1
|
Zhang D, Sun T, Bao J, Fu J. Implications of DNA damage in chronic lung disease. Front Cell Dev Biol 2024; 12:1436767. [PMID: 39544366 PMCID: PMC11560874 DOI: 10.3389/fcell.2024.1436767] [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: 05/22/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
DNA plays an indispensable role in ensuring the perpetuation of life and safeguarding the genetic stability of living organisms. The emergence of diseases linked to a wide spectrum of responses to DNA damage has garnered increasing attention within the scientific community. There is growing evidence that patterns of DNA damage response in the lungs are associated with the onset, progression, and treatment of chronic lung diseases such as chronic obstructive pulmonary disease (COPD), asthma, and bronchopulmonary dysplasia (BPD). Currently, some studies have analyzed the mechanisms by which environmental factors induce lung DNA damage. In this article, we summarize inducible factors of lung DNA damage, current indicators, and methods for diagnosing DNA damage in chronic lung diseases and explore repair mechanisms after DNA damage including nonhomologous end-joining and homology-directed repair end joining pathways. Additionally, drug treatments that may reduce DNA damage or promote repair after it occurs in the lungs are briefly described. In general, more accurate assessment of the degree of lung DNA damage caused by various factors is needed to further elucidate the mechanism of lung DNA damage and repair after damage, so as to search for potential therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Li J, Liu X, Shi Y, Xie Y, Yang J, Du Y, Zhang A, Wu J. Differentiation in TCM patterns of chronic obstructive pulmonary disease by comprehensive metabolomic and lipidomic characterization. Front Immunol 2023; 14:1208480. [PMID: 37492573 PMCID: PMC10363632 DOI: 10.3389/fimmu.2023.1208480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 07/27/2023] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a complex disease involving inflammation, cell senescence, and autoimmunity. Dialectical treatment for COPD with traditional Chinese medicine (TCM) has the advantage of fewer side effects, more effective suppression of inflammation, and improved immune function. However, the biological base of TCM pattern differentiation in COPD remains unclear. Methods Liquid Chromatography-Quadrupole-Orbitrap mass spectrometry (LC-Q-Orbitrap MS/MS) based metabolomics and lipidomics were used to analyze the serum samples from COPD patients of three TCM patterns in Lung Qi Deficiency (n=65), Lung-Kidney Qi Deficiency (n=54), Lung-Spleen Qi Deficiency (n=52), and healthy subjects (n=41). Three cross-comparisons were performed to characterize metabolic markers for different TCM patterns of COPD vs healthy subjects. Results We identified 28, 8, and 16 metabolites with differential abundance between three TCM patterns of COPD vs healthy subjects, respectively, the metabolic markers included cortisol, hypoxanthine, fatty acids, alkyl-/alkenyl-substituted phosphatidylethanolamine, and phosphatidylcholine, etc. Three panels of metabolic biomarkers specific to the above three TCM patterns yielded areas under the receiver operating characteristic curve of 0.992, 0.881, and 0.928, respectively, with sensitivity of 97.1%, 88.6%, and 91.4%, respectively, and specificity of 96.4%, 81.8%, and 83.9%, respectively. Discussion Combining metabolomics and lipidomics can more comprehensively and accurately trace metabolic markers. As a result, the differences in metabolism were proven to underlie different TCM patterns of COPD, which provided evidence to aid our understanding of the biological basis of dialectical treatment, and can also serve as biomarkers for more accurate diagnosis.
Collapse
Affiliation(s)
- Jiansheng Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinguang Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanmin Shi
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Xie
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jianya Yang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yan Du
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ang Zhang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinyan Wu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
3
|
Tao L, Lu X, Fu Z, Tian Y, Liu X, Li J, Zhao P. Tong Sai granules improves AECOPD via regulation of MAPK-SIRT1-NF-κB pathway and cellular senescence alleviation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116622. [PMID: 37210015 DOI: 10.1016/j.jep.2023.116622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/22/2023] [Accepted: 05/08/2023] [Indexed: 05/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tong Sai granules (TSG) a traditional Chinese medicine, are used to treat acute exacerbations of chronic obstructive pulmonary disease (AECOPD). Cellular senescence is considered the mechanism underlying AECOPD progression. AIM OF THE STUDY This study aimed to investigate the therapeutic mechanisms of TSG in an AECOPD rat model (established using cigarette smoke exposure and bacterial infection) and focused on the inhibition of cellular senescence in vivo and in vitro. MATERIALS AND METHODS Histological changes and levels of inflammatory cytokines, matrix metalloproteinases (MMPs), p53, and p21 were determined. A cellular senescence model was established by challenging airway epithelial cells with cigarette smoke extract (CSE) and lipopolysaccharide (LPS). Quantitative PCR, western blotting, and immunofluorescence were used to measure mRNA and protein levels. Additionally, UPLC-Q-Extractive-Orbitrap MS analysis, network analysis, and transcriptomics were used to analyze the potential compounds and molecular mechanisms of TSG. RESULTS The results showed that oral administration of TSG significantly reduced the severity of AECOPD in rats by ameliorating lung function decline and pathological injuries and increasing the levels of C-reactive protein and serum amyloid A, two well-known proinflammatory mediators of the acute phase response. Oral TSG administration also decreased the expression levels of proinflammatory cytokines (e.g., IL-6, IL-1β, and TNF-α), MMPs (e.g., MMP-2 and MMP-9), critical regulators of senescence such as p21 and p53, and the apoptotic marker γH2AX, all of which are factors in cellular senescence in lung tissue. TSG4 was isolated from TSGs using macroporous resin and found to significantly suppress cellular senescence in CSE/LPS-induced bronchial epithelial cells. Furthermore, 26 of 56 compounds identified in TSG4 were used to predict 882 potential targets. Additionally, 317 differentially expressed genes (DEGs) were detected in CSE/LPS-treated bronchial epithelial cells. Network analysis of the 882 targets and 317 DEGs revealed that TSG4 regulated multiple pathways, among which the mitogen-activated protein kinase-sirtuin 1-nuclear factor kappa B (MAPK-SIRT1-NF-κB) pathway is important in terms of antisenescent mechanisms. Moreover, in CSE/LPS-induced bronchial epithelial cells, p-p38, p-ERK1/2, p-JNK, and p-p65 levels were increased and SIRT1 levels were decreased after TSG4 treatment. Additionally, oral TSG administration decreased p-p38 and p-p65 levels and increased SIRT1 levels in the lung tissues of AECOPD model rats. CONCLUSION Collectively, these results indicate that TSGs ameliorate AECOPD by regulating the MAPK-SIRT1-NF-κB signaling pathway and subsequently suppressing cellular senescence.
Collapse
Affiliation(s)
- Liuying Tao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China.
| | - Xiaofan Lu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China
| | - Zijian Fu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Xinguang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
4
|
Tiao-Bu-Fei-Shen Formula Improves Glucocorticoid Resistance of Chronic Obstructive Pulmonary Disease via Downregulating the PI3K-Akt Signaling Pathway and Promoting GR α Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:4359616. [PMID: 36820399 PMCID: PMC9938767 DOI: 10.1155/2023/4359616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/21/2022] [Accepted: 11/24/2022] [Indexed: 02/12/2023]
Abstract
Objective To predict and determine the mechanism through which Tiao-Bu-Fei-Shen (TBFS) formula improves glucocorticoid resistance in chronic obstructive pulmonary disease (COPD), using network pharmacology, molecular docking technology, and in vitro studies. Methods The main active components and associated targets of TBFS were screened using the systems pharmacology database of traditional Chinese medicine database (TCMSP). The main COPD targets were retrieved from the Human Gene (GeneCards) and DrugBank databases. A protein-protein interaction (PPI) network was constructed using the protein interaction platform STRING and Cytoscape 3.6.1. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genome Pathway (KEGG) analyses were performed using the biological information annotation database Metascape. Molecular docking was performed using the AutoDock Vina software. THP-1 monocytes were treated with TBFS-containing serum and cigarette smoke extract (CSE) for 48 h, and cell proliferation in each group was determined using cell counting kit-8 (CCK-8). A COPD cell model was constructed by stimulating THP-1 monocytes with CSE for 12 h. A lentivirus vector for RNA interference of histone deacetylase 2 (HDAC2) gene was constructed and transfected into the THP-1 monocytes, and the transfection efficiency was verified using quantitative polymerase chain reaction (qPCR) and western blotting (WB). The expression of HDAC2 in each group of cells was detected using qPCR, and the expression of HDAC2, phosphoinositide-3 kinase (PI3K) p85α, glucocorticoid receptor α (GRα), and P-AKT1 in each group of cells was detected through WB. Results A total of 344 TBFS active components, 249 related drug targets, 1,171 COPD target proteins, and 138 drug and disease intersection targets were obtained. Visual analysis of the PPI network map revealed that the core COPD targets of TBFS were AKT1, IL-6, TNF, TP53, and IL1-β. KEGG pathway enrichment analysis resulted in the identification of 20 signaling pathways as the main pathways involved in the action of TBFS against COPD, including the PI3K-Akt, TNF, and IL-17 signaling pathways. Molecular docking experiments revealed a strong binding capacity of kaempferol, luteolin, and quercetin to the ATK1 protein in TBFS, with quercetin performing the best. PCR results showed that treatment with TBFS significantly increased the expression levels of HDAC2 in the COPD model. WB results showed that TBFS treatment significantly increased the expression levels of GRα and HDAC2 in the COPD model, while reducing the expression levels of P-AKT1. Conclusion TBFS treatment improves glucocorticoid resistance observed in COPD through downregulation of the PI3K-Akt signaling pathway and promotion of GRα expression.
