1
|
Liu Y, Zhang L, Zhao J, Lu R, Shao X, Xu K, Li J, Tian Y. Effective-Component Compatibility of Bufei Yishen Formula III Suppresses Mitochondrial Oxidative Damage in COPD: Via Pkm2/Nrf2 Pathway. Int J Chron Obstruct Pulmon Dis 2024; 19:1905-1920. [PMID: 39206144 PMCID: PMC11352541 DOI: 10.2147/copd.s468825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose The main objective of this study was to explore the mechanism of effective component compatibility of Bufei Yishen formula III (ECC-BYF III) in inhibiting mitochondrial oxidative stress in a rat model of chronic obstructive pulmonary disease (COPD). Methods A549 cells exposed to cigarette smoke extract (CSE) were used to establish a model of mitochondrial oxidative damage. The cells were treated with the plasmid encoding Pkm2 and the enzymes and proteins involved in oxidative stress and mitochondrial function were measured. A rat model of COPD was established using CS and bacteria. Two different treatments were established, ECC-BYF III (5.5 mg/kg/d) and N-acetylcysteine (54 mg/kg/day). Animals were tested for pulmonary function (Vt, PEF, FVC, FEV0.1s and Cdyn) after eight weeks of therapy and were sacrificed. Pulmonary H&E staining was performed, and the total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) content were measured. The mitochondrial function was also examined. Furthermore, the Pkm2/Nrf2 signaling pathway was evaluated. Results Overexpression of Pkm2 dramatically ameliorated the CS-induced mitochondrial oxidative damage. Further studies indicated that ECC-BYF III significantly improved mitochondrial function and inhibited oxidative stress in the lung tissues of COPD rats. Moreover, it can upregulate mitochondrial respiratory chain enzyme activity. ECC-BYF III also decreased the MDA content and increased T-SOD, GSH-Px, and T-AOC expression to facilitate oxidative homeostasis. Finally, our results indicated that the Pkm2/Nrf2 pathway is regulated by ECC-BYF III in A549 cells and lung tissue. Conclusion These results indicate that ECC-BYF III exerts a strong effective therapeutic effect against cigarette smoke combined with bacteria-induced COPD in rats by activating the Pkm2/Nrf2 signaling pathway and restoring mitochondrial oxidative stress. Although more in vivo animal model research is needed to confirm these findings, this study contributes new data to support the conventional usage of ECC-BYF III.
Collapse
Affiliation(s)
- Yang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Lanxi Zhang
- School of Basic Medicine (Zhongjing School), Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jie Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, 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
| | - Ruilong Lu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xuejie Shao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Kexin Xu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, 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
- First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Yange Tian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, 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
| |
Collapse
|
2
|
He Q, Li P, Han L, Yang C, Jiang M, Wang Y, Han X, Cao Y, Liu X, Wu W. Revisiting airway epithelial dysfunction and mechanisms in chronic obstructive pulmonary disease: the role of mitochondrial damage. Am J Physiol Lung Cell Mol Physiol 2024; 326:L754-L769. [PMID: 38625125 DOI: 10.1152/ajplung.00362.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Chronic exposure to environmental hazards causes airway epithelial dysfunction, primarily impaired physical barriers, immune dysfunction, and repair or regeneration. Impairment of airway epithelial function subsequently leads to exaggerated airway inflammation and remodeling, the main features of chronic obstructive pulmonary disease (COPD). Mitochondrial damage has been identified as one of the mechanisms of airway abnormalities in COPD, which is closely related to airway inflammation and airflow limitation. In this review, we evaluate updated evidence for airway epithelial mitochondrial damage in COPD and focus on the role of mitochondrial damage in airway epithelial dysfunction. In addition, the possible mechanism of airway epithelial dysfunction mediated by mitochondrial damage is discussed in detail, and recent strategies related to airway epithelial-targeted mitochondrial therapy are summarized. Results have shown that dysregulation of mitochondrial quality and oxidative stress may lead to airway epithelial dysfunction in COPD. This may result from mitochondrial damage as a central organelle mediating abnormalities in cellular metabolism. Mitochondrial damage mediates procellular senescence effects due to mitochondrial reactive oxygen species, which effectively exacerbate different types of programmed cell death, participate in lipid metabolism abnormalities, and ultimately promote airway epithelial dysfunction and trigger COPD airway abnormalities. These can be prevented by targeting mitochondrial damage factors and mitochondrial transfer. Thus, because mitochondrial damage is involved in COPD progression as a central factor of homeostatic imbalance in airway epithelial cells, it may be a novel target for therapeutic intervention to restore airway epithelial integrity and function in COPD.
