Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Pharm Biomed Anal ; 241: 115981, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38237543

ABSTRACT

Shenqi-Tiaoshen formula (SQTSF) is a traditional Chinese medicine (TCM) prescription that has been employed in the treatment of chronic obstructive pulmonary disease (COPD). Clinical practice has demonstrated that SQTSF is an effective prescription for stable COPD. However, owing to the complexity of TCM prescription, there is a lack of in-depth understanding of the chemical components of SQTSF and its in vivo metabolism studies. In this study, a comprehensive analytical strategy based on ultra-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF/MS) was established to identify the chemical components, the absorbed components, and the metabolites of SQTSF given by gavage in rats, and analyze their dynamic changes. As a result, 86 chemical components of SQTSF were characterized, which were mainly categorized into flavonoids, saponins, organic acids, terpenoids, etc. Among them, 13 compounds were confirmed unambiguously by reference standards. Furthermore, 20 prototype components and 46 metabolites were detected in rat plasma at different time points. It was found that one prototype component and thirteen metabolites could be detected during the entire 24 h, indicating that these compounds were slowly eliminated and thus accumulated in vivo over a prolonged duration. Interestingly, the phenomenon that three prototype components and fourteen metabolites reappeared after a period of disappearance from the plasma was found. It was also observed that different prototype components may generate the same metabolite. The metabolic processes of SQTSF in rats mainly included oxidation, reduction, hydration, demethylation, deglycosylation, methylation, acetylation, glucuronidation, glutathionylation, and associated combination reactions. Overall, the present study identified the chemical components of SQTSF and their dynamic metabolic profile in rat plasma, which provided a systematic and applicable strategy for screening and characterization of the prototype components and metabolites of TCM compound preparations.


Subject(s)
Drugs, Chinese Herbal , Pulmonary Disease, Chronic Obstructive , Rats , Animals , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods , Chromatography, High Pressure Liquid/methods , Metabolome , Drugs, Chinese Herbal/chemistry
2.
Altern Ther Health Med ; 30(1): 270-277, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37793329

ABSTRACT

Objective: Bronchial asthma is a prevalent respiratory disorder characterized by airway inflammation. This study aimed to investigate the protective effect of Pingchuanning decoction (PCN) on airway inflammation in bronchial asthma, focusing on the role of autophagy and its underlying molecular mechanism. Methods: Using an in vitro lipopolysaccharide (LPS)-induced inflammatory damage model of human airway epithelial cells (16HBE), we assessed the effect of PCN. Various experiments were performed to evaluate the expression of autophagy-related genes, autophagosome and vesicle counts, and reactive oxygen species (ROS) levels. Results: First, PCN reduced LPS-induced cellular inflammation. Second, PCN decreased the number of autophagosomes and autophagic vesicles. And third, PCN significantly reduced reactive oxygen species (ROS) levels. Most importantly, PCN also down-regulated LPS-induced expression of HMGB1, Beclin-1, and autophagy-related gene 5 (ATG5) while enhancing the expression of B-cell lymphoma 2 (Bcl-2), which further reduced the LC3II/I ratio. Conclusion: PCN reduces the 16HBE inflammatory response by inhibiting the overexpression of ROS/HMGB1/Beclin-1 mediated cell autophagy. Therefore, it may serve as a potential drug for treating bronchial asthma.


Subject(s)
Asthma , HMGB1 Protein , Humans , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Reactive Oxygen Species/therapeutic use , Beclin-1/genetics , HMGB1 Protein/genetics , HMGB1 Protein/pharmacology , HMGB1 Protein/therapeutic use , Lipopolysaccharides , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Autophagy/genetics , Inflammation/drug therapy
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(11): 961-966, 2022 Nov.
Article in Chinese | MEDLINE | ID: mdl-36328424

ABSTRACT

Objective To investigate the mechanism of the Qibaipingfei Capsule regulating chronic obstructive pulmonary (COPD) related immune cells by analyzing the single-cell transcriptome sequencing (scRNA-seq) data of COPD lung tissue and the pharmacology of Qibaipingfei Capsule. Methods The scRNA-seq data of COPD lung tissue downloaded from the gene expression omnibus (GEO) was used to obtain the COPD related immune cells and the differentially expressed RNA, and the primary active molecular and target genes of Qibaipingfei Capsule were retrieved from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). The "active molecules-immune cells-target genes" network was constructed by mapping the target genes of Qibaipingfei Capsules to the differentially expressed RNA of COPD related immune cells, and the Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and the protein-protein interaction (PPI) were administrated to analyze the molecular mechanisms of target genes. Results Twelve active molecules including quercetin, kaempferol, and formononetin of Qibaipingfei Capsule targeted multiple COPD related immune cells like macrophages, alveolar macrophages, and T cells, and genes like PPARG, JUN, HMOX1, and HIF1A which were primarily collected in pathways such as interleukin 17 signaling pathway, nuclear factor-κB (NF-κB) signaling pathway, and mitogen-activated protein kinase (MAPK) signaling pathway. Conclusions The Qibaipingfei Capsule may target multiple immune cells and intervene in inflammation and immune-related pathways to regulate the inflammation and immune response of COPD.