Collapse
|
5
|
Sun Y, Chen X, Zhang L, Yuan WA, Chen Q, Zhang YB, Liu LJ, Zhang W, Sun M. Efficiency and Safety of Baofei Granules in Chronic Obstructive Pulmonary Disease (Lung and Spleen Qi Deficiency Syndrome): A Multicenter, Randomized, Double-Blind, Placebo-Controlled Phase II Clinical Trial. Drug Des Devel Ther 2022; 16:4251-4267. [PMID: 36540716 PMCID: PMC9759976 DOI: 10.2147/dddt.s382285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Baofei Granules (BFGs) have been extensively applied in the clinical treatment of chronic obstructive pulmonary disease (COPD) and significantly have affected COPD patients with lung and spleen qi deficiency syndrome. However, the data from previous small-sample clinical trials are limited. This trial aimed to estimate the efficiency and safety of BFGs in COPD with lung and spleen qi deficiency syndrome. METHODS It is a multicenter, randomized, double-blind, placebo-controlled phase II clinical trial. The 216 stable COPD patients will be divided randomly in a ratio of 1:1. The whole trial period consists of a 4-week introductory period, a 52-week treatment period and a 48-week follow-up. Study visits occur every 4 weeks during the treatment period and every 12 weeks during the follow-up. All the subjects will receive 10g BFGs or placebo three times per day for 56 weeks and be followed up for 48 weeks. The primary efficiency evaluation outcome will be the frequency and duration of AECOPD, and the secondary efficiency evaluation outcome will be pulmonary function tests (PFTs), modified Medical Research Council (mMRC) dyspnoea scale, six-minute walking test (6MWT), COPD assessment test (CAT) score, traditional Chinese medicine (TCM) syndrome score, the frequency of emergency medication, BODE index, and the time to first Clinically important deterioration (CID). The safety evaluation outcomes will be adverse events (AEs), vital signs, physical examination, twelve-lead electrocardiogram (ECG), and laboratory examinations. All the data will be analyzed by SAS9.4. DISCUSSION This is the first and largest clinical trial that evaluates the efficiency and safety of BFGs for COPD with lung and spleen qi deficiency syndrome. It will provide valuable clinical evidence for recommendations on COPD management by the integrated TCM and western medicine. TRIAL REGISTRATION CTR20211280. Date: June 09, 2021. http://www.chinadrugtrials.org.cn/clinicaltrials.searchlistdetail.dhtml?id=383a370ecd9f43d7af6f1c8585779e1a.
Collapse
Affiliation(s)
- Yuan Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Xuan Chen
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Wei-an Yuan
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Qi Chen
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yi-bao Zhang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Lu-jiong Liu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Wei Zhang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Meng Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
6
|
Effects of Fermented Green Tea Waste Extract Gels on Oxidative Damage in Short-Term Passive Smoking Mice. Gels 2022; 8:gels8080461. [PMID: 35892720 PMCID: PMC9332167 DOI: 10.3390/gels8080461] [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: 06/06/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Passive smoking is extensively studied because of its harmfulness to human health. In this study, the effects of fermented green tea waste extract gels (GTEG) on oxidative damage in mice exposed to short-term cigarette smoke (CS) were investigated. The GTEG is prepared from green tea waste extract and microbial transglutaminase (MTGase). The lung injury model of mice was established through passive smoking for 5 days. The experimental results revealed the following findings. (1) The GTEG induced by MTGase has obvious gel properties; (2) GTEG has strong biological activity and antioxidant properties in vitro; (3) The passive smoking model was established successfully; specifically, the lung tissue of the model mice exhibited inflammatory symptoms, oxidative stress response appeared in their bodies, and their inflammatory indicators increased; (4) Compared with the passive smoking model group, the mice, which were exposed to CS and received GTEG treatment, exhibited increased food intake and body weight; increased total superoxide dismutase and glutathione peroxidase activity in serum; significant decreases (p < 0.05) in the content levels of the inflammatory factors malondialdehyde, interleukin (IL)-6, and tumor necrosis factor α (TNF-α); and inhibited expression of IL-6, IL-33, TNF-α, and IL-1β inflammatory genes. The results indicated that taking GTEG can relieve the oxidative stress injury of mice caused by short-term CS and has antioxidant properties.
Collapse
|
7
|
Zhou P, Yu W, Zhang C, Chen K, Tang W, Li X, Liu Z, Xia Q. Tiao-bu-fei-shen formula promotes downregulation of the caveolin 1-p38 mapk signaling pathway in COPD - Associated tracheobronchomalacia cell model. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115256. [PMID: 35367574 DOI: 10.1016/j.jep.2022.115256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/17/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tiao-bu-fei-shen (TBFS) formula, extensively used in Traditional Chinese Medicine (TCM), can enhance therapeutic efficacy and reduce the frequency of acute exacerbations of lung-kidney Qi deficiency in patients with chronic obstructive pulmonary disease (COPD). According to both TCM theory and long-term observation of practice, TBFS has become an effective treatment for COPD-associated tracheobronchomalacia (TBM). AIM OF THE STUDY To investigate the mechanism of the TBFS formula in treating COPD-associated TBM based on caveolin 1-p38 MAPK signaling and apoptosis. MATERIALS AND METHODS A rat COPD model was prepared by exposure to smoking combined with tracheal lipopolysaccharide injection. The trachea or bronchus chondrocytes from COPD rats were isolated, cultured, and treated with 10 ng/mL IL-1β for 24 h to develop a model of COPD-associated TBM. Normal rats were administered TBFS to prepare drug-containing serum, and CCK8 assays were used to screen the optimal drug-containing serum concentration and SB203580 dose. TBFS drug-containing serum and SB203580 were processed separately for the control, model, drug-containing serum, blocker, and drug-containing serum combined with blocker groups. Flow cytometry and CCK8 assays were used to detect apoptosis and proliferative activity. Toluidine blue staining and immunohistochemistry were used to analyze the chondrocyte proteoglycan and type II collagen content. Western blotting was used to detect the expression of caveolin 1, p-p38 MAPK, TNF-α, IL-1β, MMP-13, Bax, and Bcl-2 proteins. Quantitative PCR was used to detect the expression of caveolin 1, p38 MAPK, IL-1β, MMP-13, Bax, Bcl-2, and miR-140-5p. RESULTS The isolation and identification of bronchial chondrocytes from COPD rats revealed that 10 ng/mL IL-1β can produce a stable COPD-associated TBM model. Screened via the CCK8 method, fourth-generation bronchial chondrocytes were determined as the optimal cells, and 5 μM SB203580 and 5% low-dose drug-containing serum were the optimal intervention doses. The experimental chondrocytes of each group were treated separately for 48 h. Toluidine blue staining and immunohistochemical analysis revealed that TBFS drug-containing serum, SB203580, and TBFS drug-containing serum combined with SB203580 can effectively increase the proteoglycan and type II collagen content after chondrocyte degradation. Flow cytometry of cells treated with SB203580 and TBFS drug-containing serum combined with SB203580 revealed significantly reduced cell apoptosis and enhanced cell proliferation activity. Western blot and qPCR analyses revealed that the TBFS drug-containing serum, SB203580, and TBFS drug-containing serum combined with SB203580 effectively inhibit the expression of caveolin 1, p-p38 MAPK, MMP-13, IL-1β, TNF-α, and Bax proteins while promoting Bcl -2 protein expression. Treatment with TBFS drug-containing serum and SB203580 effectively inhibited the expression of MMP-13, p38 MAPK, caveolin 1, and Bax genes, and promoted the expression of Bcl-2 and miR-140-5p genes. CONCLUSIONS A concentration of 10 ng/mL of IL-1β can generate a stable COPD-associated TBM cell model. TBFS can improve the proteoglycan and type II collagen content, increase cell activity, and reduce the amount of chondrocyte apoptosis. The role of TBFS may be related to mechanisms of inhibiting the expression of the key signaling molecules caveolin 1 and p-p38 MAPK in the caveolin 1-p38 MAPK signaling pathway, thereby reducing the expression of the downstream effector products MMP-13, IL-1β, and TNF-α, while inhibiting the expression of the apoptotic gene Bax and improving the expression of Bcl-2 and miR-140-5p genes.
Collapse
Affiliation(s)
- Pengcheng Zhou
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Wei Yu
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Keling Chen
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Wenjun Tang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Xuelian Li
- Department of Emergency, Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Zijun Liu
- Department of Intensive Care Unit, Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| | - Qianming Xia
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan province, PR China.
| |
Collapse
|
8
|
Mao J, Li Y, Bian Q, Xuan Y, Li J, Wang Z, Feng S, Liu X, Tian Y, Li S. The Bufei Jianpi Formula Improves Mucosal Immune Function by Remodeling Gut Microbiota Through the SCFAs/GPR43/NLRP3 Pathway in Chronic Obstructive Pulmonary Disease Rats. Int J Chron Obstruct Pulmon Dis 2022; 17:1285-1298. [PMID: 35673595 PMCID: PMC9167601 DOI: 10.2147/copd.s359428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/22/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jing Mao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Ya Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Qingqing Bian
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Yinshuang Xuan
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jingmei Li
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Zhikun Wang
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Suxiang Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Suyun Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Correspondence: Suyun Li, Email
| |
Collapse
|
9
|
HUANG Y, WAN B, HUANG Z, LIN M. Effect of Jianpibufei plaster on chronic obstructive pulmonary disease in mice. FOOD SCIENCE AND TECHNOLOGY 2021. [DOI: 10.1590/fst.39320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Ying HUANG
- Guangzhou University of Chinese Medicine, China; Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, China
| | - Bin WAN
- Chinese Academy of Medical Sciences, China
| | - Zhenyan HUANG
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, China
| | - Mian LIN
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, China
| |
Collapse
|
10
|
Li J, Xie Y, Zhao P, Qin Y, Oliver BG, Tian Y, Li S, Wang M, Liu X. A chinese herbal formula ameliorates COPD by inhibiting the inflammatory response via downregulation of p65, JNK, and p38. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 83:153475. [PMID: 33545548 DOI: 10.1016/j.phymed.2021.153475] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Bufei Yishen formula (BYF), a traditional Chinese medicine (TCM), is an effective therapeutic strategy for patients with chronic obstructive pulmonary disease (COPD). PURPOSE To evaluate the efficacy of BYF and investigate its therapeutic mechanisms. METHODS A total of 134 patients completed the study: 68 patients treated by BYF combined with conventional Western medicine in the trial group; and 66 patients treated using conventional Western medicine in the control group. The efficacy of BYF was evaluated by a subgroup analysis of data obtained from a four-center, open-label, randomized controlled trial of comprehensive TCM interventions. A rat model of COPD was treated with the key active molecules (KAM) of BYF for 8 weeks. An in vitro model of COPD was also treated with KAM. RESULTS Patients treated with BYF had reduced frequency of acute exacerbation of COPD (p < 0.001) and duration (p = 0.028), dyspnea scale (p = 0.007), 6-min walking distance (p = 0.048). There were no differences observed in forced vital capacity in one second (FVC), forced expiratory volume in one second (FEV1), and FEV1 percentage of the predicted value (FEV1%). The five KAM of BYF (KAM-BYF) improved lung function, including tidal volume, minute ventilation, peak expiratory flow, FVC, FEV0.1, and FEV0.3, and pathological changes in COPD rats. Treatment with KAM-BYF markedly decreased the levels of interleukin 6 (IL6), tumor necrosis factor-α (TNF-α), matrix metalloproteinase 9 (MMP9), and MMP12 in serum and bronchial alveolar lavage fluid. In airway epithelial cells, KAM-BYF decreased the levels of TNF-α-induced IL8 and IL6. Finally, we discovered that the anti-inflammatory effects of KAM-BYF in COPD rats and BEAS-2Bs were mediated through inhibition of nuclear factor-kappaB (NF-κB) p65, c-Jun NH2-terminal kinase (JNK), and p38 mitogen-activated protein kinase signaling. CONCLUSIONS BYF exerts beneficial effects in patients with COPD via inhibition of inflammation.
