1
|
Kong Y, Wang Z, Yu H, Dong A, Song Y, Guo L, Zhu J, Sun L, Guo Y. YiQi GuBen formula alleviates airway inflammation and airway remodeling in OVA-induced asthma mice through TLR4/NF-κB signaling pathway. J Pharm Pharmacol 2024; 76:1028-1037. [PMID: 38824434 DOI: 10.1093/jpp/rgae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND We aim to investigate the effect of YiQi GuBen formula (YQGB) on airway inflammation and airway remodeling in the ovalbumin (OVA)-induced asthma model to further explore the potential mechanisms of YQGB in treating allergic asthma. METHODS Mice were divided into five groups randomly (n = 10): the control group, OVA group, OVA + Dex (0.1 mg/kg) group, OVA + low-dose (1.1 g/kg) YQGB group, and OVA + high-dose (2.2 g/kg) YQGB group. Inflammatory cell count and IgE were detected in bronchoalveolar lavage fluid (BALF). Lung tissue histopathology was observed by using H&E, PAS, Masson, and immunohistochemistry staining. qRT-PCR and western blot were applied to analyze key genes and proteins associated with TLR4 and NF-κB signaling pathways. RESULTS In OVA-induced asthma mice, YQGB decreased eosinophils and IgE in BALF. YQGB alleviated the OVA-induced inflammatory infiltration and declined IL-4, IL-5, IL-13, Eotaxin, ECP, GM-CSF, LTC4, and LTD4. YQGB attenuated the OVA-induced goblet cell metaplasia and mucus hypersecretion. YQGB mitigated the OVA-induced subepithelial fibrosis and lowered TGF-β1, E-Cadherin, Vimentin, and Fibronectin. YQGB ameliorated the OVA-induced airway smooth muscle thickening and lessened α-SMA and PDGF levels. YQGB reduced the expression of TLR4, MyD88, TRAF6, IκBα, and p65 mRNAs, and IκBα and p-p65 protein levels were also reduced. CONCLUSION YQGB exhibits the anti-asthma effect by reducing airway inflammation and airway remodeling through suppressing TLR4/NF-κB signaling pathway, and is worth promoting clinically.
Collapse
Affiliation(s)
- Yibu Kong
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Zhongtian Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Hongjun Yu
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Aiai Dong
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Yongfu Song
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Lei Guo
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Jinpu Zhu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Liping Sun
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Yinan Guo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| |
Collapse
|
2
|
Karunarathne WAHM, Lee KT, Choi YH, Kang CH, Lee MH, Kim SH, Kim GY. Investigating rutin as a potential transforming growth factor-β type I receptor antagonist for the inhibition of bleomycin-induced lung fibrosis. Biofactors 2024; 50:477-492. [PMID: 38006284 DOI: 10.1002/biof.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/26/2023] [Indexed: 11/27/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung condition characterized by the abnormal regulation of extracellular matrix (ECM) and epithelial-mesenchymal transition (EMT). In this study, we investigated the potential of rutin, a natural flavonoid, in attenuating transforming growth factor-β (TGF-β)-induced ECM regulation and EMT through the inhibition of the TGF-β type I receptor (TβRI)-mediated suppressor of mothers against decapentaplegic (SMAD) signaling pathway. We found that non-toxic concentrations of rutin attenuated TGF-β-induced ECM-related genes, including fibronectin, elastin, collagen 1 type 1, and TGF-β, as well as myoblast differentiation from MRC-5 lung fibroblast cells accompanied by the downregulation of α-smooth muscle actin. Rutin also inhibited TGF-β-induced EMT processes, such as wound healing, migration, and invasion by regulating EMT-related gene expression. Additionally, rutin attenuated bleomycin-induced lung fibrosis in mice, thus providing a potential therapeutic option for IPF. The molecular docking analyses in this study predict that rutin occludes the active site of TβRI and inhibits SMAD-mediated fibrotic signaling pathways in lung fibrosis. These findings highlight the potential of rutin as a promising anti-fibrotic prodrug for lung fibrosis and other TGF-β-induced fibrotic and cancer-related diseases; however, further studies are required to validate its safety and effectiveness in other experimental models.
