR-128 to boost PDGF-BB-induced inflammatory reaction and phenotypic transformation of ASMCs to aggravate the occurrence and development of childhood bronchial asthma.
Collapse
Affiliation(s)
- Danyang Song
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Yajing Jiang
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Qiuju Zhao
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Jinling Li
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Yuqi Zhao
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| |
Collapse
|
37
|
Follistatin-Like 1 Induces the Activation of Type 2 Innate Lymphoid Cells to Promote Airway Inflammation in Asthma. Inflammation 2021; 45:904-918. [PMID: 34757553 DOI: 10.1007/s10753-021-01594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
Asthma is a chronic disease closely related to airway inflammation. It has been proven that type 2 innate lymphoid cells (ILC2s) play an essential role in airway inflammation in asthma. Furthermore, there is growing evidence that Follistatin-like 1 (FSTL1) can participate in various inflammatory reactions mediated by the JAK/STAT signaling pathway, among others. Therefore, we put forward a new hypothesis: FSTL1 promotes asthmatic airway inflammation by activating ILC2. This study generated an ovalbumin-sensitized asthma model in C57BL/6 and Fstl1+/- mice. The results showed that the absolute number and the proportion of ILC2 in the ovalbumin-challenged Fstl1+/- group were lower than in the ovalbumin-challenged wild-type group. We also measured the levels of Th2-type cytokines in the serum and bronchoalveolar lavage fluid (BALF) of mice and found that the corresponding cytokines in the Fstl1+/- were lower than in the wild-type groups. Finally, we tested whether MEK-JAK-STAT-GATA3 is the specific pathway for FSTL1 to activate ILC2, and further tested our working hypothesis by adding various inhibitors of proteins from this pathway. Overall, these findings reveal that FSTL1 can activate ILC2 through MEK-JAK-STAT-GATA3 to promote airway inflammation and participate in the pathogenesis of asthma.
Collapse
|
38
|
He H, Cao L, Wang Z, Wang Z, Miao J, Li XM, Miao M. Sinomenine Relieves Airway Remodeling By Inhibiting Epithelial-Mesenchymal Transition Through Downregulating TGF-β1 and Smad3 Expression In Vitro and In Vivo. Front Immunol 2021; 12:736479. [PMID: 34804018 PMCID: PMC8602849 DOI: 10.3389/fimmu.2021.736479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/14/2021] [Indexed: 01/07/2023] Open
Abstract
Airway remodeling is associated with dysregulation of epithelial-mesenchymal transition (EMT) in patients with asthma. Sinomenine (Sin) is an effective, biologically active alkaloid that has been reported to suppress airway remodeling in mice with asthma. However, the molecular mechanisms behind this effect remain unclear. We aimed to explore the potential relationship between Sin and EMT in respiratory epithelial cells in vitro and in vivo. First, 16HBE cells were exposed to 100 μg/mL LPS and treated with 200 μg/mL Sin. Cell proliferation, migration, and wound healing assays were performed to evaluate EMT, and EMT-related markers were detected using Western blotting. Mice with OVA-induced asthma were administered 35 mg/kg or 75 mg/kg Sin. Airway inflammation and remodeling detection experiments were performed, and EMT-related factors and proteins in the TGF-β1 pathway were detected using IHC and Western blotting. We found that Sin suppressed cell migration but not proliferation in LPS-exposed 16HBE cells. Sin also inhibited MMP7, MMP9, and vimentin expression in 16HBE cells and respiratory epithelial cells from mice with asthma. Furthermore, it decreased OVA-specific IgE and IL-4 levels in serum, relieved airway remodeling, attenuated subepithelial collagen deposition, and downregulating TGF-β1and Smad3 expression in mice with asthma. Our results suggest that Sin suppresses EMT by inhibiting IL-4 and downregulating TGF-β1 and Smad3 expression.