Collapse
Affiliation(s)
- Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Yang Xie
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Yanqin Qin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Brian G Oliver
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Woolcock Institute of Medical Research, Respiratory Cellular and Molecular Biology, The University of Sydney, New South Wales 2037, Australia
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Suyun Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Minghang Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| |
Collapse
|
11
|
Zhao P, Liu X, Dong H, Tian Y, Feng S, Zhao D, Ren Z, Zhang L, Li J. Bufei Yishen Formula Restores Th17/Treg Balance and Attenuates Chronic Obstructive Pulmonary Disease via Activation of the Adenosine 2a Receptor. Front Pharmacol 2020; 11:1212. [PMID: 32848801 PMCID: PMC7427463 DOI: 10.3389/fphar.2020.01212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/24/2020] [Indexed: 01/24/2023] Open
Abstract
Bufei Yishen formula (BYF) is a Traditional Chinese Medicine (TCM) reported to ameliorate chronic obstructive pulmonary disease (COPD) by regulating the balance between T helper (Th) 17 and regulatory T (Treg) cells. However, its mechanism remains unknown. Therefore, this study aimed to explore the underlying mechanisms of BYF. Naïve CD4+ T cells were exposed to anti-CD3, anti-CD28, transforming growth factor (TGF)-β, and/or interleukin (IL)-6 to promote their differentiation into Th17 or Treg cells. A rat model of cigarette smoke- and bacterial infection-induced COPD was established and orally treated with BYF and/or an adenosine 2a receptor (A2aR) antagonist. Then, the rats were sacrificed, their lung tissues were removed for histological analysis, and their spleens were collected to evaluate Th17 and Treg cells. The results showed that BYF significantly suppressed Th17 cell differentiation and its related cytokines and enhanced Treg cell differentiation and its related cytokines. In addition, BYF activated the A2aR, increased the levels of p-signal transducer and activator of transcription (STAT)5, and decreased the level of p-STAT3 in Treg and Th17 cells. The A2aR antagonist suppressed the changes induced by BYF treatment in Th17 and Treg cells. Furthermore, the A2aR antagonist diminished the therapeutic effect of BYF on COPD, as indicated by the lung injury scores, bronchiole wall thickness, small pulmonary vessels wall thickness, bronchiole stenosis, alveolar diameters, decrease in inflammatory cytokines, increase in alveolar number, and lung functions. Similarly, the A2aR antagonist reversed the effects of BYF on the proportion of Th17 and Treg cells in the spleen. Additionally, BYF increased the protein and mRNA levels of A2aR and regulated the phosphorylation of STAT3 and STAT5 in spleen and lung tissues, which were inhibited by cotreatment with the A2aR antagonist. In conclusion, this study suggested that BYF exhibited its anti-COPD efficacy by restoring the Th17/Treg balance via activating A2aR, which may provide evidence for the clinical application of BYF in the treatment of COPD.
Collapse
Affiliation(s)
- Peng Zhao
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xuefang Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Haoran Dong
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yange Tian
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Suxiang Feng
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
| | - Di Zhao
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhouxin Ren
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lanxi Zhang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiansheng Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.,Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
12
|
Li J, Ma J, Tian Y, Zhao P, Liu X, Dong H, Zheng W, Feng S, Zhang L, Wu M, Zhu L, Liu S, Zhao D. Effective-component compatibility of Bufei Yishen formula II inhibits mucus hypersecretion of chronic obstructive pulmonary disease rats by regulating EGFR/PI3K/mTOR signaling. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112796. [PMID: 32344236 DOI: 10.1016/j.jep.2020.112796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/07/2020] [Accepted: 03/23/2020] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The effective-component compatibility of Bufei Yishen formula I (ECC-BYF I), a combination of 10 compounds, including total ginsenosides, astragaloside IV, icariin, and paeonol, etc., is derived from Bufei Yishen formula (BYF). The efficacy and safety of ECC-BYF I is equal to BYF. However, the composition of ECC-BYF I needs to be further optimized. Based on the beneficial effects of BYF and ECC-BYF I on chronic obstructive pulmonary disease (COPD), this study aimed to optimize the composition of ECC-BYF I and to explore the effects and mechanisms of optimized ECC-BYF I (ECC-BYF II) on mucus hypersecretion in COPD rats. MATERIALS AND METHODS ECC-BYF I was initially optimized to six groups: optimized ECC-BYF I (OECC-BYF I)-A~F. Based on a COPD rat model, the effects of OECC-BYF I-A~F on COPD rats were evaluated. R-value comprehensive evaluation was used to evaluate the optimal formula, which was named ECC-BYF II. The changes in goblet cells and expression of mucins and the mRNA and proteins involved in the epidermal growth factor receptor/phosphoinositide-3-kinase/mammalian target of rapamycin (EGFR/PI3K/mTOR) pathway were evaluated to explore the effects and mechanisms of ECC-BYF II on mucus hypersecretion. RESULTS ECC-BYF I and its six optimized groups, OECC-BYF I-A~F, had beneficial effects on COPD rats in improving pulmonary function and lung tissue pathology, reducing inflammation and oxidative stress, and improving the protease/anti-protease imbalance and collagen deposition. R-value comprehensive evaluation found that OECC-BYF I-E (paeonol, icariin, nobiletin, total ginsenoside, astragaloside IV) was the optimal formula for improving the comprehensive effects (lung function: VT, MV, PEF, EF50, FVC, FEV 0.1, FEV 0.1/FVC; histological changes: MLI, MAN; IL-1β, IL-6, TNF-α, MMP-9, TIMP-1, T-AOC, LPO, MUC5AC, Collagen I and Collagen III). OECC-BYF I-E was named ECC-BYF II. Importantly, the effect of ECC-BYF II showed no significant difference from BYF and ECC-BYF I. ECC-BYF II inhibited mucus hypersecretion in COPD rats, which manifested as reducing the expression of MUC5AC and MUC5B and the hyperplasia rate of goblet cells. The mRNA and protein expression levels of EGFR, PI3K, Akt, and mTOR were increased in COPD rats and were obviously downregulated after ECC-BYF II administration. CONCLUSION ECC-BYF II, which consists of paeonol, icariin, nobiletin, total ginsenoside and astragaloside IV, has beneficial effects equivalent to BYF and ECC-BYF I on COPD rats. ECC-BYF II significantly inhibited mucus hypersecretion, which may be related to the regulation of the EGFR/PI3K/mTOR pathway.
Collapse
Affiliation(s)
- Jiansheng Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Jindi Ma
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Yange Tian
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Peng Zhao
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Xuefang Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Haoran Dong
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Wanchun Zheng
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Suxiang Feng
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Lanxi Zhang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Mingming Wu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Lihua Zhu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Shuai Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Di Zhao
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., 450046, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
13
|
Li JS, Liu XF, Dong HR, Zheng WC, Feng SX, Tian YG, Zhao P, Ma JD, Ren ZX, Xie Y. Effective-constituent compatibility-based analysis of Bufei Yishen formula, a traditional herbal compound as an effective treatment for chronic obstructive pulmonary disease. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:351-362. [PMID: 32565294 DOI: 10.1016/j.joim.2020.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Critical effective constituents were identified from Bufei Yishen formula (BYF), a traditional herbal compound and combined as effective-constituent compatibility (ECC) of BYF I, which may have potential bioactive equivalence to BYF. METHODS The active constituents of BYF were identified using four cellular models and categorised into Groups 1 (Bufeiqi), 2 (Bushen), 3 (Huatan) and 4 (Huoxue) according to Chinese medicinal theory. An orthogonal design and a combination method were used to determine the optimal ratios of effective constituents in each group and the ratios of "Groups 1 to 4" according to their pharmacological activity. We also comprehensively assessed bioactive equivalence between the BYF and the ECC of BYF I in a rat model of chronic obstructive pulmonary disease (COPD). RESULTS We identified 12 active constituents in BYF. The numbers of constituents in Groups 1 to 4 were 3, 2, 5 and 2, respectively. We identified the optimal ratios of effective constituents within each group. In Group 1, total ginsenosides:Astragalus polysaccharide:astragaloside IV ratio was 9:5:2. In Group 2, icariin:schisandrin B ratio was 100:12.5. In Group 3, nobiletin:hesperidin:peimine:peiminine:kaempferol ratio was 4:30:6.25:0:0. In Group 4, paeoniflorin:paeonol ratio was 4:1. An orthogonal design was then used to establish the optimal ratios of Group 1, Group 2, Group 3 and Group 4 in ECC of BYF I. The ratio for total ginsenosides:Astragalus polysaccharide:astragaloside IV:icariin:schisandrin B:nobiletin:hesperidin:peimine:paeoniflorin:paeonol was determined to be 22.5:12.5:5:100:12.5:4:30:6.25:25:6.25. A comprehensive evaluation confirmed that ECC of BYF I presented with bioactive equivalence to the original BYF. CONCLUSION Based on the ECC of traditional Chinese medicine formula method, the effective constituents of BYF were identified and combined in a fixed ratio as ECC of BYF I that was as effective as BYF itself in treating rats with COPD.