Collapse
Affiliation(s)
- Qinglan He
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihua Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Chen Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Meiling Jiang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyu Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yuanyuan Cao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weibing Wu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
3
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Kong J, Xiang Q, Ge W, Wang Y, Xu F, Shi G. Network pharmacology mechanisms and experimental verification of licorice in the treatment of ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117691. [PMID: 38176667 DOI: 10.1016/j.jep.2023.117691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Licorice is widely used in the treatment of ulcerative colitis (UC) and has good antioxidant and anti-inflammatory effects, but its specific active ingredients and mechanisms of action are still unknown. THE PURPOSE OF THE STUDY To elucidate the specific molecular mechanisms of licorice in the treatment of UC and to experimentally verify its activity. METHODS Through network pharmacology, the active ingredients of licorice and the molecular targets of UC were identified. A traditional Chinese medicine (TCM)-components-target-disease network diagram was established, and the binding energies of the active ingredient and targets of licorice were verified by molecular docking. A BALB/c mice model of UC was established by treatment with 3% dextran sulfate sodium (DSS). The effect of licorice on colon tissue injury was histologically assessed. The expression of IL-6 and IL-17 in colon tissue was detected by immunohistochemistry (IHC). Transmission electron microscopy (TEM) was used to observe morphological changes in mitochondria in the colon. Caco2 cells were treated with lipopolysaccharide (LPS) for 24 h to establish the cell inflammatory damage model, and cells were exposed to different concentrations of drug-containing serum of Licorice (DCSL) for 24 h. In cells treated with the drug, the contents of oxidation markers were measured and ELISA was used to determine the levels of inflammatory factors in the cells. TEM was used to observe morphological changes in mitochondria. ZO-1 and occludin were detected by Western blotting. DCSL effects on autophagy were evaluated by treating cells with DCSL and autophagy inhibitor for 24 h after LPS injection. Small interfering ribonucleic acid (si-RNA) was used to silence Nrf2 gene expression in Caco2 cells to observe the effects of DCSL on autophagy through the Nrf2/PINK1 pathway. Nrf2, PINK1, HO-1, Parkin, P62, and LC3 were detected by Western blotting. RESULTS Ninety-one active ingredients and 339 action targets and 792 UC disease targets were identified, 99 of which were overlapping targets. Molecular docking was used to analyze the binding energies of liquiritin, liquiritigenin, glycyrrhizic acid, and glycyrrhetinic acid to the targets, with glycyrrhetinic acid having the strongest binding energy. In the UC mouse model, licorice improved colon histopathological changes, reduced levels of IL-6 and IL-17 and repaired mitochondrial damage. In the LPS-induced inflammation model of Caco2 cells, DCSL decreased MDA, IL-1β, Il-6, and TNF-α levels and increased those of Superoxide Dismutase (SOD), glutathione peroxidase (GSH-PX), and IL-10, and improved the morphological changes of mitochondria. Increased expression of Nrf2, PINK1, Parkin, HO-1, ZO-1, occludin, P62, and LC3 promoted autophagy and reduced inflammation levels. CONCLUSION Licorice improves UC, which may be related to the activation of the Nrf2/PINK1 signaling pathway that regulates autophagy.
Collapse
Affiliation(s)
- Jinrong Kong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China
| | - Qingzhen Xiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China
| | - Wanyue Ge
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China
| | - Yunlai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China
| | - Fan Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China.
| | - Gaoxiang Shi
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei City, Anhui Province, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei City, Anhui Province, China.
| |
Collapse
|
5
|
Qin Y, Zhai J, Yang J, Li H, Tian Y, Liu X, Zhao P, Li J. Effective-component compatibility of Bufei Yishen formula alleviates chronic obstructive pulmonary disease inflammation by regulating GSK3β-mediated NLRP3 inflammasome activation. Biomed Pharmacother 2023; 168:115614. [PMID: 37862971 DOI: 10.1016/j.biopha.2023.115614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023] Open
Abstract
Glycogen synthase kinase 3β (GSK3β) has been associated with sensing many different stimuli to trigger the NLRP3 inflammasome, which plays a crucial role in promoting the inflammatory response in diseases, including chronic obstructive pulmonary disease (COPD). Bufei Yishen formula (BYF), a traditional Chinese herbal medicine, has beneficial effects on COPD. Effective-component compatibility of BYF (ECC-BYF), optimized from BYF, is equally effective as BYF in inhibiting COPD inflammation. However, the exact mechanism by which ECC-BYF regulates the activation of NLRP3 inflammasome to inhibit COPD inflammation remains unclear. Hence, we investigated the mechanisms underlying the alleviation of COPD inflammation by ECC-BYF through the inhibition of GSK3β-mediated NLRP3 inflammasome activation by experimental rat model of COPD and lipopolysaccharide/adenosine triphosphate (LPS/ATP) induced macrophages. The data showed that ECC-BYF significantly improved the lung function, attenuated histopathological damage, and alleviated inflammatory cell infiltration and alveolar destruction. Further, it significantly inhibited inflammatory cytokine production and downregulated the phosphorylation of GSK3β by inhibiting the activation of NLRP3 inflammasome in the rat model of COPD. Moreover, ECC-BYF suppressed the activation of the NLRP3 inflammasome by increasing the phosphorylation at serine 9 and decreasing the phosphorylation at tyrosine 216 of GSK3β, followed by the inhibition of IL-1β secretion in macrophages. Together, ECC-BYF effectively ameliorates COPD by suppressing inflammation, which is dependent on the regulation of GSK3β-mediated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jiena Zhai
- 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jingfan Yang
- 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Haibo 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, 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 and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - 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 and 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, Henan Province, China.