Collapse
Affiliation(s)
| | - Kyoung Tae Lee
- Forest Bioresources Department, Forest Microbiology Division, National Institute of Forest Science, Suwon, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan, Republic of Korea
| | - Chang-Hee Kang
- Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Mi-Hwa Lee
- Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Sang-Hun Kim
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
3
|
Zhu JQ, Tian YY, Chan KL, Hu Z, Xu QQ, Lin ZX, Xian YF. Modified Qing-Zao-Jiu-Fei decoction attenuated pulmonary fibrosis induced by bleomycin in rats via modulating Nrf2/NF-κB and MAPKs pathways. Chin Med 2024; 19:10. [PMID: 38229198 DOI: 10.1186/s13020-024-00882-5] [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: 10/31/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Qing-Zao-Jiu-Fei Decoction (QZJFD) is a famous herbal formula commonly prescribed for the treatment of lung-related diseases in the ancient and modern times. Trichosanthis Fructus (TF) and Fritillariae Thunbergii Bulbus (FTB) are widely used for treatment of cough and pulmonary disease. In order to identify a more effective formula for treatment of pulmonary fibrosis, we intend to add TF and FTB in QZJFD to form a modified QZJFD (MQZJFD). In this study, we aims to explore MQZJFD as an innovative therapeutic agent for pulmonary fibrosis using bleomycin (BLM)-treated rats and to unravel the underlying molecular mechanisms. METHODS BLM was given to SD rats by intra-tracheal administration of a single dose of BLM (5 mg/kg). QZJFD (3 g/kg) and MQZJFD (1, 2 and 4 g/kg) was given intragastrically daily to rats for 14 days (from day 15 to 28) after BLM administration for 14 consecutive days. RESULTS MQZJFD was found to contain 0.29% of amygdalin, 0.020% of lutin, 0.077% of glycyrrhizic acid and 0.047% of chlorogenic acid. BLM treatment could induce collagen deposition in the lung tissues of rats, indicating that the pulmonary fibrosis rat model had been successfully established. MQZJFD have better effects than the original QZJFD in reducing the pulmonary structure damage and collagen deposition of rat lung fibrosis induced by BLM. MQZJFD could reduce the hydroxyproline content in lung tissues of BLM-treated rats. The biomarkers of fibrosis such as matrix metalloproteinase 9 (MMP9), collagen I and α-smooth muscle actin (α-SMA) were remarkably reduced after treatment with MQZJFD. MQZJFD also have anti-oxidant stress effects by inhibiting the level of malondialdehyde (MDA), but enhancing the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and the level of glutathione (GSH) in the lung tissues of BLM-treated rats. Moreover, the MQZJFD markedly suppressed the over expressions of p-p65/p65 and p-IκBα/IκBα, but upregulated the Nrf2. MQZJFD also suppressed the protein expressions of p-ERK1/2/ERK1/2, p-p38/p38 and p-JNK/JNK in the lung tissues of BLM-treated rats. CONCLUSIONS MQZJFD could improve the pulmonary fibrosis induced by BLM in rats via inhibiting the fibrosis and oxidative stress via suppressing the activation of NF-κB/Nrf2 and MAPKs pathways.
Collapse
Affiliation(s)
- Jia-Qian Zhu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
| | - Yuan-Yang Tian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
| | - Kam Leung Chan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
| | - Zhen Hu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
| | - Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China.
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
4
|
Liang D, Liu L, Zhao Y, Luo Z, He Y, Li Y, Tang S, Tang J, Chen N. Targeting extracellular matrix through phytochemicals: a promising approach of multi-step actions on the treatment and prevention of cancer. Front Pharmacol 2023; 14:1186712. [PMID: 37560476 PMCID: PMC10407561 DOI: 10.3389/fphar.2023.1186712] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Extracellular matrix (ECM) plays a pivotal and dynamic role in the construction of tumor microenvironment (TME), becoming the focus in cancer research and treatment. Multiple cell signaling in ECM remodeling contribute to uncontrolled proliferation, metastasis, immune evasion and drug resistance of cancer. Targeting trilogy of ECM remodeling could be a new strategy during the early-, middle-, advanced-stages of cancer and overcoming drug resistance. Currently nearly 60% of the alternative anticancer drugs are derived from natural products or active ingredients or structural analogs isolated from plants. According to the characteristics of ECM, this manuscript proposes three phases of whole-process management of cancer, including prevention of cancer development in the early stage of cancer (Phase I); prevent the metastasis of tumor in the middle stage of cancer (Phase II); provide a novel method in the use of immunotherapy for advanced cancer (Phase III), and present novel insights on the contribution of natural products use as innovative strategies to exert anticancer effects by targeting components in ECM. Herein, we focus on trilogy of ECM remodeling and the interaction among ECM, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), and sort out the intervention effects of natural products on the ECM and related targets in the tumor progression, provide a reference for the development of new drugs against tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Dan Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjie Zhao
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Zhenyi Luo
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Yadi He
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Liu H, Wang C. The genus Asarum: A review on phytochemistry, ethnopharmacology, toxicology and pharmacokinetics. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114642. [PMID: 34537281 DOI: 10.1016/j.jep.2021.114642] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/28/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In essentially every quadrant of the globe, many species of genus Asarum are used as a common herbal medicine and appear in many formulas or Kampo. Crude drug from several medicinal plants of genus Asarum (MA) known as Asari Radix et Rhizoma (ARR) has been proven to have the functions of dispelling cold, relieving pain, and reducing phlegm according to Traditional Chinese Medicine (TCM) theory for thousands of years. AIM OF THE STUDY This article reviews the ethnopharmacology, phytochemistry, pharmacology, toxicology and metabolic kinetics related research of genus Asarum to evaluate its ethnopharmacology use and future opportunities for research. MATERIALS AND METHODS Information on relevant studies of the genus Asarum was gathered via the Internet using Baidu Scholar, Web of Science, Elsevier, ResearchGate, ACS, Pudmed and Chinese National Knowledge Infrastructure (CNKI). Additionally, information was also obtained from some local books, PhD, MS's dissertations and Pharmacopeias. RESULTS The genus Asarum has played an important role in herbal treatment. At present, more than 277 compounds have been isolated or identified from genus Asarum. Among them, volatile oil and lignans are the major active constituents and important chemotaxonomic markers. Modern pharmacological studies indicated that genus Asarum and its active compounds possess a wide range of pharmacological effects, especially analgesic, anti-inflammatory, neuroprotective, cardiovascular protection, antitussive, immunosuppressive, anti-tumor, and microbicidal activities. CONCLUSIONS Based on this review, therapeutic potential of genus Asarum has been demonstrated with the pharmacological effects on inflammation, CNS, respiratory regulation, cardiovascular diseases, cancer and microbial infection. The available literature showed that the major activities of the genus Asarum can be attributed to the active lignans and essential oils. Further in-depth studies on the aspects of the genus for mechanism of actions, metabolism, pharmacokinetics, toxicology, drug interactions, and clinical trials are still limited, thereby intensive research and assessments should be performed.
Collapse
Affiliation(s)
- Hanze Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
6
|
Reichard A, Wanner N, Stuehr E, Alemagno M, Weiss K, Queisser K, Erzurum S, Asosingh K. Quantification of airway fibrosis in asthma by flow cytometry. Cytometry A 2018; 93:952-958. [PMID: 29659138 DOI: 10.1002/cyto.a.23373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/23/2018] [Accepted: 03/20/2018] [Indexed: 01/31/2023]
Abstract
Airway fibrosis is a prominent feature of asthma, contributing to the detrimental consequences of the disease. Fibrosis in the airway is the result of collagen deposition in the reticular lamina layer of the subepithelial tissue. Myofibroblasts are the leading cell type involved with this collagen deposition. Established methods of collagen deposition quantification present various issues, most importantly their inability to quantify current collagen biosynthesis occurring in airway myofibroblasts. Here, a novel method to quantify myofibroblast collagen expression in asthmatic lungs is described. Single cell suspensions of lungs harvested from C57BL/6 mice in a standard house dust mite model of asthma were employed to establish a flow cytometric method and compare collagen production in asthmatic and non-asthmatic lungs. Cells found to be CD45- αSMA+ , indicative of myofibroblasts, were gated, and median fluorescence intensity of the anti-collagen-I antibody labeling the cells was calculated. Lung myofibroblasts with no, medium, or high levels of collagen-I expression were distinguished. In asthmatic animals, collagen-I levels were increased in both medium and high expressers, and the number of myofibroblasts with high collagen-I content was elevated. Our findings determined that quantification of collagen-I deposition in myofibroblastic lung cells by flow cytometry is feasible in mouse models of asthma and indicative of increased collagen-I expression by asthmatic myofibroblasts. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Andrew Reichard
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Nicholas Wanner
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Eric Stuehr
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Mario Alemagno
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Kelly Weiss
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Kimberly Queisser
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio
| | - Serpil Erzurum
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio.,Respiratory Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Kewal Asosingh
- Department of Inflammation and Immunity, The Cleveland Clinic, Cleveland, Ohio.,Flow Cytometry Core, Lerner Research Institute The Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
7
|
Ngo QMT, Lee HS, Nguyen VT, Kim JA, Woo MH, Min BS. Chemical constituents from the fruits of Ligustrum japonicum and their inhibitory effects on T cell activation. PHYTOCHEMISTRY 2017; 141:147-155. [PMID: 28618355 DOI: 10.1016/j.phytochem.2017.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 06/07/2023]
Abstract
A previously undescribed nor-dammarane, 3β,20,23-trihydroxy-24,25,26,27-tetranordammarane; three previously undescribed secoiridoid glycosides, ligujaponosides A-B, and iso-oleonuzhenide; and twenty three known compounds, were isolated from the fruits of Ligustrum japonicum Thunb. Their chemical structures were elucidated by extensive spectroscopic analyses, including 1D and 2D NMR, and HRMS. The isolated compounds were screened for immunosuppressive effects on T activated cells by evaluating interleukin-2 (IL-2) production. Among them, sesamin inhibited IL-2 production in Jurkat T cells with an IC50 value of 38 ± 2 μM. In addition, sesamin inhibited the phosphorylation of extracellular signal-regulated protein kinase (ERK), a member of the mitogen-activated protein kinase (MAPK) family, in phorbol 12-myristate 13-acetate (PMA)/A23187-stimulated T cells. Therefore, sesamin was demonstrated to inhibit T cell activation via regulation of MAPK phosphorylation pathway.
Collapse
Affiliation(s)
- Quynh-Mai Thi Ngo
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, 38430, Republic of Korea
| | - Hyun-Su Lee
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, 38430, Republic of Korea
| | - Van Thu Nguyen
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, 38430, Republic of Korea
| | - Jeong Ah Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Mi Hee Woo
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, 38430, Republic of Korea
| | - Byung Sun Min
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, 38430, Republic of Korea.
| |
Collapse
|
8
|
Lee HY, Kim IK, Yoon HK, Kwon SS, Rhee CK, Lee SY. Inhibitory Effects of Resveratrol on Airway Remodeling by Transforming Growth Factor-β/Smad Signaling Pathway in Chronic Asthma Model. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2017; 9:25-34. [PMID: 27826959 PMCID: PMC5102832 DOI: 10.4168/aair.2017.9.1.25] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 05/25/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
Purpose Asthma is a chronic airway disease characterized by airway remodeling, leading to a progressive decline in lung function. Therapeutic agents that attenuate airway remodeling can complement the limited effects of traditional glucocorticoids. In this study, we investigated the effect of resveratrol on allergic airway inflammation and remodeling in a murine model of chronic bronchial asthma. Methods Peribronchial smooth muscle thickening that developed in mice challenged with a 3-month repeated exposure to ovalbumin (OVA) was used to study airway remodeling. Oral resveratrol was administered daily during the OVA challenge. The expression of TGF-β1/Smad signaling proteins and downstream mesenchymal markers in the presence or absence of resveratrol was examined in bronchial epithelial cells. Results OVA sensitization and chronic challenge increased airway hyperresponsiveness, inflammation, goblet cell hyperplasia, α-smooth muscle actin (SMA), and collagen deposition. Resveratrol effectively suppressed OVA-induced airway inflammation and remodeling. The expression of TGF-β1/phosphorylated Smad2/3 was increased in the lung tissues of OVA-challenged mice but effectively inhibited by resveratrol. In bronchial epithelial cells, the TGF-β1-induced expression of the mesenchymal markers snail, slug, vimentin, and α-SMA was suppressed by resveratrol treatment. Conclusions Resveratrol effectively ameliorated both airway inflammation and airway structural changes in a mouse model of bronchial asthma. These effects were mediated by decreased TGF-β1 expression, in turn suppressing TGF-β1/Smad signaling and the epithelial-mesenchymal transition process. Our results demonstrate the potential benefits of resveratrol for the treatment of airway remodeling associated with bronchial asthma.
Collapse
Affiliation(s)
- Hwa Young Lee
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Kyoung Kim
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyoung Kyu Yoon
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soon Suk Kwon
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chin Kook Rhee
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Sook Young Lee
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
9
|
Shen C, Jiang L, Shao H, You C, Zhang G, Ding S, Bian T, Han C, Meng Q. Targeted killing of myofibroblasts by biosurfactant di-rhamnolipid suggests a therapy against scar formation. Sci Rep 2016; 6:37553. [PMID: 27901027 PMCID: PMC5128858 DOI: 10.1038/srep37553] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/01/2016] [Indexed: 01/06/2023] Open
Abstract
Pathological myofibroblasts are often involved in skin scarring via generating contractile force and over-expressing collagen fibers, but no compound has been found to inhibit the myofibroblasts without showing severe toxicity to surrounding physiological cells. Here we report that di-rhamnolipid, a biosurfactant secreted by Pseudomonas aeruginosa, showed potent effects on scar therapy via a unique mechanism of targeted killing the myofibroblasts. In cell culture, the fibroblasts-derived myofibroblasts were more sensitive to di-rhamnolipid toxicity than fibroblasts at a concentration-dependent manner, and could be completely inhibited of their specific functions including α-SMA expression and collagen secretion/contraction. The anti-fibrotic function of di-rhamnolipid was further verified in rabbit ear hypertrophic scar models by presenting the significant reduction of scar elevation index, type I collagen fibers and α-SMA expression. In this regard, di-rhamnolipid treatment could be suggested as a therapy against skin scarring.
Collapse
Affiliation(s)
- Chong Shen
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Lifang Jiang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Huawei Shao
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Chuangang You
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Guoliang Zhang
- Ocean College, Zhejiang University of Technology, Hangzhou, PR China
| | - Sitong Ding
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Tingwei Bian
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Chunmao Han
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Qin Meng
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| |
Collapse
|