Collapse
Affiliation(s)
- Jian-Sheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China.
| | - Xue-Fang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hao-Ran Dong
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Wan-Chun Zheng
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Su-Xiang Feng
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Yan-Ge Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jin-di Ma
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhou-Xin Ren
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Yang Xie
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| |
Collapse
|
14
|
Wang W, Zha G, Zou JJ, Wang X, Li CN, Wu XJ. Berberine Attenuates Cigarette Smoke Extract-induced Airway Inflammation in Mice: Involvement of TGF-β1/Smads Signaling Pathway. Curr Med Sci 2019; 39:748-753. [PMID: 31612392 DOI: 10.1007/s11596-019-2101-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/02/2019] [Indexed: 02/06/2023]
Abstract
Although several studies confirmed that berberine may attenuate airway inflammation in mice with chronic obstructive pulmonary disease (COPD), its underlying mechanisms were not clear until now. We aimed to establish an experiment mouse model for COPD and to investigate the effects of berberine on airway inflammation and its possible mechanism in COPD model mice induced by cigarette smoke extract (CSE). Twenty SPF C57BL/6 mice were randomly divided into PBS control group, COPD model group, low-dose berberine group and high-dose berberine group, 5 mice in each group. The neutrophils and macrophages were examined by Wright's staining. The levels of inflammatory cytokines TNF-α and IL-6 in bronchoalveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assay. The expression levels of TGF-β1, Smad2 and Smad3 mRNA and proteins in lung tissues were respectively detected by quantitative real-time polymerase chain reaction and Western blotting. It was found that CSE increased the number of inflammation cells in BALF, elevated lung inflammation scores, and enhanced the TGF-β1/Smads signaling activity in mice. High-dose berberine restrained the alterations in the COPD mice induced by CSE. It was concluded that high-dose berberine ameliorated CSE-induced airway inflammation in COPD mice. TGF-β1/Smads signaling pathway might be involved in the mechanism. These findings suggested a therapeutic potential of high-dose berberine on the CSE-induced airway inflammation.
Collapse
Affiliation(s)
- Wen Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Gan Zha
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin-Jing Zou
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xun Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chun-Nian Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao-Jun Wu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
15
|
Tang B, Xu Q, Xuan L, Wang H, Zhang H, Wang X, Kang P. Circ 0001434 RNA reduces inflammation in acute lung injury model through Wnt/β-catenin and NF-κB by miR-625-5p. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3290-3300. [PMID: 31934172 PMCID: PMC6949842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/26/2019] [Indexed: 06/10/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common and complicated inflammatory lung diseases. Circ RNAs have emerged as a novel class of gene regulatory molecules that play vital roles in multiple complex diseases, including ALI. In this study, we aimed to identify potential regulators of Circ 0001434 on acute lung injury (ALI) and to explore their roles in lipopolysaccharide (LPS)-induced ALI. In a mouse ALI model, Circ 0001434 expression was effectively down-regulated, compared with the control group. Up-regulation of Circ 0001434 effectively decreased the inflammation of ALI in an in vitro model. Down-regulation of Circ 0001434 effectively promoted inflammation in ALI in an in vitro model. Over-expression of Circ 0001434 induced Wnt and β-catenin protein expression, and suppressed NF-κB p65 protein expression in the ALI in vitro model by miR-625-5p. Down-regulation of Circ 0001434 significantly suppressed Wnt and β-catenin, and induced NF-κB p65 protein expression in the ALI in vitro model by miR-625-5p. Wnt reduced the function of Circ 0001434 on inflammation in ALI in an in vitro model. The inhibition of miR-625-5p reversed the function of anti-Circ 0001434 on inflammation in ALI vitro model. Taken together, Circ 0001434 mediates ALI-induced lung inflammation through Wnt/β-catenin and NF-κB activation by miR-625-5p.
Collapse
Affiliation(s)
- Bi Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Qingmei Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Ling Xuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Hongju Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Heng Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Xiaojing Wang
- Clinical and Basic Provincial Laboratory of Respiratory System Diseases of Anhui Province, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| | - Pinfang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical CollegeBengbu, Anhui Province, P. R. China
| |
Collapse
|
16
|
Lin L, Hou G, Han D, Yin Y, Kang J, Wang Q. Ursolic acid alleviates airway-vessel remodeling and muscle consumption in cigarette smoke-induced emphysema rats. BMC Pulm Med 2019; 19:103. [PMID: 31170951 PMCID: PMC6555740 DOI: 10.1186/s12890-019-0826-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND This study assessed the effects of ursolic acid (UA) on airway-vessel remodeling and muscle atrophy in cigarette smoke (CS)-induced emphysema rats and investigated potential underlying mechanisms. METHODS Emphysema was induced in a rat model with 3 months of CS exposure. Histology and immunohistochemistry (IHC) stains were used to assess airway-vessel remodeling and muscle atrophy-associated changes. Levels of cleaved-caspase3, 8-OHdG, and S100A4 were measured in airways and associated vessels to evaluate cell apoptosis, oxidant stress, epithelial-to-mesenchymal transition (EMT), and endothelial-to-mesenchymal transition (EndMT)-associated factors. Western blot and/or IHC analyses were performed to measure transforming growth factor-beta 1(TGF-β1)/Smad2.3, alpha-smooth muscle actin (α-SMA), and insulin-like growth factor 1 (IGF1) expression. We also gave cultured HBE and HUVEC cells Cigarette Smoke Extract (CSE) administration and UA intervention. Using Western blot method to measure TGF-β1/Smad2.3, α-SMA, S100A4, and IGF1 molecules expression. RESULTS UA decreased oxidant stress and cell apoptosis in airway and accompanying vascular walls of cigarette smoke-induced emphysema model rats. UA alleviated EMT, EndMT, changes associated with airway-vessel remodeling and muscle atrophy. The UA effects were associated with IGF1 and TGF-β1/Smad2.3 pathways. CONCLUSIONS UA reduced EMT, EndMT, airway-vessel remodeling, and musculi soleus atrophy in CS-induced emphysema model rats at least partly through IGF1 and TGF-β1/Smad2.3 signaling pathways.
Collapse
Affiliation(s)
- Li Lin
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| | - Gang Hou
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| | - Dan Han
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| | - Yan Yin
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| | - Jian Kang
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| | - Qiuyue Wang
- Institute of Respiratory Disease, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001 China
| |
Collapse
|
17
|
Ma J, Tian Y, Li J, Zhang L, Wu M, Zhu L, Liu S. Effect of Bufei Yishen Granules Combined with Electroacupuncture in Rats with Chronic Obstructive Pulmonary Disease via the Regulation of TLR-4/NF- κB Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:6708645. [PMID: 31275415 PMCID: PMC6560336 DOI: 10.1155/2019/6708645] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The combined therapy of Bufei Yishen granules (BY) and electroacupuncture (EA) has shown good effects clinically in treating chronic obstructive pulmonary disease (COPD). The present study aimed to observe the effects of the BY + EA combination in a COPD rat model and dissect the potential mechanisms via Toll-like receptor (TLR) 4/nuclear factor kappa B (NF-κB) signaling. METHODS The COPD rats were treated with normal saline, aminophylline, Bufei Yishen granules, electroacupuncture, or Bufei Yishen granules combined with electroacupuncture. The pulmonary function; lung tissue histology; levels of inflammatory factors; expression levels of TLR-4, inhibitor of nuclear factor kappa B (IκB), and NF-κB; and activation of NF-κB in the lung tissues were evaluated. RESULTS Pulmonary function was markedly decreased in the COPD rats, and the lung tissue histology of the COPD rats showed severe pathological changes. The pulmonary function and lung tissue morphology in the treatment groups (APL, BY, EA, and BY + EA) were improved. The increased levels of the inflammatory cytokines interleukin (IL)-1β and IL-6 indicated a chronic inflammatory state in the COPD rats. In the BY, EA, and BY + EA groups, the levels of IL-1β and IL-6 were decreased, especially in the BY + EA group. In addition, the mRNA and protein expression levels of TLR-4, IκB, and NF-κB were obviously downregulated in the BY and BY + EA groups; and the NF-κB p65 activation was significantly decreased in the BY, EA, and BY + EA groups. CONCLUSIONS Bufei Yishen granules and electroacupuncture have curative effects in COPD rats, and the combination therapy of Bufei Yishen granules and electroacupuncture is superior. The TLR-4/NF-κB pathway may be involved in the potential mechanisms by which Bufei Yishen granules and electroacupuncture reduce inflammation.
Collapse
Affiliation(s)
- Jindi Ma
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Yange Tian
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Jiansheng Li
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Lanxi Zhang
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Mingming Wu
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Lihua Zhu
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Shuai Liu
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| |
Collapse
|
18
|
Chen J, Si L, Zhou L, Deng Y. Role of bone marrow mesenchymal stem cells in the development of PQ‑induced pulmonary fibrosis. Mol Med Rep 2019; 19:3283-3290. [PMID: 30816470 DOI: 10.3892/mmr.2019.9976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/11/2019] [Indexed: 11/06/2022] Open
Abstract
Paraquat (PQ) poisoning‑induced pulmonary fibrosis is one of the primary causes of mortality in patients with PQ poisoning. The potential mechanism of PQ‑induced pulmonary fibrosis was thought to be mediated by inflammation. Recently, bone marrow‑derived mesenchymal stem cells (BMSCs) have been considered as a potential strategy for the treatment of fibrotic disease due to their anti‑inflammatory and immunosuppressive effects. In the present study, an increased accumulation of BMSCs in a mouse model of PQ‑induced pulmonary fibrosis following their transplantation, markedly improving the survival rate of mice with PQ poisoning. In addition, the results indicated that BMSC transplantation may inhibit the production of pro‑inflammatory cytokines, including tumor necrosis factor‑α interleukin (IL)‑1β, IL‑6 and IL‑10 in the lung tissues of PQ‑poisoned mice, and ultimately attenuate the pulmonary fibrosis. In vitro, BMSCs may suppress PQ‑induced epithelial‑to‑mesenchymal transition and protect pulmonary epithelial cells from PQ‑induced apoptosis. These findings suggest that BMSC transplantation may be a promising treatment for pulmonary fibrosis induced by PQ poisoning.