Collapse
Affiliation(s)
- Hongjuan He
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lihua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zheng Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhenzhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiu-Min Li
- Microbiology and Immunology, and Department of Otolaryngology, New York Medical College, New York, NY, United States
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
39
|
Zhao J, Zhang J, Tang S, Wang J, Liu T, Zeng R, Zhu W, Zhang K, Wu J. The different functions of short and long thymic stromal lymphopoietin isoforms in autophagy-mediated asthmatic airway inflammation and remodeling. Immunobiology 2021; 226:152124. [PMID: 34333403 DOI: 10.1016/j.imbio.2021.152124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 11/16/2022]
Abstract
Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling as well as hyper-responsiveness. Thymic stromal lymphopoietin (TSLP), which is a crucial inflammatory cytokine in immune homeostasis, consists of two isoforms, the long isoform lfTSLP and short isoform sfTSLP. The lfTSLP promotes inflammation and plays a pivotal role in asthma pathogenesis, while sfTSLP had been reported to have anti-asthma effects. Experiments have shown that lfTSLP could induce autophagy in hepatocytes. It is unknown whether lfTSLP or sfTSLP could influence autophagy and affect the progression of asthma. Using house dust mite (HDM)-stimulated airway smooth muscle cells as an in vitro model and HDM-induced asthma mice as in vivo model, we found that lfTSLP could induce autophagy and remodeling, while sfTSLP has the reverse effect. Strikingly, sfTSLP treatment in vivo reversed HDM-mediated activation of inflammation and airway remodeling, partly determined by autophagy change. These findings may help us understand the function of TSLP isoforms in the pathogenesis of asthma, and they support the use of drugs targeting sfTSLP and TSLP for asthma treatment.
Collapse
Affiliation(s)
- Jiping Zhao
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Shuangmei Tang
- Department of Otolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junfei Wang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tian Liu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Rong Zeng
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Weichun Zhu
- Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Kangda Zhang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jinxiang Wu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
40
|
Follistatin-Like Proteins: Structure, Functions and Biomedical Importance. Biomedicines 2021; 9:biomedicines9080999. [PMID: 34440203 PMCID: PMC8391210 DOI: 10.3390/biomedicines9080999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022] Open
Abstract
Main forms of cellular signal transmission are known to be autocrine and paracrine signaling. Several cells secrete messengers called autocrine or paracrine agents that can bind the corresponding receptors on the surface of the cells themselves or their microenvironment. Follistatin and follistatin-like proteins can be called one of the most important bifunctional messengers capable of displaying both autocrine and paracrine activity. Whilst they are not as diverse as protein hormones or protein kinases, there are only five types of proteins. However, unlike protein kinases, there are no minor proteins among them; each follistatin-like protein performs an important physiological function. These proteins are involved in a variety of signaling pathways and biological processes, having the ability to bind to receptors such as DIP2A, TLR4, BMP and some others. The activation or experimentally induced knockout of the protein-coding genes often leads to fatal consequences for individual cells and the whole body as follistatin-like proteins indirectly regulate the cell cycle, tissue differentiation, metabolic pathways, and participate in the transmission chains of the pro-inflammatory intracellular signal. Abnormal course of these processes can cause the development of oncology or apoptosis, programmed cell death. There is still no comprehensive understanding of the spectrum of mechanisms of action of follistatin-like proteins, so the systematization and study of their cellular functions and regulation is an important direction of modern molecular and cell biology. Therefore, this review focuses on follistatin-related proteins that affect multiple targets and have direct or indirect effects on cellular signaling pathways, as well as to characterize the directions of their practical application in the field of biomedicine.
Collapse
|
41
|
Kaur D, Chachi L, Gomez E, Sylvius N, Brightling CE. Interleukin-18, IL-18 binding protein and IL-18 receptor expression in asthma: a hypothesis showing IL-18 promotes epithelial cell differentiation. Clin Transl Immunology 2021; 10:e1301. [PMID: 34194747 PMCID: PMC8234286 DOI: 10.1002/cti2.1301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 01/15/2023] Open
Abstract
Objective In asthma, genome‐wide association studies have shown that interleukin‐18 (IL‐18) receptor 1 gene (IL‐18R1) and sputum IL‐18 are increased during exacerbations. However, the role of the IL‐18 axis in bronchial epithelial function is unclear. To investigate IL‐18, IL‐18 binding protein (BP) and IL‐18R expression in bronchial biopsies and sputum samples from patients with asthma, and to determine its functional role using in vitro bronchial epithelial cells. Methods The expression of IL‐18, IL‐18BP and IL‐18Rα was examined in subjects with asthma and healthy controls in bronchial biopsies by immunohistochemistry and IL‐18 and IL‐18BP release in sputum. In epithelial cells, the mRNA and protein expression of IL‐18, IL‐18BP, IL‐18Rα and IL‐18Rβ was assessed by qPCR, flow cytometry, Western blotting and immunofluorescence respectively. IL‐18 function in epithelial cells was examined by intracellular calcium, wound repair, synthetic activation and epithelial differentiation changes. Results In biopsies from subjects with asthma, the IL‐18 expression was not different in the lamina propria compared with controls but was decreased in the epithelium. In contrast, the IL‐18BP was decreased in the lamina propria in asthma and was absent in the bronchial epithelium. IL‐18 was released in sputum with IL‐18BP elevated in patients with asthma. The IL‐18Rα expression was not different between health and disease. In vitro, IL‐18‐stimulated bronchial epithelial cells increased intracellular calcium, wound repair, metabolic activity, morphological changes and epithelial cellular differentiation. Conclusion In asthma, the dynamic interaction between IL‐18, its cognate receptor and natural inhibitor is complex, with differences between airway compartments. Upregulation of IL‐18 can promote epithelial activation and cellular differentiation.
Collapse
Affiliation(s)
- Davinder Kaur
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Latifa Chachi
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Edith Gomez
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Nicolas Sylvius
- Genomic Core Facility Department of Genetics University of Leicester Adrian Building, University Road, G23 Leicester LE1 7RH UK
| | - Christopher E Brightling
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| |
Collapse
|
42
|
Wu H, Wang D, Shi H, Liu N, Wang C, Tian J, Wang X, Zhang Z. PM 2.5 and water-soluble components induce airway fibrosis through TGF-β1/Smad3 signaling pathway in asthmatic rats. Mol Immunol 2021; 137:1-10. [PMID: 34175710 DOI: 10.1016/j.molimm.2021.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 01/12/2023]
Abstract
Epidemiological studies have suggested that fine particulate matter (PM2.5) and asthma have been independently associated with pulmonary fibrosis but rarely studied together. Furthermore, it is unknown whether airway fibrosis in asthma is more attributable to water-soluble ions of PM2.5. Our current study was to explore the potential mechanism of PM2.5 and water-soluble components on airway fibrosis in ovalbumin (OVA)-sensitized asthmatic rats. Rats were intratracheally instilled with PM2.5 and water-soluble components every 3 days for 4 times or 8 times. Histopathological examination demonstrated that lung inflammatory and airway fibrosis were induced after PM2.5 and water-soluble components exposure. Meanwhile, PM2.5, in particular water-soluble extracts, increased expression of collagen 1 (COL-1), connective tissue growth factor (CTGF), interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), Smad family member 3 (Smad3), and p-Smad3, whereas decreased secretion of heme oxygenase-1 (HO-1). However, pretreating asthmatic rats with SB432542, the inhibitor of TGF-β1, and SIS3 HCl, the antagonist of Smad3, both reversed the activation of airway fibrosis induced by water-soluble extracts. Therefore, TGF-β1/Smad3 signaling pathway may be responsible for the pathological process of airway fibrosis in asthmatic rats following PM2.5 and water-soluble components exposure.
Collapse
Affiliation(s)
- Hongyan Wu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Dan Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China.
| |
Collapse
|
43
|
Carlier FM, de Fays C, Pilette C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front Physiol 2021; 12:691227. [PMID: 34248677 PMCID: PMC8264588 DOI: 10.3389/fphys.2021.691227] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells, which provide a complex and adaptive module that ensures first-line defense against external toxics, irritants, antigens, and pathogens. The underlying mechanisms of host protection encompass multiple physical, chemical, and immune pathways. In the lung, inhaled agents continually challenge the airway epithelial barrier, which is altered in chronic diseases such as chronic obstructive pulmonary disease, asthma, cystic fibrosis, or pulmonary fibrosis. In this review, we describe the epithelial barrier abnormalities that are observed in such disorders and summarize current knowledge on the mechanisms driving impaired barrier function, which could represent targets of future therapeutic approaches.
Collapse
Affiliation(s)
- François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology and Lung Transplant, Centre Hospitalier Universitaire UCL Namur, Yvoir, Belgium
| | - Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| |
Collapse
|
44
|
Wang S, Jiang Z, Li L, Zhang J, Zhang C, Shao C. Ameliorative effects of eosinophil deficiency on immune response, endoplasmic reticulum stress, apoptosis, and autophagy in fungus-induced allergic lung inflammation. Respir Res 2021; 22:173. [PMID: 34098934 PMCID: PMC8186139 DOI: 10.1186/s12931-021-01770-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 06/04/2021] [Indexed: 01/21/2023] Open
Abstract
Background Respiratory fungal exposure is known to be associated with various allergic pulmonary disorders. Eosinophils have been implicated in tissue homeostasis of allergic inflammation as both destructive effector cells and immune regulators. What contributions eosinophils have in Aspergillus fumigatus (Af)-induced allergic lung inflammation is worthy of investigating. Methods We established the Af-exposed animal asthmatic model using eosinophil-deficient mice, ∆dblGATA1 mice. Airway inflammation was assessed by histopathological examination and total cell count of bronchoalveolar lavage fluid (BALF). The protein level in BALF and lung mRNA level of type 2 cytokines IL-4, IL-5, and IL-13 were detected by ELISA and qRT-PCR. We further studied the involvement of endoplasmic reticulum (ER) stress, apoptosis, and autophagy by western blots, qRT-PCR, immunofluorescence, TUNEL, or immunohistochemistry. RNA-Seq analysis was utilized to analyze the whole transcriptome of Af-exposed ∆dblGATA1 mice. Results Hematoxylin and eosin (HE) staining and periodic acid–Schiff staining (PAS) showed that airway inflammation and mucus production were alleviated in Af-challenged ∆dblGATA1 mice compared with wild-type controls. The protein and mRNA expressions of IL-4, IL-5, and IL-13 were reduced in the BALF and lung tissues in Af-exposed ∆dblGATA1 mice. The results demonstrated that the significantly increased ER stress markers (GRP78 and CHOP) and apoptosis executioner caspase proteases (cleaved caspase-3 and cleaved caspase-7) in Af-exposed wild-type mice were all downregulated remarkably in the lungs of ∆dblGATA1 mice with Af challenge. In addition, the lung autophagy in Af-exposed ∆dblGATA1 mice was found elevated partially, manifesting as higher expression of LC3-II/LC3-I and beclin1, lower p62, and downregulated Akt/mTOR pathway compared with Af-exposed wild-type mice. Additionally, lung RNA-seq analysis of Af-exposed ∆dblGATA1 mice showed that biological processes about chemotaxis of lymphocytes, neutrophils, or eosinophils were enriched but without statistical significance. Conclusions In summary, eosinophils play an essential role in the pathogenesis of Af-exposed allergic lung inflammation, whose deficiency may have relation to the attenuation of type 2 immune response, alleviation of ER stress and apoptosis, and increase of autophagy. These findings suggest that anti-eosinophils therapy may provide a promising direction for fungal-induced allergic pulmonary diseases.
Collapse
Affiliation(s)
- Sijiao Wang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liyang Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Changzhou Shao
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Department of Pulmonary Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 361015, China.
| |
Collapse
|
45
|
Duan XY, Sun Y, Zhao ZF, Shi YQ, Ma XY, Tao L, Liu MW. Baicalin attenuates LPS-induced alveolar type II epithelial cell A549 injury by attenuation of the FSTL1 signaling pathway via increasing miR-200b-3p expression. Innate Immun 2021; 27:294-312. [PMID: 34000873 PMCID: PMC8186156 DOI: 10.1177/17534259211013887] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In China, baicalin is the main active component of Scutellaria baicalensis, which has been used in the treatment of inflammation-related diseases, such as inflammation-induced acute lung injury. However, its specific mechanism remains unclear. This study examined the protective effect of baicalin on LPS-induced inflammation injury of alveolar epithelial cell line A549 and explored its protective mechanism. Compared with the LPS-induced group, the proliferation inhibition rates of alveolar type II epithelial cell line A549 intervened by different concentrations of baicalin decreased significantly, as did the levels of inflammatory factors IL-6, IL-1β, prostaglandin 2 and TNF-α in the supernatant. The expression levels of inflammatory proteins inducible NO synthase (iNOS), NF-κB65, phosphorylated ERK (p-ERK1/2), and phosphorylated c-Jun N-terminal kinase (p-JNK1) significantly decreased, as did the protein expression of follistatin-like protein 1 (FSTL1). In contrast, expression of miR-200b-3p significantly increased in a dose-dependent manner. These results suggested that baicalin could significantly inhibit the expression of inflammation-related proteins and improve LPS-induced inflammatory injury in alveolar type II epithelial cells. The mechanism may be related to the inhibition of ERK/JNK inflammatory pathway activation by increasing the expression of miR-200b-3p. Thus, FSTL1 is the regulatory target of miR-200b-3p.
Collapse
Affiliation(s)
- Xin-Ya Duan
- Department of Tuberculosis Diseases, Third People's Hospital of Kunming City, China
| | - Yang Sun
- Department of Nephrology, The Sixth Affiliated Hospital of Kunming Medical University, China
| | - Zhu-Feng Zhao
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, China
| | - Yao-Qing Shi
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, China
| | - Xun-Yan Ma
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, China
| | - Li Tao
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, China
| | - Ming-Wei Liu
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, China
| |
Collapse
|
46
|
Pontemezzo E, Foglio E, Vernucci E, Magenta A, D’Agostino M, Sileno S, Astanina E, Bussolino F, Pellegrini L, Germani A, Russo MA, Limana F. miR-200c-3p Regulates Epitelial-to-Mesenchymal Transition in Epicardial Mesothelial Cells by Targeting Epicardial Follistatin-Related Protein 1. Int J Mol Sci 2021; 22:4971. [PMID: 34067060 PMCID: PMC8125323 DOI: 10.3390/ijms22094971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Recent findings suggest that epithelial to mesenchymal transition (EMT), a key step during heart development, is involved in cardiac tissue repair following myocardial infarction (MI). MicroRNAs (miRNAs) act as key regulators in EMT processes; however, the mechanisms by which miRNAs target epicardial EMT remain largely unknown. Here, by using an in vitro model of epicardial EMT, we investigated the role of miRNAs as regulators of this process and their potential targets. EMT was induced in murine epicardial-mesothelial cells (EMCs) through TGF β1 treatment for 48, 72, and 96 h as indicated by the expression of EMT-related genes by qRT-PCR, WB, and immunofluorescence. Further, enhanced expression of stemness genes was also detected. Among several EMT-related miRNAs, miR-200c-3p expression resulted as the most strongly suppressed. Interestingly, we also found a significant upregulation of Follistatin-related protein 1 (FSTL1), a miR-200c predicted target already identified as a potent cardiogenic factor produced by epicardial cells that promotes regeneration following MI. Dual-luciferase reporter assay demonstrated that miR-200c-3p directly targeted the 3'-untranslated region of FSTL1 in EMCs. Consistently, WB analysis showed that knockdown of miR-200c-3p significantly increased FSTL1 expression, whereas overexpression of miR-200c-3p counteracted TGF β1-mediated FSTL1 upregulation. Importantly, FSTL1 silencing maintained epithelial features in EMCs, despite EMT induction by TGF β1, and attenuated EMT-associated traits, including migration and stemness. In conclusion, epicardial FSTL1, an important cardiogenic factor in its secreted form, induces EMT, stemness, and migration of EMCs in a miR-200c-3p dependent pathway.
Collapse
Affiliation(s)
- Elena Pontemezzo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.P.); (E.F.); (E.V.); (L.P.)
| | - Eleonora Foglio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.P.); (E.F.); (E.V.); (L.P.)
| | - Enza Vernucci
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.P.); (E.F.); (E.V.); (L.P.)
| | - Alessandra Magenta
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (A.M.); (M.D.); (S.S.); (A.G.)
- Institute of Translational Pharmacology (IFT), Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Marco D’Agostino
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (A.M.); (M.D.); (S.S.); (A.G.)
| | - Sara Sileno
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (A.M.); (M.D.); (S.S.); (A.G.)
| | - Elena Astanina
- Department of Oncology, University of Turin, 10060 Candiolo, Italy; (E.A.); (F.B.)
- Candiolo Cancer Institute-FPO-IRCCS, 10060 Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Turin, 10060 Candiolo, Italy; (E.A.); (F.B.)
- Candiolo Cancer Institute-FPO-IRCCS, 10060 Candiolo, Italy
| | - Laura Pellegrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.P.); (E.F.); (E.V.); (L.P.)
| | - Antonia Germani
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (A.M.); (M.D.); (S.S.); (A.G.)
| | - Matteo Antonio Russo
- IRCCS San Raffaele Pisana and MEBIC Consortium, 00166 Rome, Italy;
- Department of Human Science and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Federica Limana
- Department of Human Science and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| |
Collapse
|
47
|
Pharmacogenetic Polygenic Risk Score for Bronchodilator Response in Children and Adolescents with Asthma: Proof-of-Concept. J Pers Med 2021; 11:jpm11040319. [PMID: 33923870 PMCID: PMC8073919 DOI: 10.3390/jpm11040319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 11/19/2022] Open
Abstract
Genome-wide association studies (GWAS) of response to asthma medications have primarily focused on Caucasian populations, with findings that may not be generalizable to minority populations. We derived a polygenic risk score (PRS) for response to albuterol as measured by bronchodilator response (BDR), and examined the PRS in a cohort of Hispanic school-aged children with asthma. We leveraged a published GWAS of BDR to identify relevant genetic variants, and ranked the top variants according to their Combined Annotation Dependent Depletion (CADD) scores. Variants with CADD scores greater than 10 were used to compute the PRS. Once we derived the PRS, we determined the association of the PRS with BDR in a cohort of Hispanic children with asthma (the Genetics of Asthma in Costa Rica Study (GACRS)) in adjusted linear regression models. Mean BDR in GACRS participants was5.6% with a standard deviation of 10.2%. We observed a 0.63% decrease in BDR in response to albuterol for a standard deviation increase in the PRS (p = 0.05). We also observed decreased odds of a BDR response at or above the 12% threshold for a one standard deviation increase in the PRS (OR = 0.80 (95% CI 0.67 to 0.95)). Our findings show that combining variants from a pharmacogenetic GWAS into a PRS may be useful for predicting medication response in asthma.
Collapse
|
48
|
Wang WL, Luo XM, Zhang Q, Zhu HQ, Chen GQ, Zhou Q. The lncRNA PVT1/miR-590-5p/FSTL1 axis modulates the proliferation and migration of airway smooth muscle cells in asthma. Autoimmunity 2021; 54:138-147. [PMID: 33825599 DOI: 10.1080/08916934.2021.1897977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Asthma is a prevalent chronic inflammatory airway disease that is characterised by airway remodelling and airway hyperresponsiveness. Abnormal proliferation and migration of airway smooth muscle cells (ASMCs) contribute to airway remodelling in asthma. However, the molecular mechanism underlying an increased ASMC mass in asthma remains elusive. Herein, we aimed at investigating the regulation of lncRNA PVT1 on ASMCs and focussing on the mechanism in the proliferation and migration. METHODS Expression levels of lncRNA PVT1 and miR-590-5p in the serum collected from 24 children with asthma and 10 control children were determined by qRT-PCR. ASMCs proliferation and migration prior to and post platelet-derived growth factor subunit B (PDGF-BB) stimulation were examined by CCK-8 test and transwell assay. Dual-luciferase reporter assay was performed to determine miR-590-5p interaction with lncRNA PVT1 and follistatin-like 1 (FSTL1). Expression of lncRNA PVT1, miR-590-5p, FSTL1, C-Myc, cyclin D1, and cyclin-dependent kinase 1 (CDK1) was tested by quantitative real-time PCR (qRT-PCR) and immunoblotting analysis. RESULTS The expression level of lncRNA PVT1 was higher but the expression level of miR-590-5p was lower in the serum of children with asthma than in control children. The expression level of lncRNA PVT1 was negatively correlated with the expression level of miR-590-5p in asthma. LncRNA PVT1 was upregulated upon PDGF-BB stimulation. LncRNA PVT1 knockdown by its specific shRNA repressed PDGF-BB-induced promotion of proliferation and migration in ASMCs and triggered an elevated miR-590-5p along with declined C-Myc, cyclin D1, and CDK1. The effects of lncRNA PVT1 knockdown on PDGF-BB-induced ASMCs were lost upon miR-590-5p inhibition. MiR-590-5p targeted FSTL1 gene and declined its expression, thus suppressing ASMC proliferation and migration following PDGF-BB stimulation and downregulating C-Myc, cyclin D1, and CDK1 expressions. The effects of miR-590-5p on PDGF-BB-induced ASMCs were lost upon FSTL1 overexpression. CONCLUSION These results support the notion that the lncRNA PVT1/miR-590-5p/FSTL1 axis modulates ASMCs proliferation and migration following PDGF-BB stimulation, providing a potential therapeutic target to attenuate airway remodelling in asthma.
Collapse
Affiliation(s)
- Wen-Lan Wang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Xiao-Ming Luo
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Hai-Qiao Zhu
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Guo-Qing Chen
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhou
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| |
Collapse
|
49
|
Yan Q, Forno E, Herrera-Luis E, Pino-Yanes M, Qi C, Rios R, Han YY, Kim S, Oh S, Acosta-Pérez E, Zhang R, Hu D, Eng C, Huntsman S, Avila L, Boutaoui N, Cloutier MM, Soto-Quiros ME, Xu CJ, Weiss ST, Lasky-Su J, Kiedrowski MR, Figueiredo C, Bomberger J, Barreto ML, Canino G, Chen W, Koppelman GH, Burchard EG, Celedón JC. A genome-wide association study of severe asthma exacerbations in Latino children and adolescents. Eur Respir J 2021; 57:2002693. [PMID: 33093117 PMCID: PMC8026735 DOI: 10.1183/13993003.02693-2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022]
Abstract
Severe asthma exacerbations are a major cause of school absences and healthcare costs in children, particularly those in high-risk racial/ethnic groups.To identify susceptibility genes for severe asthma exacerbations in Latino children and adolescents, we conducted a meta-analysis of genome-wide association studies (GWAS) in 4010 Latino youth with asthma in four independent cohorts, including 1693 Puerto Ricans, 1019 Costa Ricans, 640 Mexicans, 256 Brazilians and 402 members of other Latino subgroups. We then conducted methylation quantitative trait locus, expression quantitative trait locus and expression quantitative trait methylation analyses to assess whether the top single nucleotide polymorphism (SNP) in the meta-analysis is linked to DNA methylation and gene expression in nasal (airway) epithelium in separate cohorts of Puerto Rican and Dutch children and adolescents.In the meta-analysis of GWAS, an SNP in FLJ22447 (rs2253681) was significantly associated with 1.55 increased odds of severe asthma exacerbation (95% CI 1.34-1.79, p=6.3×10-9). This SNP was significantly associated with DNA methylation of a CpG site (cg25024579) at the FLJ22447 locus, which was in turn associated with increased expression of KCNJ2-AS1 in nasal airway epithelium from Puerto Rican children and adolescents (β=0.10, p=2.18×10-7).SNP rs2253681 was significantly associated with both DNA methylation of a cis-CpG in FLJ22447 and severe asthma exacerbations in Latino youth. This may be partly explained by changes in airway epithelial expression of a gene recently implicated in atopic asthma in Puerto Rican children and adolescents (KCNJ2-AS1).
Collapse
Affiliation(s)
- Qi Yan
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Shared first authors
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Shared first authors
| | - Esther Herrera-Luis
- Genomics and Health Group, Dept of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Spain
- Shared first authors
| | - Maria Pino-Yanes
- Genomics and Health Group, Dept of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Cancan Qi
- Dept of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Raimon Rios
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Soyeon Kim
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sam Oh
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Edna Acosta-Pérez
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Rong Zhang
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donglei Hu
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Celeste Eng
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Scott Huntsman
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lydiana Avila
- Dept of Pediatrics, Hospital Nacional de Niños, San José, Costa Rica
| | - Nadia Boutaoui
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Cheng-Jian Xu
- CiiM and TWINCORE, joint ventures between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Dept of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan R Kiedrowski
- Dept of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Camila Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Jennifer Bomberger
- Dept of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mauricio L Barreto
- Instituto de Saúde Coletiva, Federal University of Bahia, Salvador, Brazil
| | - Glorisa Canino
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Wei Chen
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gerard H Koppelman
- Dept of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Esteban G Burchard
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
- Shared senior authors
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh Medical Centre, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Shared senior authors
| |
Collapse
|
50
|
Lu X, Li R, Yan X. Airway hyperresponsiveness development and the toxicity of PM2.5. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:6374-6391. [PMID: 33394441 DOI: 10.1007/s11356-020-12051-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/10/2020] [Indexed: 04/16/2023]
Abstract
Airway hyperresponsiveness (AHR) is characterized by excessive bronchoconstriction in response to nonspecific stimuli, thereby leading to airway stenosis and increased airway resistance. AHR is recognized as a key characteristic of asthma and is associated with significant morbidity. At present, many studies on the molecular mechanisms of AHR have mainly focused on the imbalance in Th1/Th2 cell function and the abnormal contraction of airway smooth muscle cells. However, the specific mechanisms of AHR remain unclear and need to be systematically elaborated. In addition, the effect of air pollution on the respiratory system has become a worldwide concern. To date, numerous studies have indicated that certain concentrations of fine particulate matter (PM2.5) can increase airway responsiveness and induce acute exacerbation of asthma. Of note, the concentration of PM2.5 does correlate with the degree of AHR. Numerous studies exploring the toxicity of PM2.5 have mainly focused on the inflammatory response, oxidative stress, genotoxicity, apoptosis, autophagy, and so on. However, there have been few reviews systematically elaborating the molecular mechanisms by which PM2.5 induces AHR. The present review separately sheds light on the underlying molecular mechanisms of AHR and PM2.5-induced AHR.
Collapse
Affiliation(s)
- Xi Lu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, China
| | - Rongqin Li
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, China
| | - Xixin Yan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|