| |
Collapse
|
6
|
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
|
7
|
Li K, Mei X, Xu K, Jia L, Zhao P, Tian Y, Li J. Comparative study of cigarette smoke, Klebsiella pneumoniae, and their combination on airway epithelial barrier function in mice. ENVIRONMENTAL TOXICOLOGY 2023; 38:1133-1142. [PMID: 36757011 DOI: 10.1002/tox.23753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/09/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The airway epithelium acts as a physical barrier to protect pulmonary airways against pathogenic microorganisms and toxic substances, such as cigarette smoke (CS), bacteria, and viruses. The disruption of the structural integrity and dysfunction of the airway epithelium is related to the occurrence and progression of chronic obstructive pulmonary disease. PURPOSE The aim of this study is to compare the effects of CS, Klebsiella pneumoniae (KP), and their combination on airway epithelial barrier function. METHODS The mice were exposed to CS, KP, and their combination from 1 to 8 weeks. After the cessation of CS and KP at Week 8, we observed the recovery of epithelial barrier function in mice for an additional 16 weeks. To compare the epithelial barrier function among different groups over time, the mice were sacrificed at Weeks 4, 8, 16, and 24 and then the lungs were harvested to detect the pulmonary pathology, inflammatory cytokines, and tight junction proteins. To determine the underlying mechanisms, the BEAS-2B cells were treated with an epidermal growth factor receptor (EGFR) inhibitor (AG1478). RESULTS The results of this study suggested that the decreased lung function, increased bronchial wall thickness (BWT), elevated inflammatory factors, and reduced tight junction protein levels were observed at Week 8 in CS-induced mice and these changes persisted until Week 16. In the KP group, increased BWT and elevated inflammatory factors were observed only at Week 8, whereas in the CS + KP group, decreased lung function, lung tissue injury, inflammatory cell infiltration, and epithelial barrier impairment were observed at Week 4 and persisted until Week 24. To further determine the mechanisms of CS, bacteria, and their combination on epithelial barrier injury, we investigated the changes of EGFR and its downstream protein in the lung tissues of mice and BEAS-2B cells. Our research indicated that CS, KP, or their combination could activate EGFR, which can phosphorylate and activate ERK1/2, and this effect was more pronounced in the CS + KP group. Furthermore, the EGFR inhibitor AG1478 suppressed the phosphorylation of ERK1/2 and subsequently upregulated the expression of ZO-1 and occludin. In general, these results indicated that the combination of CS and KP caused more severe and enduring damage to epithelial barrier function than CS or KP alone, which might be associated with EGFR/ERK1/2 signaling. CONCLUSION Epithelial barrier injury occurred earlier, was more severe, and had a longer duration when induced by the combination of CS and KP compared with the exposure to CS or KP alone, which might be associated with EGFR/ERK signaling.
Collapse
Affiliation(s)
- Kangchen Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofeng Mei
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Kexin Xu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lidan Jia
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiansheng Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
8
|
Effects of Ipratropium Bromide Combined with Traditional Chinese Medicine Intervention on the Pulmonary Function and Psychological Status of Patients with Chronic Obstructive Pulmonary Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:6483785. [PMID: 36798728 PMCID: PMC9928514 DOI: 10.1155/2023/6483785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/28/2022] [Accepted: 10/09/2022] [Indexed: 02/10/2023]
Abstract
Recently, most scholars have advocated multidisciplinary comprehensive intervention measures for chronic obstructive pulmonary disease (COPD) to improve lung function, relieve symptoms of dyspnea, and improve quality of life. Traditional Chinese medicine (TCM) has rich experience in the treatment of various respiratory system diseases and the rehabilitation of their syndrome differentiation. In this study, total 68 patients with COPD from November 2019 to November 2021 in the hospitals were divided into the control group, ipratropium bromide (IB)-treated group, and IB + TCM-treated group for clinical efficacy observation and to explore the effect of IB combined with TCM on the pulmonary function and psychological status of COPD patients. Patients in the control group were subjected to routine oxygen inhalation, cough and expectorant, and antiviral treatments, while the patients in the IB-treated group were treated with IB and those received in the control group. Patients in the IB + TCM-treated group were treated with IB and TCM intervention. All patients were treated for a month. The results showed that after different interventions, the levels of FEV1, FEV1% pred, FVC, and PEF (P < 0.05) were significantly increased in all the groups, while levels of TNF-α, IL-6, IL-8, and CRP in serum as well as Hamilton Anxiety Scale and Hamilton Depression scores were significantly decreased. Compared with the control group and IB-treated group, the IB + TCM-treated group presented the greatest changes on all abovementioned indicators and the lowest total incidence of adverse reactions, indicating the biggest improvement of IB + TCM on the symptoms of COPD patients. Therefore, the combination of IB and TCM intervention effectively improved the pulmonary function and psychological status of COPD patients and could be used as an important adjunct for COPD treatment.
Collapse
|