Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Main subject
Language
Affiliation country
Publication year range
1.
Phytomedicine ; 123: 155211, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38061286

ABSTRACT

BACKGROUND/PURPOSE: Exposure to particles with an aerodynamic diameter of ≤2.5 µm (PM2.5) increased various lung diseases, which lack effective treatment. Massive evidence links PM2.5 to the development of allergic lung diseases like asthma. Modified Guo-Min Decoction (MGMD) is a traditional Chinese formula for allergic diseases. However, whether MGMD could improve PM2.5-induced lung injury and the underlying mechanism remain unclear and we aimed to explore. STUDY DESIGN/METHODS: Male Wistar rats (200-220 g) were intratracheally instilled of PM2.5 suspension daily for 4 weeks to establish PM2.5-induced lung injury model. MGMD (2.1 g/kg) treatment by gavage was started 1 week before, at the same time or 1 week after the instillation of PM2.5 suspension, namely the pre-, sync- or post-administration groups. HE and Masson staining were used to observe morphological changes. Immunohistochemistry staining was used to detect macrophage and neutrophil infiltration. The levels of inflammatory cytokines in the bronchoalveolar lavage fluid were detected by ELISA. The main components of MGMD were detected by UHPLC-LTQ-Orbitrap MSn. Network pharmacology was used to identify the key targets mediating the effect of MGMD in treating PM2.5-induced lung injury. Changes in the expression of target proteins were examined by western blot. In-vitro experiments were carried out in Beas2b cells to evaluate the protective effect and mechanism of MGMD against PM2.5 induced injury. RESULTS: Exposure to PM2.5 suspension resulted in disarrangement of tracheal epithelium, neutrophil and M1 macrophage infiltration and collagen deposition, and significantly increased IgE, IL-1ß and IL-17 secretion and NLRP3 expression, which were inhibited by MDMD treatment and pre-MGMD treatment showed the best effect. By UHPLC-LTQ-Orbitrap MSn, 46 main compounds were identified in MGMD. Using network pharmacology approach, we found MGMD attenuate PM2.5-induced lung damage by targeting 216 genes, and PPI network, GO and KEGG analysis all indicated that PI3K-AKT and MAPK pathways were important. Western blot showed that PM2.5 suspension exposure increased PI3K, AKT, ERK and JNK phosphorylation, which were reversed by MGMD intervention significantly. In vitro, the viability of Beas2b cells was significantly decreased after PM2.5 suspension exposure, and was obviously upregulated after MGMD-containing serum or LY294002 treatment. CONCLUSION: The present study demonstrated that MGMD could improve PM2.5-induced lung injury through reducing inflammation and pulmonary fibrosis, which was probably mediated by inhibition of the PI3K-AKT and MAPK signaling pathways, and NLRP3 inflammasome. The results of this study support and provide scientific evidence for the clinical application of MGMD on PM2.5-induced lung injury. Pre-treatment, sync-treatment, and post-treatment is the highlight of this study.


Subject(s)
Lung Injury , Rats , Animals , Male , Lung Injury/chemically induced , Lung Injury/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphatidylinositol 3-Kinases/metabolism , Rats, Wistar , Signal Transduction , Particulate Matter/toxicity
2.
Article in English | MEDLINE | ID: mdl-36523421

ABSTRACT

Objective: To explore the effects and mechanisms of Bufei Huoxue Capsule (BHC) on chronic obstructive pulmonary disease (COPD) based on network pharmacology. Methods: The effective components and related targets of BHC were collected by searching TCMSP, HERB, and ETCM databases, after which the related targets of COPD were obtained on GeneCards and OMIM databases. The common targets were imported into the STRING database and Cytoscape database to construct a target interaction network and screen core targets. Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the Metascape platform. According to the prediction results of network pharmacology, the action mechanism was further examined in an animal model of COPD. The pathological changes of lung tissue were observed by HE staining; goblet cells and mucus secretion in lung tissue were observed by AB-PAS staining, airway collagen deposition was observed by Masson staining, and the expression of NE, TGF-ß1, P-EGFR/EGFR, P-ERK1/2/ERK1/2, P-JNK/JNK, and P-P38/P38MAPK protein was detected by Western blot analysis. Results: A total of 379 targets related to BHC and 7391 targets related to COPD were obtained, including 313 potential targets of BHC in treating chronic obstructive pulmonary disease, with JUN, AKT1, TNF, IL6, EGFR, MAPK1, and MAPK14 as the core targets. Through enrichment analysis, BHC may interfere with COPD by regulating the MAPK signal pathway, HIF-1 signal pathway, NF-κB signal pathway, cAMP signal pathway, cGMP-PKG signal pathway, and so on. Animal experiments showed that the BHC could reduce airway inflammatory cell infiltration, inhibit airway epithelial goblet cell proliferation, reduce mucus secretion, and improve small airway collagen fiber deposition in COPD model rats. Besides, BHC could downregulate the protein expression of NE, TGF-ß1, P-EGFR, P-ERK1/2, and P-P38MAPK. Conclusion: BHC can reduce airway inflammation, inhibit mucus hypersecretion, and improve airway remodeling by regulating the MAPK signal transduction pathway.

3.
Article in English | MEDLINE | ID: mdl-30800170

ABSTRACT

Airway mucus hypersecretion is the main pathogenic factor in acute exacerbation of chronic obstructive pulmonary disease (AECOPD) and the control of mucus secretion is closely associated with survival. Louqin Zhisou decoction (LQZS) has been found to improve lung function and reduce sputum in AECOPD patients, but the mechanism remains unclear. This study aimed to explore the mechanism of LQZS against mucus hypersecretion in lung tissues of rat AECOPD model. Wistar rats were used to establish AECOPD model by intratracheal instillation of LPS in combination with the continuous cigarette smoking. Rats were administrated LQZS/clarithromycin (CAM)/distilled water via gavage every day and all rats were sacrificed after 30 days. BALF and lung tissues were obtained. Lung morphology, cytokines levels, MUC5AC mRNA transcription and protein expression, phosphorylation of the EGFR-PI3K-AKT signaling pathway, and molecules involved in Th17/Treg balance were evaluated. The results demonstrated that LQZS protected rats from decline in pulmonary function and ameliorated lung injury. LQZS treatment decreased the number of goblet cells in airway and suppressed MUC5AC mRNA and protein expression of lung tissues. Furthermore, LQZS attenuated the level of phospho-EGFR, phospho-PI3K and phospho-AKT in AECOPD rats. In addition, LQZS could inhibit the production of proinflammatory cytokines in BALF, including IL-6 and IL-17A and downregulate the secretion of NE and MCP-1, indicating that LQZS could limit inflammatory responses in AECOPD. Moreover, LQZS reversed RORγt and Foxp3 expression, the key transcription factors of Th17 and Treg, respectively. In conclusion, this research demonstrated the inhibitory effects of LQZS against mucus hypersecretion in AECOPD via suppressing EGFR-PI3K-AKT signaling pathway and restoring Th17/Treg balance.

SELECTION OF CITATIONS
SEARCH DETAIL