Collapse
Affiliation(s)
- Jianjun Chen
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Linjie Si
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Liangliang Zhou
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Yijun Deng
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| |
Collapse
|
19
|
Bufei Jianpi Granules Reduce Quadriceps Muscular Cell Apoptosis by Improving Mitochondrial Function in Rats with Chronic Obstructive Pulmonary Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1216305. [PMID: 30723509 PMCID: PMC6339712 DOI: 10.1155/2019/1216305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/03/2018] [Accepted: 12/27/2018] [Indexed: 01/08/2023]
Abstract
Background Cell apoptosis is an important mechanism underlying skeletal muscle dysfunction in chronic obstructive pulmonary disease (COPD) patients, and mitochondrial dysfunction is recognized as a central aspect contributing to skeletal muscle deterioration. Bufei Jianpi granules have been confirmed effective for improving motor function in COPD patients, but the specific mechanism for this improved function remains unknown. This study explored the mechanisms by which Bufei Jianpi granules improve cell apoptosis and mitochondrial dysfunction in COPD. Methods Sprague-Dawley rats were randomized into control, model, Bufei Jianpi, and aminophylline groups. A stable COPD rat model was induced with respective repeated cigarette smoke inhalation and intragastric bacterial infection, and rats were sacrificed after 20 weeks; the quadriceps muscle was harvested from each rat. Skeletal muscle mitochondria were extracted for measurements of mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore openings (mPTPs). ATP levels were determined with a firefly luciferase-based ATP assay kit. The rates of cell apoptosis were determined by the transferase-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL) method. Cyto C and caspase-3 mRNA and protein levels were measured by qPCR and western blotting. Results ATP, MMP, and mPTPs were markedly decreased in COPD rats, while cell apoptosis, caspase-3, and Cyto C were increased (P<0.01). All aforementioned parameters were improved in treatment groups (P<0.05). ATP, MMP, and mPTPs were significantly higher in the Bufei Jianpi group than in the aminophylline group, while cell apoptosis, caspase-3, and Cyto C were lower (P<0.05). Conclusions Bufei Jianpi granules can inhibit mitochondrial dysfunction and cell apoptosis in peripheral muscles, which might be the mechanism involved in improving skeletal muscle function in COPD patients.
Collapse
|
20
|
Li AY, Wang JJ, Yang SC, Zhao YS, Li JR, Liu Y, Sun JH, An LP, Guan P, Ji ES. Protective role of Gentianella acuta on isoprenaline induced myocardial fibrosis in rats via inhibition of NF-κB pathway. Biomed Pharmacother 2018; 110:733-741. [PMID: 30554111 DOI: 10.1016/j.biopha.2018.12.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 10/27/2022] Open
Abstract
Gentianella acuta (Michx.) Hulten (G. acuta) has been widely used in Mongolian medicines for the treatment of cardiovascular diseases in Ewenki and Oroqen, Inner Mongolia autonomous region, China. The aim of this study was to investigate the effects and related mechanism of G. acuta on isoproterenol (ISO)-induced oxidative stress, fibrosis, and myocardial damage in rats. Male Sprague Dawley rats were randomly divided into the normal control group, ISO induced group and ISO+G. acuta treatment group. Rats were administered with ISO subcutaneously (5 mg/kg/day) for 7 days, and were orally administered simultaneously with aqueous extracts of G. acuta for 21 days. This investigation showed G. acuta treatment ameliorated cardiac structural disorder, excessive collagenous fiber accumulation and cardiac malfunction. Compared with the ISO induced model group, G. acuta treatment increased superoxide dismutase (SOD) activities and glutathione (GSH) level, prevented the rise of malondialdehyde (MDA), and decreased hydroxyproline contents in the heart tissues. Moreover, G. acuta reduced the expression of transforming growth factor β1 (TGF-β1) and connective tissue growth factor (CTGF), and inhibited the expression and activation of NF-κB-P65 in myocardial tissues. These results suggested that G. acuta protects against ISO-induced cardiac malfunction probably by preventing oxidative stress, and fibrosis, and the mechanism might be through inhibiting NF-κB pathway.
Collapse
Affiliation(s)
- Ai-Ying Li
- Hebei key laboratory of Chinese medicine research on cardio-cerebrovascular disease and Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Jing-Jing Wang
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Sheng-Chang Yang
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Ya-Shuo Zhao
- Scientific Research Center, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Jie-Ru Li
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Yu Liu
- Hebei key laboratory of Chinese medicine research on cardio-cerebrovascular disease and Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Jia-Huan Sun
- Hebei key laboratory of Chinese medicine research on cardio-cerebrovascular disease and Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Li-Ping An
- Hebei key laboratory of Chinese medicine research on cardio-cerebrovascular disease and Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - Peng Guan
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China
| | - En-Sheng Ji
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
21
|
miRNA-1246 suppresses acute lung injury-induced inflammation and apoptosis via the NF-κB and Wnt/β-catenin signal pathways. Biomed Pharmacother 2018; 108:783-791. [DOI: 10.1016/j.biopha.2018.09.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/22/2018] [Accepted: 09/08/2018] [Indexed: 02/03/2023] Open
|
22
|
Li J, Li Y, Lu X, Wang H, Wang Y, Li H, Wu Z. Dynamic Characteristics of Sequential Acute Exacerbations and Risk Windows in AECOPD Rats Induced by Cigarette-Smoke and Exposure to Klebsiella pneumoniae. Biol Pharm Bull 2018; 41:1543-1553. [PMID: 30058599 DOI: 10.1248/bpb.b18-00148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The risk-window (RW) of chronic obstructive pulmonary disease (COPD) is a period after an acute exacerbation (AE) but before the following stable phase, in which exacerbations are easy to relapse. We established a sequential COPD-AE-RW rat model by cigarette-smoke and bacterial exposures in the first 8 weeks, and was challenged with Klebsiella pneumonia to mimic an AE on Day 1 of week 9, and found that body temperature, white blood cell, neutrophils, serum amyloid A (SAA) and C-reactive protein (CRP) increased in AECOPD rats 24 h after challenge, and declined in 3-6 d, while lung function declined in 48 h, and recovered in 7-16 d. When sacrificed, pulmonary forced expiratory volume (FEV)100 and FEV300 decreased, while elevated bronchoalveolar lavage fluid (BALF) neutrophils and marked airway inflammation, remodeling and emphysema were observed. Sequential COPD-AE-RW rat model was established successfully and AE phase lasts for approximately 5-7 d, followed by a 10-d around risk-window.
Collapse
Affiliation(s)
- Jiansheng Li
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Institute for Geriatrics, Henan University of Traditional Chinese Medicine (TCM).,Institute for Respiratory Diseases and the Level Three Laboratory of Respiration Pharmacology of TCM, the First Affiliated Hospital, Henan University of TCM
| | - Ya Li
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Institute for Respiratory Diseases and the Level Three Laboratory of Respiration Pharmacology of TCM, the First Affiliated Hospital, Henan University of TCM.,Central Laboratory, the First Affiliated Hospital, Henan University of TCM
| | - Xiaofan Lu
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Respiratory Department, the Second Clinical Medical College, Henan University of Chinese Medicine
| | - Haifeng Wang
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of TCM
| | - Yang Wang
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of TCM
| | - Hangjie Li
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM).,Department of Respiratory Diseases, the Chinese Medicine Hospital of Xuchang
| | - Zhaohuan Wu
- Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development of Henan University of Traditional Chinese Medicine (TCM)
| |
Collapse
|
23
|
Zhao P, Li J, Tian Y, Mao J, Liu X, Feng S, Li J, Bian Q, Ji H, Zhang L. Restoring Th17/Treg balance via modulation of STAT3 and STAT5 activation contributes to the amelioration of chronic obstructive pulmonary disease by Bufei Yishen formula. JOURNAL OF ETHNOPHARMACOLOGY 2018; 217:152-162. [PMID: 29454913 DOI: 10.1016/j.jep.2018.02.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 02/05/2018] [Accepted: 02/14/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Bufei Yishen formula (BYF), a Traditional Chinese Medicine (TCM), has been extensively applied in clinical treatment of chronic obstructive pulmonary disease (COPD) and provides an effective treatment strategy for the syndrome of lung-kidney qi deficiency in COPD patients. Here, we investigated its anti-COPD mechanism in COPD rats in relation to the balance between T helper (Th) 17 cells and regulatory T (Treg) cells. METHODS Rat model of cigarette smoke- and bacterial infection-induced COPD was established, and orally treated with BYF for 12 consecutive weeks. Then, the rats were sacrificed, their lung tissues were removed for histological analysis, and spleens and mesenteric lymph nodes (MLNs) were collected to evaluate the Th17 and Treg cells. RESULTS Oral treatment of BYF markedly suppressed the disease progression and alleviated the pathological changes of COPD. It also decreased the bronchoalveolar lavage fluid (BALF) levels of pro-inflammatory cytokines, including IL-1β, IL-6, TNF-α and Th17-related IL-17A, and induced a significant increase in Treg-related IL-10. Furthermore, BYF treatment obviously decreased the proportion of CD4+RORγt+ T (Th17) cell and increased the proportion of CD4+CD25+Foxp3+ T (Treg) cell, leading to restore the Th17/Treg balance. BYF treated groups also decreased RORγt and increased Foxp3 expression in the spleens and MLNs. BYF further inhibited the phosphorylation of signal transducer and activator of transcription-3 (STAT3) and boosted the phosphorylation of STAT5, that were critical transcription factors for TH17 and Treg differentiation. CONCLUSION these results demonstrated that BYF exerted its anti-COPD efficacy by restoring Th17/Treg balance via reciprocally modulating the activities of STAT3 and STAT5 in COPD rats, which may help to elucidate the underlying immunomodulatory mode of BYF on COPD treatment.