Subject(s)
Drugs, Chinese Herbal , Pulmonary Disease, Chronic Obstructive , Humans , Computational Biology , Pulmonary Disease, Chronic Obstructive/genetics , Gene Ontology , Inflammation , RNA , Drugs, Chinese Herbal/pharmacology
4.
Article in English | MEDLINE | ID: mdl-34093717

ABSTRACT

OBJECTIVE: Qiyusanlong (QYSL) formula has been used in the clinic for more than 20 years and has been proved to have pronounced efficacy in the treatment of non-small-cell lung cancer (NSCLC). This work aims to evaluate the molecular mechanism of QYSL formula action on NSCLC, specifically in relation to autophagy induction. METHODS: In vitro, CCK-8 was used to detect the effect of QYSL serum on cell viability in A549 cells. In vivo, A549 cells were implanted subcutaneously in nude mice to establish a xenograft model. TUNEL staining was used to measure cell apoptosis and TEM to observe the autophagy-related morphological changes in vitro and in vivo. Western blotting, RT-qPCR, and immunofluorescence were used to measure autophagy-related proteins. In addition, rapamycin (an inhibitor of mTOR and inducer of autophagy) and MHY1485 (an activator of mTOR and inhibitor of autophagy) were used to determine whether QYSL-induced autophagy was regulated by the mTOR pathway. RESULTS: QYSL serum inhibited the cell viability of A549 cells in a concentration-dependent manner. In vivo, the QYSL formula inhibited xenograft growth. The QYSL formula promoted apoptosis in A549 cells and induced autophagosome formation in vitro and in vivo. In addition, the QYSL formula downregulated the expression of mTOR and p62, while it upregulated the expression of ATG-7 and Beclin-1 and increased the LC3-II/LC3-I ratio. QYSL serum inhibited p-mTOR in a similar manner to rapamycin while reducing the activating effects of MHY1485 on p-mTOR. CONCLUSION: The QYSL formula has anti-lung cancer effects and promotes autophagy through the mTOR signaling pathway.

5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(2): 115-120, 2019 Feb.
Article in Chinese | MEDLINE | ID: mdl-30975275

ABSTRACT

Objective To explore the effect of Qibai Pingfei capsule (QPC) on the inflammation and oxidative stress in a chronic obstructive pulmonary disease (COPD) rat models with the syndromes of qi deficiency and phlegm and blood stasis by regulating the SIRT1/FoxO3a pathway. Methods A total of 80 male SD rats were randomly divided into 4 groups with 20 animals in each group: a non-diseased group, a non-treated diseased group, a diseased group treated with QPC, and a diseased group treated with placebo. The COPD rat models with the syndromes of qi deficiency and phlegm and blood stasis were then developed with established protocols. After the corresponding treatments, the serum levels of superoxide dismutase (SOD), malondialdehyde (MDA), interleukine 1ß (IL-1ß), and IL-2 were determined by ELISA; the protein levels of SIRT1 and FoxO3a were quantified by Western blot analysis; the mRNA levels of the SIRT1 and FoxO3a genes were also measured by real-time quantitative PCR. Results First of all, compared with the non-diseased group, the serum levels of MDA, IL-1ß, and IL-2 were elevated in the diseased group, while the level of SOD was reduced. Both mRNA and protein levels of SIRT1 decreased, while the levels of FoxO3a increased in the lung tissues of the diseased group. Compared with the diseased group treated with placebo, the diseased group treated with QPC had reduced serum levels of MDA, IL-1ß and IL-2, elevated SOD, increased mRNA and protein levels of SIRT1 and decreased levels of FoxO3a, thereby restoring their levels partially under the disease state. Conclusion QPC can alleviate inflammation and oxidative stress of COPD rats with syndrome of qi deficiency and phlegm and blood stasis effectively, potentially through regulating the expression level of the SIRT1/FoxO3a pathway.


Subject(s)
Drugs, Chinese Herbal , Gene Expression Regulation , Inflammation , Oxidative Stress , Pulmonary Disease, Chronic Obstructive , Qi , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Forkhead Box Protein O3/genetics , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Male , Oxidative Stress/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Rats , Rats, Sprague-Dawley , Sirtuin 1/genetics , Syndrome
6.
J Cell Biochem ; 120(3): 3833-3844, 2019 03.
Article in English | MEDLINE | ID: mdl-30260006

ABSTRACT

BACKGROUND: Pingchuanning decoction is a well-known traditional Chinese medicine for the treatment of airway inflammatory diseases, including asthma. However, the potential mechanism by which Pingchuanning decoction contributes to the amelioration of airway inflammation remains unknown. METHODS: A rat model of asthma was well established by inducing ovalbumin. Lipopolysaccharide-stimulated rat tracheal epithelial (RTE) cells were used as cellular model. Lung histopathology and goblet cell hyperplasia were assessed by hematoxylin-eosin (HE) and periodic acid Schiff staining, respectively. Total inflammatory cells count and RTE cell apoptosis were analyzed by flow cytometry. The autophagic activities were evaluated by immunohistochemical and immunofluorescence analysis and Western blot analysis of autophagy-related proteins. We also detected the effects of Pingchuanning decoction on phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) and high-mobility group box 1 (HMGB1)-mediated toll-like receptor 4 (TLR4)/NF-κB pathways-related proteins and inflammatory cytokines using the Western blot analysis and enzyme-linked immunosorbent assay. RESULTS: Pingchuanning decoction effectively attenuated pulmonary pathology and autophagy. Treatment with Pingchuanning decoction activated PI3K/Akt/mTOR pathway and inhibited HMGB1/TLR4/NF-κB pathway, which could be overturned by LY294002, a PI3K antagonist, or rapamycin (Rapa), an autophagy inducer. CONCLUSION: Pingchuanning decoction exerted a therapeutic effect on asthma by inhibiting autophagy via PI3K/Akt /mTOR signaling pathway.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Drugs, Chinese Herbal/pharmacology , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/metabolism , Autophagy/drug effects , Autophagy/genetics , Dexamethasone/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Ovalbumin/administration & dosage , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL