1
|
Lyu Y, Feng W, Song J, Wang C, Fu Y, Zhao B, Meng Y. Zedoarondiol inhibits human bronchial smooth muscle cell proliferation through the CAV-1/PDGF signalling pathway. Sci Rep 2024; 14:13145. [PMID: 38849430 PMCID: PMC11161633 DOI: 10.1038/s41598-024-63970-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Airway remodelling in lung diseases can be treated by inhibiting excessive smooth muscle cell proliferation. Zedoarondiol (Zed) is a natural compound isolated from the Chinese herb Curcuma longa. The caveolin-1 (CAV-1) is widely expressed in lung cells and plays a key role in platelet-derived growth factor (PDGF) signalling and cell proliferation. This study aims to investigate the effect of Zed on human bronchial smooth muscle cell (HBSMC) proliferation and explore its potential molecular mechanisms. We assessed the effect of Zed on the proliferation of PDGF-stimulated HBSMCs and performed proteomic analysis to identify potential molecular targets and pathways. CAV1 siRNA was used to validate our findings in vitro. In PDGF-stimulated HBSMCs, Zed significantly inhibited excessive proliferation of HBSMCs. Proteomic analysis of zedoarondiol-treated HBSMCs revealed significant enrichment of differentially expressed proteins in cell proliferation-related pathways and biological processes. Zed inhibition of HBSMC proliferation was associated with upregulation of CAV1, regulation of the CAV-1/PDGF pathway and inhibition of MAPK and PI3K/AKT signalling pathway activation. Treatment of HBSMCs with CAV1 siRNA partly reversed the inhibitory effect of Zed on HBSMC proliferation. Thus, this study reveals that zedoarondiol potently inhibits HBSMC proliferation by upregulating CAV-1 expression, highlighting its potential value in airway remodelling and related diseases.
Collapse
Affiliation(s)
- Yinglan Lyu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wandi Feng
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jingze Song
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, 11 North 3Rd Ring Eastern Road, Beijing, 100029, China
| | - Yu Fu
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, 11 North 3Rd Ring Eastern Road, Beijing, 100029, China
| | - Yanyan Meng
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, 11 North 3Rd Ring Eastern Road, Beijing, 100029, China.
| |
Collapse
|
2
|
Saunders RM, Biddle M, Amrani Y, Brightling CE. Stressed out - The role of oxidative stress in airway smooth muscle dysfunction in asthma and COPD. Free Radic Biol Med 2022; 185:97-119. [PMID: 35472411 DOI: 10.1016/j.freeradbiomed.2022.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022]
Abstract
The airway smooth muscle (ASM) surrounding the airways is dysfunctional in both asthma and chronic obstructive pulmonary disease (COPD), exhibiting; increased contraction, increased mass, increased inflammatory mediator release and decreased corticosteroid responsiveness. Due to this dysfunction, ASM is a key contributor to symptoms in patients that remain symptomatic despite optimal provision of currently available treatments. There is a significant body of research investigating the effects of oxidative stress/ROS on ASM behaviour, falling into the following categories; cigarette smoke and associated compounds, air pollutants, aero-allergens, asthma and COPD relevant mediators, and the anti-oxidant Nrf2/HO-1 signalling pathway. However, despite a number of recent reviews addressing the role of oxidative stress/ROS in asthma and COPD, the potential contribution of oxidative stress/ROS-related ASM dysfunction to asthma and COPD pathophysiology has not been comprehensively reviewed. We provide a thorough review of studies that have used primary airway, bronchial or tracheal smooth muscle cells to investigate the role of oxidative stress/ROS in ASM dysfunction and consider how they could contribute to the pathophysiology of asthma and COPD. We summarise the current state of play with regards to clinical trials/development of agents targeting oxidative stress and associated limitations, and the adverse effects of oxidative stress on the efficacy of current therapies, with reference to ASM related studies where appropriate. We also identify limitations in the current knowledge of the role of oxidative stress/ROS in ASM dysfunction and identify areas for future research.
Collapse
Affiliation(s)
- Ruth M Saunders
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK.
| | - Michael Biddle
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Yassine Amrani
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Christopher E Brightling
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
3
|
Wang X, Xu R, Chi D, Dai C, Sheng M. Role of NEAT1/MiR-9-5p/SLC26A2 Pathway on Human Airway Smooth Muscle Cell. Yonsei Med J 2021; 62:858-867. [PMID: 34427073 PMCID: PMC8382724 DOI: 10.3349/ymj.2021.62.9.858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Asthma is a serious inflammatory disease of the respiratory system in which airway smooth muscle cells (ASMCs) play a key role. This study aimed to investigate the expression of SLC26A2 in human ASMCs (HASMCs) and the regulatory mechanism of SLC26A2 in the proliferation and inflammatory factor production of HASMCs. MATERIALS AND METHODS We obtained the asthma-associated differential mRNA SLC26A2 by bioinformatics analysis in childhood acute asthma samples. To investigate its role in airway inflammation and airway remodeling, we treated HASMCs with platelet-derived growth factor (PDGF) in an in vitro model and determined SLC26A2 expression in cells using western blotting. Cell proliferation was detected by MTT and EdU assays, and cell contractile phenotype marker proteins were measured. Cell migration and production of inflammatory factors were determined by Transwell and ELISA assays. Additionally, the upstream regulatory miRNA and LncRNA of SLC26A2 were identified by bioinformatics, luciferase reporter gene, and RIP analyses. RESULTS SLC26A2 was significantly upregulated in bioinformatics analysis of pediatric asthma-related sample. PDGF treatment up-regulated SLC26A2 expression in HASMCs, whereas the knockdown of SLC26A2 inhibited PDGF-stimulated proliferation, migration, and production of inflammatory factors, and enhanced the expression of cell contractile phenotype marker proteins in HASMCs. Luciferase reporter and RIP experiments validated that NEAT1 targeted miR-9-5p to regulate SLC26A2, thereby influencing the biological function of PDGF-induced HASMCs. CONCLUSION These findings indicate that NEAT1-mediated miR-9-5p targeting of SLC26A2 inhibits the PDGF-induced proliferation and production of inflammatory factors in HASMCs. These findings highlight potential therapeutic targets for asthma and airway inflammation.
Collapse
Affiliation(s)
- Xiangying Wang
- Department of Rheumatology and Immunology in Children, Hangzhou Children's Hospital, Hangzhou, China
| | - Ruju Xu
- Department of Rheumatology and Immunology in Children, Hangzhou Children's Hospital, Hangzhou, China
| | - Di Chi
- Department of Rheumatology and Immunology in Children, Hangzhou Children's Hospital, Hangzhou, China
| | - Chufeng Dai
- Department of Rheumatology and Immunology in Children, Hangzhou Children's Hospital, Hangzhou, China
| | - Meiling Sheng
- Department of Rheumatology and Immunology in Children, Hangzhou Children's Hospital, Hangzhou, China.
| |
Collapse
|
4
|
Abstract
Receptor for advanced glycation end products (RAGE) is an immunoglobulin-like receptor present on cell surface. RAGE binds to an array of structurally diverse ligands, acts as a pattern recognition receptor (PRR) and is expressed on cells of different origin performing different functions. RAGE ligation leads to the initiation of a cascade of signaling events and is implicated in diseases, such as inflammation, cancer, diabetes, vascular dysfunctions, retinopathy, and neurodegenerative diseases. Because of the significant involvement of RAGE in the progression of numerous diseases, RAGE signaling has been targeted through use of inhibitors and anti-RAGE antibodies as a treatment strategy and therapy. Here in this review, we have summarized the physical and physiological aspects of RAGE biology in mammalian system and the importance of targeting this molecule in the treatment of various RAGE mediated pathologies. Highlights Receptor for advanced glycation end products (RAGE) is a member of immunoglobulin superfamily of receptors and involved in many pathophysiological conditions. RAGE ligation with its ligands leads to initiation of distinct signaling cascades and activation of numerous transcription factors. Targeting RAGE signaling through inhibitors and anti-RAGE antibodies can be promising treatment strategy.
Collapse
Affiliation(s)
- Nitish Jangde
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India.,Manipal Academy of Higher Education, Manipal, India
| | - Rashmi Ray
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India
| | - Vivek Rai
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
5
|
Gap-134, a Connexin43 activator, prevents age-related development of ventricular fibrosis in Scn5a +/- mice. Pharmacol Res 2020; 159:104922. [PMID: 32464326 DOI: 10.1016/j.phrs.2020.104922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/30/2020] [Accepted: 05/10/2020] [Indexed: 01/05/2023]
Abstract
Down-regulation of Connexin43 (Cx43) has often been associated with the development of cardiac fibrosis. We showed previously that Scn5a heterozygous knockout mice (Scn5a+/-), which mimic familial progressive cardiac conduction defect, exhibit an age-dependent decrease of Cx43 expression and phosphorylation concomitantly with activation of TGF-β pathway and fibrosis development in the myocardium between 45 and 60 weeks of age. The aim of this study was to investigate whether Gap-134 prevents Cx43 down-regulation with age and fibrosis development in Scn5a+/- mice. We observed in 60-week-old Scn5a+/- mouse heart a Cx43 expression and localization remodeling correlated with fibrosis. Chronic administration of a potent and selective gap junction modifier, Gap-134 (danegaptide), between 45 and 60 weeks, increased Cx43 expression and phosphorylation on serine 368 and prevented Cx43 delocalization. Furthermore, we found that Gap-134 prevented fibrosis despite the persistence of the conduction defects and the TGF-β canonical pathway activation. In conclusion, the present study demonstrates that the age-dependent decrease of Cx43 expression is involved in the ventricular fibrotic process occurring in Scn5a+/- mice. Finally, our study suggests that gap junction modifier, such as Gap-134, could be an effective anti-fibrotic agent in the context of age-dependent fibrosis in progressive cardiac conduction disease.
Collapse
|
6
|
Kardas G, Daszyńska-Kardas A, Marynowski M, Brząkalska O, Kuna P, Panek M. Role of Platelet-Derived Growth Factor (PDGF) in Asthma as an Immunoregulatory Factor Mediating Airway Remodeling and Possible Pharmacological Target. Front Pharmacol 2020; 11:47. [PMID: 32116722 PMCID: PMC7033439 DOI: 10.3389/fphar.2020.00047] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
Asthma is a chronic and heterogenic disease of the respiratory system, one of the most common lung diseases worldwide. The underlying pathologies, which are chronic inflammatory process and airway remodeling (AR), are mediated by numerous cells and cytokines. Particularly interesting in this field is the platelet-derived growth factor (PDGF), one of the members of the human growth factor family. In this article, the authors analyze the available data on the role of PDGF in asthma in experimental models and in human research. PDGF is expressed in airway by various cells contributing to asthma pathogenesis—mast cells, eosinophils, and airway epithelial cells. Research confirms the thesis that this factor is also secreted by these cells in the course of asthma. The main effects of PDGF on bronchi are the proliferation of airway smooth muscle (ASM) cells, migration of ASM cells into the epithelium and enhanced collagen synthesis by lung fibroblasts. The importance of AR in asthma is well recognized and new therapies should also aim to manage it, possibly targeting PDGFRs. Further studies on new and already existing drugs, mediating the PDGF signaling and related to asthma are necessary. Several promising drugs from the tyrosine kinase inhibitors group, including nilotinib, imatinib masitinib, and sunitinib, are currently being clinically tested and other molecules are likely to emerge in this field.
Collapse
Affiliation(s)
- Grzegorz Kardas
- Clinic of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Łódź, Poland
| | | | - Mateusz Marynowski
- Clinic of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Łódź, Poland
| | - Oliwia Brząkalska
- Clinic of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Łódź, Poland
| | - Piotr Kuna
- Clinic of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Łódź, Poland
| | - Michał Panek
- Clinic of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
7
|
Qu D, Ling Z, Tan X, Chen Y, Huang Q, Li M, Liu T, Hou C, Chen Y. High mobility group protein B1 (HMGB1) interacts with receptor for advanced glycation end products (RAGE) to promote airway smooth muscle cell proliferation through ERK and NF- κB pathways. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3268-3278. [PMID: 31934170 PMCID: PMC6949814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND High-mobility graoup box protein 1 (HMGB1) has been shown to mediate a wide range of pathologic responses by interacting with RAGE (receptor for advanced glycation endproducts) and TLRs (Toll-like receptors). Our previous study showed that HMGB1 has been involved in pathogenesis of airway remodeling in an allergen-induced chronic mice asthma model. Increased airway smooth muscle (ASM) mass is a vital feature of airway remodeling. OBJECTIVE To evaluate the effect of HMGB1 on proliferation of ASMs and the underlying mechanisms. METHODS Rat airway smooth muscle cells (RASMs) were obtained by primary explant techniques. We investigated the effect of HMGB1 on the proliferation of RASMs. To identify which receptors and signaling pathways be involved in proliferation of RASMs, we performed western blot and CCK-8 assay by specific receptor blockade and inhibition of MAPK (p38, JNK and ERK) and NF-κB signaling pathways. RESULTS HMGB1 stimulated RASMs proliferation in a dose- and time-dependent manner and also increased proliferating cell nuclear antigen (PCNA) and RAGE expression of RASMs. The inhibitor of RAGE, but not TLR2 and TLR4, reversed HMGB1-induced RASM proliferation and PCNA expression. Incubation of RASMs with HMGB1 caused a rapid increase in P65 and ERK phosphorylation. RASM proliferation and PCNA expression toward HMGB1 were significantly inhibited by the inhibitors of ERK and NF-κB. CONCLUSION HMGB1 induces proliferation of RASMs through a RAGE-dependent activation of ERK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Dongming Qu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Zhougui Ling
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical UniversityLiuzhou 545005, China
| | - Xiaoyu Tan
- Department of Respiratory Medicine, The Second Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Qinghua Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Mengze Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Tangjuan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Changchun Hou
- Department of Respiratory Medicine, The Second Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Yiqiang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, China
| |
Collapse
|
8
|
刘 慧, 刘 嘉, 彭 美, 黎 玉, 李 春. [Effect of acupuncture on TGF-β1/Smads pathway in mice with airway remodeling mic]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1372-1377. [PMID: 30514688 PMCID: PMC6744113 DOI: 10.12122/j.issn.1673-4254.2018.11.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of acupuncture on TGF-β1/Smads signaling pathway in the lung tissue of mice with airway remodeling. METHODS Thirty specific pathogen-free mice were randomly divided into blank group, model group and acupuncture group (n=10). Mouse models of asthma were established in the model group and the acupuncture group, and the mice in the latter group received 7 acupuncture therapies (at bilateral Fei Shu, Da Zhui and Zu Sanli, 20 min each time) every other day, starting on the 10th day after the modeling. At 24 h after the last acupuncture, the mice were subjected to inhalation of 1% OVA for 3 days, and 24 h after the last challenge, the mice were given methacholine chloride (Mch) inhalation at different concentrations for measurement of lung resistance using a noninvasive stroke volume meter. HE staining was used to observe the pathological changes in the lung tissues, and TGF-β1 levels in the the bronchoalveolar lavage fluid (BALF) and serum were detected using ELISA; Western blotting was used to detect the differential protein expressions in the airway smooth muscles between the two groups. The airway smooth muscle cells were isolated from the mice in the acupuncture group and treated with a TGF- β1 inhibitor (LY2157299), and the relative expressions of type-Ⅰ and Smads proteins were detected using Western blotting. RESULTS The mice in the model showed obvious tracheal fistula with airway pathologies including lumen narrowing, bronchial mucosa thickening, dissociation of the epithelial cells, and thickening of the alveolar septum and airway smooth muscles. These pathological changes were obviously milder in the acupuncture group. The asthmatic mice exhibited significantly increased lung resistance in positive correlation with Mch concentration. Serum TGF-β1 level was significantly elevated in asthmatic mice (P < 0.05); TGF-β1 levels in the serum and BALF were significantly lower in the acupuncture group than in the model group (P < 0.05). In the model group, the expressions of α-SMA, TGF-β1 and Smads in the airway smooth muscles were significantly higher than those in the other two groups (both P < 0.05). In cultured airway smooth muscle cells, the expressions of type-Ⅰ and Smads were significantly higher in cells treated with LY2157299 than in the control cells (P>0.05). CONCLUSIONS Acupuncture can inhibit airway remodeling by inhibiting the expression of airway TGF-β1 and down-regulating the expression of Smads and α-SMA to reduce airway inflammatory response. Airway expressions of type-Ⅰ and Smads proteins remain high after inhibiting TGF-β1. Acupuncture may control asthma progression through the TGF-β1/Smads pathway.
Collapse
Affiliation(s)
- 慧慧 刘
- 广州中医药大学第一附属医院 麻醉科,广东 广州 510405Department of Anesthesiology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - 嘉羿 刘
- 中山大学附属孙逸仙医院麻醉科,广东 广州 510120Department of Anesthesiology, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Guangzhou 510120, China
| | - 美玉 彭
- 广州中医药大学第一附属医院 麻醉科,广东 广州 510405Department of Anesthesiology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - 玉辉 黎
- 广州中医药大学第一附属医院 麻醉科,广东 广州 510405Department of Anesthesiology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - 春桥 李
- 广州中医药大学第一附属医院 耳鼻喉科,广东 广州 510405Department of Otolaryngology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|