Collapse
MESH Headings
- Animals
- Cigarette Smoking/adverse effects
- Cytokines/metabolism
- Disease Models, Animal
- Disease Progression
- Drugs, Chinese Herbal/pharmacology
- Female
- Immunologic Factors/pharmacology
- Inflammation Mediators/metabolism
- Klebsiella pneumoniae/pathogenicity
- Lung/drug effects
- Lung/metabolism
- Lung/pathology
- Male
- Phenotype
- Phosphorylation
- Pulmonary Disease, Chronic Obstructive/etiology
- Pulmonary Disease, Chronic Obstructive/immunology
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/prevention & control
- Rats, Sprague-Dawley
- STAT3 Transcription Factor/metabolism
- STAT5 Transcription Factor/metabolism
- Signal Transduction/drug effects
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Jing Mao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Suxiang Feng
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Junzi Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Qingqing Bian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Huige Ji
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Lanxi Zhang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| |
Collapse
|
24
|
Cui P, Xin H, Yao Y, Xiao S, Zhu F, Gong Z, Tang Z, Zhan Q, Qin W, Lai Y, Li X, Tong Y, Xia Z. Human amnion-derived mesenchymal stem cells alleviate lung injury induced by white smoke inhalation in rats. Stem Cell Res Ther 2018; 9:101. [PMID: 29650044 PMCID: PMC5898065 DOI: 10.1186/s13287-018-0856-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/17/2018] [Accepted: 03/26/2018] [Indexed: 02/08/2023] Open
Abstract
Background White smoke inhalation (WSI) is an uncommon but potentially deadly cause of acute lung injury and acute respiratory distress syndrome for which no effective pharmaceutical treatment has been developed. This study aimed to determine the protective effects of human amnion-derived mesenchymal stem cells (hAMSCs) against WSI-induced lung injury in rats. Methods hAMSCs were injected into rats via the tail vein 4 h after WSI. At 1, 3, 7, 14, and 28 days after cell injection, hAMSCs labeled with PKH26 in lung, heart, liver, and kidney tissues were observed by fluorescence microscopy. The lung injury score was determined by hematoxylin and eosin staining. Lung fibrosis was assessed by Masson’s trichrome staining. The computed tomography (CT) score was assessed by CT scanning. The wet/dry weight ratio was calculated. The levels of interleukin (IL)-1β, IL-6, and IL-10 were determined by enzyme-linked immunosorbent assays. The expression of surfactant protein (SP)-A, SP-C, and SP-D was measured by Western blotting. Results The injected hAMSCs were primarily distributed in the lung tissues in WSI-induced rats. Compared with the model and phosphate-buffered saline (PBS) group, hAMSC treatment led to reduced lung injury, lung fibrosis, CT score, and inflammation levels in WSI-induced mice. hAMSC treatment also resulted in increased cell retention in the lung, partial pressure of oxygen (PaO2), and PaO2/fraction of inspired oxygen (FiO2) levels, and pulmonary SP-A, SP-C, and SP-D expression compared with that in the model and PBS group. Conclusions hAMSCs are a potential cell-based therapy for WSI-induced lung injury.
Collapse
Affiliation(s)
- Pei Cui
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Haiming Xin
- Department of Burns, Plastic and Wound repair surgery, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Yongming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, 100048, People's Republic of China
| | - Shichu Xiao
- Department of Burn surgery, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Feng Zhu
- Department of Burn surgery, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Zhenyu Gong
- Department of Burns, Plastic and Wound repair surgery, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Zhiping Tang
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Qiu Zhan
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Wei Qin
- Department of Burns, Plastic and Wound repair surgery, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Yanhua Lai
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Xiaohui Li
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China
| | - Yalin Tong
- Research Laboratory of Burns and Trauma, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China. .,Department of Burns, Plastic and Wound repair surgery, the 181st Hospital of Chinese PLA, Guilin, 541002, People's Republic of China.
| | - Zhaofan Xia
- Department of Burn surgery, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
25
|
Wu J, Li X, Qin Y, Cheng J, Hao G, Jin R, Zhu C. Jinwei Tang modulates HDAC2 expression in a rat model of COPD. Exp Ther Med 2018; 15:2604-2610. [PMID: 29456664 DOI: 10.3892/etm.2018.5707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 04/07/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect of a Traditional Chinese Herbal Medicine (TCHM), named Jinwei Tang on histone deacetylase 2 (HDAC2) and its role in the regulation of corticosteroid resistance in a rat model of chronic obstructive pulmonary disease (COPD). Male Wistar rats were divided into five groups (each n=10): COPD group, established by the intratracheal instillation of lipopolysaccharide and passive smoke exposure, and control, budesonide, theophylline + budesonide and Jinwei Tang + budesonide groups. Lung function was measured, lung tissue histopathology was examined and HDAC2 expression in the lung was assessed by immunohistochemistry. In addition, protein levels of interleukin-8 (IL-8), tumor necrosis factor (TNF)-α and HDAC2 in lung homogenate were quantified by ELISA. The rat COPD model exhibited alterations of the ratio of forced expiratory volume in 0.2 sec (FEV0.2) to the forced vital capacity, FEV0.2, dynamic compliance and airway resistance. HDAC2 expression was markedly reduced in the lung tissue of the COPD group compared with the control group, and treatment with Jinwei Tang + budesonide or theophylline + budesonide resulted in significant attenuation of the reduction of HDAC2 expression in the lungs (P<0.05). However, treatment with budesonide alone did not significantly alter HDAC2 expression. In the Jinwei Tang + budesonide and theophylline + budesonide groups, IL-8 and TNF-α expression was significantly decreased (P<0.05) and the HDAC2 level increased (P<0.05) compared with that in the COPD group. In conclusion, Jinwei Tang modulates airway inflammation and may enhance the anti-inflammatory effect of glucocorticoid through the upregulation of HADC2 expression in a rat model of COPD.
Collapse
Affiliation(s)
- Jianjun Wu
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Xin Li
- Department of Ocular Diseases, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, P.R. China
| | - Yang Qin
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Juan Cheng
- Department of Pathology, Dongzhimen Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100700, P.R. China
| | - Gaimei Hao
- Department of Pathology, Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Ruifeng Jin
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Chenjun Zhu
- Department of Encephalopathy, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| |
Collapse
|
26
|
Long-Term Effects of TCM Yangqing Kangxian Formula on Bleomycin-Induced Pulmonary Fibrosis in Rats via Regulating Nuclear Factor- κB Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:2089027. [PMID: 29387126 PMCID: PMC5745787 DOI: 10.1155/2017/2089027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/04/2017] [Accepted: 10/30/2017] [Indexed: 12/28/2022]
Abstract
Objective We aimed to evaluate the therapeutic effects and long-term effects of YKF and dissect the potential mechanisms. Materials and Methods IPF rats were given YKF, prednisone, or pirfenidone, respectively, from day 1 to day 42, followed by a 28-day nonintervention interval through day 70. Forced vital capacity (FVC), histopathology, hydroxyproline (HYP) contents, lung coefficient, blood inflammatory cell populations, inflammatory cytokine levels of the lung tissues, and the expression of proteins involved in nuclear factor- (NF-) κB signaling pathway were evaluated on days 7, 14, 28, 42, and 70. Results HYP contents, Ashcroft scores, lung coefficient, and pulmonary fibrosis blood cell populations increased significantly in IPF rats, while FVC declined. All the above-mentioned parameters were improved in treatment groups from day 7 up to day 70, especially in YKF group. The mRNA and protein expressions of tumor necrosis factor- (TNF-) α significantly decreased, while interferon- (IFN-) γ significantly increased, and phosphorylations of cytoplasm inhibitor of nuclear factor kappa-B kinase β (IKKβ), inhibitor of nuclear factor kappa-B α (IκBα), and NF-κB were obviously downregulated in YKF group from day 7 to day 70. Conclusion YKF has beneficial protective and long-term effects on pulmonary fibrosis by anti-inflammatory response and alleviating fibrosis.
Collapse
|
27
|
Tian Y, Li Y, Li J, Feng S, Li S, Mao J, Xie Y, Liu X, Dong H, Zheng W, Wang M. Bufei Yishen Granules Combined with Acupoint Sticking Therapy Suppress Inflammation in Chronic Obstructive Pulmonary Disease Rats: Via JNK/p38 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2017; 2017:1768243. [PMID: 29234369 PMCID: PMC5682917 DOI: 10.1155/2017/1768243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/16/2017] [Accepted: 10/02/2017] [Indexed: 12/14/2022]
Abstract
The present study was initiated to explore the mechanism of the effects of Bufei Yishen granules combined with acupoint sticking therapy (Shu-Fei Tie) on inflammation regulated by c-Jun N-terminal kinase (JNK) and p38 MAPK signaling in COPD rats. Seventy-two rats were divided into healthy control (Control), Model, Bufei Yishen (BY), acupoint sticking (AS), Bufei Yishen + acupoint sticking (BY + AS), and aminophylline (APL) groups (n = 12 each). COPD rats were exposed to cigarette smoke and bacteria and were given the various treatments from weeks 9 through 20; all animals were sacrificed at the end of week 20. MCP-1, IL-2, IL-6, and IL-10 concentrations in BALF and lung tissue as well as JNK and p38 mRNA and protein levels in lung were measured. The results showed that all the four treatment protocols (BY, AS, BY + AS, and APL) markedly reduced the concentrations of IL-2, IL-6, and MCP-1 and levels of JNK and p38 MAPK mRNA, and the effects of Bufei Yishen granules combined with acupoint sticking therapy were better than acupoint sticking therapy only and aminophylline. In conclusion, the favorable effect of Bufei Yishen granules combined with Shu-Fei Tie may be due to decreased inflammation through regulation of the JNK/p38 signaling pathways.
Collapse
Affiliation(s)
- Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Ya Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Central Laboratory and Respiratory Pharmacological Laboratory of Chinese Medicine, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute of Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Suxiang Feng
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Suyun Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute of Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Jing Mao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Yang Xie
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute of Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Haoran Dong
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Wanchun Zheng
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Minghang Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute of Respiratory Diseases, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450008, China
| |
Collapse
|
28
|
Integrating 3-omics data analyze rat lung tissue of COPD states and medical intervention by delineation of molecular and pathway alterations. Biosci Rep 2017; 37:BSR20170042. [PMID: 28450497 PMCID: PMC5479022 DOI: 10.1042/bsr20170042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 12/30/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious health problem. However, the molecular pathogenesis of COPD remains unknown. Here, we explored the molecular effects of cigarette smoke and bacterial infection in lung tissues of COPD rats. We also investigated therapeutic effects of aminophylline (APL) on the COPD rats and integrated transcriptome, proteome, and metabolome data for a global view of molecular mechanisms of COPD progression. Using molecular function and pathway analyses, the genes and proteins regulated in COPD and APL-treated rats were mainly attributed to oxidoreductase, antioxidant activity, energy and fatty acid metabolism. Furthermore, we identified hub proteins such as Gapdh (glyceraldehyde-3-phosphate dehydrogenase), Pkm (pyruvate kinase isozymes M1/M2), and Sod1 (superoxide dismutase 1), included in energy metabolism and oxidative stress. Then, we identified the significantly regulated metabolic pathways in lung tissues of COPD- and APL-treated rats, such as arachidonic acid, linoleic acid, and α-linolenic acid metabolism, which belong to the lipid metabolism. In particular, we picked the arachidonic acid metabolism for a more detailed pathway analysis of transcripts, proteins, and metabolites. We could observe an increase in metabolites and genes involved in arachidonic acid metabolism in COPD rats and the decrease in these in APL-treated rats, suggesting that inflammatory responses were up-regulated in COPD rats and down-regulated in APL-treated rats. In conclusion, these system-wide results suggested that COPD progression and its treatment might be associated with oxidative stress, lipid and energy metabolism disturbance. Additionally, we demonstrated the power of integrated omics for the elucidation of genes, proteins, and metabolites’ changes and disorders that were associated with COPD.
Collapse
|
29
|
da Silva IRV, de Araujo CLP, Dorneles GP, Peres A, Bard AL, Reinaldo G, Teixeira PJZ, Lago PD, Elsner VR. Exercise-modulated epigenetic markers and inflammatory response in COPD individuals: A pilot study. Respir Physiol Neurobiol 2017; 242:89-95. [PMID: 28435027 DOI: 10.1016/j.resp.2017.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/20/2017] [Accepted: 04/14/2017] [Indexed: 12/17/2022]
Abstract
The study investigated the effects of exercise on epigenetic signals and systemic cytokine levels in chronic obstructive pulmonary disease (COPD) individuals. Ten participants of a pulmonary rehabilitation program were submitted to 24 sessions of a supervisioned exercise protocol thrice-weekly (90min/session). Blood samples were collected at baseline, after the 1st session, before and after the 24th session. A DNA hypomethylation status was observed after the 1st session when compared at baseline, while global histone H4 acetylation status was unaltered in any time-points evaluated. No significant changes were observed on cytokine levels after the 1st session. A significant enhancement on interleukin 6 (IL-6) and a decrease on transforming growth factor-beta (TGF-β) levels were found after the 24th session when compared to the pre 24th session. Moreover, 23 sessions of exercise were able to diminish significantly the basal levels of IL-6 and interleukin 8 (IL-8). These data suggest a potential role of epigenetic machinery in mediating the anti-inflammatory effects of exercise in COPD patients.
Collapse
Affiliation(s)
- Ivy Reichert Vital da Silva
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | | | - Gilson Pires Dorneles
- Cellular and Molecular Immunology Lab., Department of Health Basic Sciences,Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
| | - Alessandra Peres
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil; Cellular and Molecular Immunology Lab., Department of Health Basic Sciences,Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
| | - Andreia Luciana Bard
- Curso de Fisioterapia do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - Gustavo Reinaldo
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
| | - Paulo José Zimermann Teixeira
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil; Médico responsável pelo Serviço de Reabilitação Pulmonar do Pavilhão Pereira Filho do Hospital Santa Casa de Misericórdia de Porto Alegre, RS, Brazil
| | - Pedro Dal Lago
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
| | - Viviane Rostirola Elsner
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil.
| |
Collapse
|
30
|
Mei X, Wang HX, Li JS, Liu XH, Lu XF, Li Y, Zhang WY, Tian YG. Dusuqing granules (DSQ) suppress inflammation in Klebsiella pneumonia rat via NF-κB/MAPK signaling. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:216. [PMID: 28415997 PMCID: PMC5392945 DOI: 10.1186/s12906-017-1736-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 04/08/2017] [Indexed: 01/31/2023]
Abstract
Background Dusuqing granules (DSQ) have been used in the treatment of bacterial pneumonia clinically, with remarkable benefits. This study was initiated to explore the effects of DSQ on pulmonary inflammation by regulating nuclear factor (NF)-κB/mitogen-activated protein kinase (MAPK) signaling in bacterial pneumonia rats. Methods Rat model was duplicated with Klebsiella pneumonia by a one-time intratracheal injection. Rats were randomized into control, model, DSQ and levofloxacin (LVX) groups. After administrated with appropriate medicines for 7 days, lung tissues were harvested and prepared for pathological analysis, and interleukin (IL)-1, IL-6, monocyte chemotactic protein (MCP)-1and macrophage inflammatory protein (MIP)-2 detections. NF-κB mRNA was measured by real-time qPCR, and the phosphorylation and total proteins of P38MAPK, JNK46/54, ERK42/44 were determined by Western blotting. Results Marked pathological impairments were observed in model rats, whereas were improved in DSQ group. The cytokines levels, NF-κB mRNA expression and the phosphorylation of P38MAPK, JNK46/54 and ERK42/44 proteins were significantly higher in model group, and were significantly depressed in DSQ group. Conclusion The protective effects of DSQ on Klebsiella pneumonia might be attributed to its inactivative effects of NF-κB/ MAPK pathway.
Collapse
|
31
|
Li Y, Tian YG, Li JS, Dong YQ, Wang MH, Feng SX, Li LL, Mao J, Wang LL, Luo S. Bufei Yishen granules combined with acupoint sticking therapy suppress oxidative stress in chronic obstructive pulmonary disease rats: Via regulating peroxisome proliferator-activated receptor-gamma signaling. JOURNAL OF ETHNOPHARMACOLOGY 2016; 193:354-361. [PMID: 27562320 DOI: 10.1016/j.jep.2016.08.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 08/11/2016] [Accepted: 08/20/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) is clinically used under the guidance of its unique theory system. Bufei Yishen (BY) granules, an oral Chinese herbal formula, is confirmed effective for treating the syndrome of lung-kidney qi deficiency in chronic obstructive pulmonary disease (COPD) patients. Shu-Fei Tie ointment is another prescription for acupoint sticking (AS) therapy based on the theory of treating an internal disease by external treatment on proper acupoints. The beneficial effects of BY granules combined with Shu-Fei Tie have been proved in previous clinical trials. However, the underlying mechanism remains unclear. The present study was initiated to explore the antioxidative mechanism of the integrated therapy of BY granules and acupoint sticking via regulating by peroxisome proliferator activated receptor-gamma (PPARγ) signaling in a cigarette-smoke/bacterial exposure induced COPD rat model. MATERIALS AND METHODS Rats were randomized into Control, Model, BY, AS, BY+AS and aminophylline (APL) groups. COPD rats were induced by cigarette-smoke and bacterial exposures, and were administrated with normal saline, BY granules, AS, BY+AS or aminophylline from week 9 and sacrificed at week 20. Activity of superoxide dismutase (SOD) and levels of methane dicarboxylic aldehyde (MDA) in peripheral blood and bronchoalveolar lavage fluid (BALF) were determined by hydroxylamine and thiobarbituric acid methods. The gene and protein expressions of PPARγ in the lung tissues were analyzed by quantitative polymerase chain reaction and western blot. RESULTS Serum and BALF SOD decreased significantly in Model group (P<0.01), while MDA increased (P<0.01). Compared to COPD rats, serum SOD was higher in all treatment groups (P<0.01), and BALF SOD was higher in BY and BY+AS groups (P<0.01); serum and BALF MDA was lower in all treatment groups (P<0.01). Serum and BALF SOD was higher in BY+AS group than in AS group, while MDA was lower (P<0.05). BALF SOD increased in BY+AS group compared with APL group, while MDA decreased (P<0.05). PPARγ mRNA and protein and the phosphorylation of PPARγ (p-PPARγ) decreased in COPD rats (P<0.01), and increased in all treatment groups (P<0.01). PPARγ mRNA was higher in BY+AS group than in AS group (P<0.05), PPARγ and p-PPARγ were higher in BY+AS group than in AS and APL groups (P<0.05, P<0.01); PPARγ protein was higher in BY group than in APL group (P<0.05). CONCLUSION Bufei Yishen granules, Shu-Fei Tie and their combination have beneficial effects in stable COPD, and can attenuate the oxidative stress, and the activation of PPARγ signaling might be involved in the underlying mechanisms, but there are no obvious synergistic effect of Bufei Yishen granules and Shu-Fei Tie.
Collapse
Affiliation(s)
- Ya Li
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China; Central Laboratory, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China; The Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development in Henan Province, Zhengzhou, Henan 450046, China.
| | - Yan-Ge Tian
- The Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development in Henan Province, Zhengzhou, Henan 450046, China; Institute for Geriatrics, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Jian-Sheng Li
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China; The Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development in Henan Province, Zhengzhou, Henan 450046, China; Institute for Geriatrics, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Yu-Qiong Dong
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China.
| | - Ming-Hang Wang
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China; The Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development in Henan Province, Zhengzhou, Henan 450046, China.
| | - Su-Xiang Feng
- The Collaborative Innovation Center for Respiratory Diseases Diagnostics, Treatment and New Drug Research and Development in Henan Province, Zhengzhou, Henan 450046, China.
| | - Lin-Lin Li
- Institute for Geriatrics, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| | - Jing Mao
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China.
| | - Li-Li Wang
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China.
| | - Shan Luo
- Institute for Respiratory Diseases, the First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China.
| |
Collapse
|
32
|
Tian Y, Li J, Li Y, Dong Y, Yao F, Mao J, Li L, Wang L, Luo S, Wang M. Effects of Bufei Yishen Granules Combined with Acupoint Sticking Therapy on Pulmonary Surfactant Proteins in Chronic Obstructive Pulmonary Disease Rats. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8786235. [PMID: 27699176 PMCID: PMC5028822 DOI: 10.1155/2016/8786235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/16/2016] [Indexed: 12/22/2022]
Abstract
Our previous studies have demonstrated the beneficial effects of Bufei Yishen granules combined with acupoint sticking therapy (the integrated therapy) in chronic obstructive pulmonary disease (COPD), but the underlying mechanism remains unclear. Dysfunction of pulmonary surfactant proteins (SPs, including SP-A, SP-B, SP-C, and SP-D) may be included in pathophysiology of COPD. This study aimed to explore the mechanism of the integrated therapy on SPs. COPD rat models were established. The treatment groups received Bufei Yishen granules or acupoint sticking or their combination. Using aminophylline as a positive control drug. The levels of SPs in serum, BALF, and lung were measured. The results showed that the integrated therapy markedly reduced the levels of SPs in serum and increased these indicators in the lung. The integrated therapy was better than aminophylline in reducing the levels of SPs and was better than Bufei Yishen granules in reducing SP-A, SP-C, and SP-D in serum. The integrated therapy was better than aminophylline and Bufei Yishen granules in increasing SP-A, SP-B, and SP-D mRNA in the lung. SP-A and SP-D in BALF were positively correlated with PEF and EF50. The levels of SPs are associated with airway limitation. The beneficial effects of the integrated therapy may be involved in regulating pulmonary surfactant proteins.
Collapse
Affiliation(s)
- Yange Tian
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Jiansheng Li
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Ya Li
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
- Central Laboratory and Respiratory Pharmacological Laboratory of Chinese Medicine, The First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450008, China
| | - Yuqiong Dong
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Fengjia Yao
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Jing Mao
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Linlin Li
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Lili Wang
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Shan Luo
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Minghang Wang
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan 450046, China
- Institute for Geriatrics, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, China
- Institute for Respiratory Diseases, The First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450008, China
| |
Collapse
|
33
|
Sequential Treatments with Tongsai and Bufei Yishen Granules Reduce Inflammation and Improve Pulmonary Function in Acute Exacerbation-Risk Window of Chronic Obstructive Pulmonary Disease in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1359105. [PMID: 27563333 PMCID: PMC4983671 DOI: 10.1155/2016/1359105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022]
Abstract
Background. Sequential treatments of Chinese medicines for acute exacerbation of chronic obstructive pulmonary disease (AECOPD) risk window (RW) have benefits for preventing reoccurrences of AEs; however, the effects on pulmonary function, pulmonary, and systemic inflammatory biomarkers remain unclear. Methods. Cigarette-smoke/bacterial infections induced rats were randomized into Control, COPD, AECOPD, Tongsai Granule/normal saline (TSG/NS), moxifloxacin + salbutamol/NS (MXF+STL/NS), TSG/Bufei Yishen Granule (BYG), MXF+STL/STL, and TSG+MXF+STL/BYG+STL groups and given corresponding medicine(s) in AE- and/or RW phase. Body temperature, pulmonary function, blood cytology, serum amyloid A (SAA) and C-reactive protein (CRP), pulmonary histomorphology and myeloperoxidase (MPO), polymorphonuclear (PMN) elastase, interleukins IL-1β, IL-6, and IL-10, and tumor necrosis factor- (TNF-) α expressions were determined. Results. Body temperature, inflammatory cells and cytokines, SAA, CRP, and pulmonary impairment were higher in AECOPD rats than stable COPD, while pulmonary function declined and recovered to COPD level in 14–18 days. All biomarkers were improved in treated groups with shorter recovery times of 4–10 days, especially in TSG+MXF+STL/BYG+STL group. Conclusion. Sequential treatments with Tongsai and Bufei Yishen Granules, during AECOPD-RW periods, can reduce inflammatory response and improve pulmonary function and shorten the recovery courses of AEs, especially the integrated Chinese and Western medicines.
Collapse
|
34
|
Evidences of Herbal Medicine-Derived Natural Products Effects in Inflammatory Lung Diseases. Mediators Inflamm 2016; 2016:2348968. [PMID: 27445433 PMCID: PMC4942669 DOI: 10.1155/2016/2348968] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022] Open
Abstract
Pulmonary inflammation is a hallmark of many respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory syndrome distress (ARDS). Most of these diseases are treated with anti-inflammatory therapy in order to prevent or to reduce the pulmonary inflammation. Herbal medicine-derived natural products have been used in folk medicine and scientific studies to evaluate the value of these compounds have grown in recent years. Many substances derived from plants have the biological effects in vitro and in vivo, such as flavonoids, alkaloids, and terpenoids. Among the biological activities of natural products derived from plants can be pointed out the anti-inflammatory, antiviral, antiplatelet, antitumor anti-allergic activities, and antioxidant. Although many reports have evaluated the effects of these compounds in experimental models, studies evaluating clinical trials are scarce in the literature. This review aims to emphasize the effects of these different natural products in pulmonary diseases in experimental models and in humans and pointing out some possible mechanisms of action.
Collapse
|
35
|
Liu Z, Li W, Lv J, Xie R, Huang H, Li Y, He Y, Jiang J, Chen B, Guo S, Chen L. Identification of potential COPD genes based on multi-omics data at the functional level. MOLECULAR BIOSYSTEMS 2016; 12:191-204. [DOI: 10.1039/c5mb00577a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A novel systematic approach MMMG (Methylation–MicroRNA–MRNA–GO) to identify potential COPD genes and their classifying performance evaluation.
Collapse
Affiliation(s)
- Zhe Liu
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Wan Li
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Junjie Lv
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Ruiqiang Xie
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Hao Huang
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Yiran Li
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Yuehan He
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Jing Jiang
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Binbin Chen
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Shanshan Guo
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| | - Lina Chen
- College of Bioinformatics Science and Technology
- Harbin Medical University
- Harbin
- China
| |
Collapse
|
36
|
Dong Y, Li Y, Sun Y, Mao J, Yao F, Tian Y, Wang L, Li L, Li S, Li J. Bufei Jianpi granules improve skeletal muscle and mitochondrial dysfunction in rats with chronic obstructive pulmonary disease. Altern Ther Health Med 2015; 15:51. [PMID: 25888379 PMCID: PMC4378020 DOI: 10.1186/s12906-015-0559-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/18/2015] [Indexed: 12/31/2022]
Abstract
Background Bufei Jianpi granules has been confirmed effective in improving pulmonary function, alleviating acute exacerbations, improving six-minute walk distance and quality of life, and benefited in 12-month follow-up in chronic obstructive pulmonary disease (COPD) patients with syndrome of lung-spleen qi deficiency. Skeletal muscle dysfunction (SMD), an important extrapulmonary complication, occurs in the very initiation of COPD and is closely related to morbidity and mortality. To evaluate the efficacy of Bufei Jianpi granules on SMD, we observed skeletal muscular function and histomorphology, mitochondrial morphormetry and proteins in COPD rats induced by cigarette-smoke and Klebsiella pneumoniae. Methods Seventy-two Sprague–Dawley rats were randomized into Control + Saline, Control + Bufei Jianpi, Control + Aminophylline, COPD + Saline, COPD + Bufei Jianpi and COPD + Aminophylline groups. From week 9 to 20, rats were administrated intragastricly by normal saline, Bufei Jianpi granules and aminophylline, respectively. Muscular tension and fatigue index of intercostal muscle, quadriceps, biceps and soleus were detected by using electrophysiological technology. Pathological and ultrastructural changes and expressions of mitochondrial Bcl-2 nineteen-kilodalton interacting protein 3 (Bnip3) and cytoplasm cytochrome C (Cyto C) in the four skeletal muscles were observed by using optical and electron microscope and western blotting. Results There was no statistical difference among the control rats treated with saline, Bufei Jianpi granules or aminophylline in above-mentioned parameters. Muscular tension, mitochondria volume density (Vv) and compared membrane surface (δm) of the four muscles were significantly lower in COPD + Saline group compared to Control + Saline group, while fatigue index, mitochondria surface area (δ), Bnip3 and Cyto C were higher (P < 0.05). COPD rats showed more morphological changes in muscle tissues than controls, such as atrophy, degeneration, necrosis and matrix hyperplasia. Utrastructurally, mitochondria populations decreased significantly in the four muscles, and were shrunken and even cavitation changed. The up-mentioned parameters were improved in Bufei Jianpi group (P < 0.05) in the four muscles. Conclusions Bufei Jianpi granules can improve skeletal muscle function via improving mitochondria population and function, reducing apoptotic factors such as Bnip3 and Cyto C, and is more effective than aminophylline.
Collapse
|
37
|
Qi Y, Shang JY, Ma LJ, Sun BB, Hu XG, Liu B, Zhang GJ. Inhibition of AMPK expression in skeletal muscle by systemic inflammation in COPD rats. Respir Res 2014; 15:156. [PMID: 25481457 PMCID: PMC4265440 DOI: 10.1186/s12931-014-0156-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/24/2014] [Indexed: 11/26/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a disease characterized by airflow limitation and inflammation. Meanwhile, COPD also is associated with metabolic disorders, such as skeletal muscle weakness. Strikingly, activation of AMP-activated protein kinase (AMPK) exerts critical roles in energy metabolism. However, it remains unclear whether and how the expression levels of AMPK are affected in the COPD model rats which may lead to the dysfunction of the skeletal muscle in these rats. Methods Here we developed a rat model of COPD, and we investigated the morphological changes of peripheral skeletal muscle and measured the levels of tumor necrosis factor -α (TNF-α) and AMPK in skeletal muscle by using approaches that include immunohistochemistry and polymerase chain reaction (PCR). Results We found that the expression levels of both AMPK mRNA and protein in skeletal muscles were significantly reduced in the COPD model rats, in comparison to those from the control rats, the COPD model rats that received treatments with AICAR and resveratrol, whereas the expression levels of TNF-α were elevated in COPD rats. Conclusion Such findings indicate that AMPK may serve as a target for therapeutic intervention in the treatment of muscle weakness in COPD patients.
Collapse
Affiliation(s)
- Yong Qi
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Jun-yi Shang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Li-jun Ma
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Bei-bei Sun
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Xin-gang Hu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Bao Liu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital (He'nan Provincial People's Hospital), 7 Weiwu Road, ZhengZhou, 450003, China.
| | - Guo-jun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, ZhengZhou, 450003, China.
| |
Collapse
|
38
|
Ren J, An HY. Effects of ROCK inhibitor Y-27632 on TGF-β1/CTGF pathway. Shijie Huaren Xiaohua Zazhi 2014; 22:3932-3936. [DOI: 10.11569/wcjd.v22.i26.3932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Y-27632, a pyrimidine derivative, is a recently developed synthetic specific inhibitor of Rho associated coiled-coil forming protein kinase (ROCK), and it inhibits the process of hepatic fibrosis by regulating a variety of biological effects mediated by ROCK. Recent studies have found that the transforming growth factor β1 (TGF-β1)/connective tissue growth factor (CTGF) signaling pathway is involved in liver fibrosis. TGF-β1 induces the expression of its downstream molecule CTGF, resulting in the increase of extracellular matrix and liver fibrosis. Y-27632 can inhibit the expression of TGF-β1 and CTGF. This paper attempts to explain the anti-fibrosis effect of Y-27632 in terms of the impact of Y-27632 on the TGF-β1/CTGF pathway, with an aim to better understand the functional target of Y-27632 and provide a theoretical basis for the targeted therapy of liver fibrosis.
Collapse
|