Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add filters








Language
Year range
1.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 246-255, 2022.
Article in Chinese | WPRIM | ID: wpr-940642

ABSTRACT

Autophagy and tumor immune escape are important biological mechanisms in the process of tumor cell proliferation and metastasis, involving multiple signaling pathways. The interaction of autophagy and tumor immune escape seriously affects the treatment and prognosis of tumor diseases. However, the correlation between autophagy and tumor immune escape is still not fully elucidated. Recent studies have shown that autophagy can affect the activity of immune cells by regulating the presentation of antigens in tumor cells, the release of cytokines, and the degradation of immune checkpoint proteins, thereby positively or negatively regulating tumor cell immune escape. The activation of autophagy in tumor cells can inhibit the activation of the innate immune sensing pathway of stimulator of interferon genes (STING)-type Ⅰ interferon (IFN-Ⅰ) to inhibit its immunogenicity and cytotoxic T lymphocytes (CTLs), which promotes tumor immune escape. While autophagy suppression can reduce the infiltration of M2 macrophages, promote the binding of natural killer group 2, member D (NKG2D) to its ligand, and inhibit the recognition of immune checkpoint proteins, thereby exerting an immune-killing effect and inhibiting tumor immune escape. Traditional Chinese medicine (TCM) has unique advantages in anti-tumor research, especially in the unilateral regulation of autophagy or improvement of tumor immunity, but the research based on the regulation of autophagy and tumor immunity by TCM is insufficient. A few studies have shown that Chinese medicine monomers and compounds can exert an anti-tumor effect by regulating cell autophagy and interfering with tumor immune escape, but there is still a lack of systematic elaboration. The present study reviewed correlation between autophagy and tumor immune escape and regulation of autophagy by Chinese medicine to interfere with tumor immune escape to provide new ideas for research on mechanism of TCM against tumor diseases and development of innovative TCM drugs against tumors.

2.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 179-189, 2022.
Article in Chinese | WPRIM | ID: wpr-940435

ABSTRACT

ObjectiveTo predict the therapeutic target genes and related signaling pathways of Qinghuangsan (QHP) in the treatment of acute myeloid leukemia (AML) by network pharmacology,molecular docking,and further clarify its mechanisms through in vitro cell experiment. MethodThe active components and targets of QHP were retrieved from traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP),traditional Chinese medicine integrated database (TCMID),TargetNet and SwissTargetPrediction databases,and AML-related target genes were obtained by GeneCards and online mendelian inheritance in man (OMIM) databases. After screening the common targets of QHP and AML,the protein-protein interaction (PPI) network of the common targets was constructed with STRING,followed by gene ontology (GO) term and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis based on RStudio software and clusterProfiler,Bioconductor packages. At the same time,Cytoscape software is used to construct the network of "disease-component-target" and "compound-target-pathway". Select the active ingredients of QHP for molecular docking with the top 8 targets in the "compound-target-pathway" network. In vitro cell experiment and Western blot were used to further verify the anti-AML effect of QHP. ResultThe prediction results show that there are 11 main active components of QHP,and 22 common targets of QHP and AML are collected. KEGG pathway analysis results show that phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) and mitogen-activated protein kinase (MAPK) signaling pathways may play a key role in the treatment of AML disease by QHP. "Compound-target-pathway" network analysis showed that the top 8 targets include Akt1,phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA),mitogen-activated protein kinase kinase 1 (MAP2K1),TP53,serine/threonine kinase (RAF1),B cell lymphoma(Bcl)-2,cysteine aspartic acid specific protease(Caspase)-9 and JUN. Molecular docking results showed that 3-indolyl-β-D-glucopyranoside was optimally docked with MAP2K1,isovitexin docked with PIK3CA,and indirubin docked with Bcl-2. Cell experiments show that 3-indolyl-β-D-glucopyranoside,isovitexin and indirubin can effectively inhibit the proliferation of AML cells,regulate the MAPK/PI3K signaling pathway,and inhibit the expression of Bcl-2 protein. ConclusionQHP can treat AML through "multi-component,multi-target,multi-pathway" synergistic treatment,and its mechanism of pharmacology may be related to the regulation of MAPK signaling pathway and PI3K/Akt signaling pathway.

3.
Journal of Experimental Hematology ; (6): 1003-1010, 2017.
Article in Chinese | WPRIM | ID: wpr-271878

ABSTRACT

<p><b>OBJECTIVE</b>To explore the effect of arsenic trioxide combined with itraconazole on proliferation and apoptosis of KG1a cells and its potential mechanism.</p><p><b>METHODS</b>The cell morphology was observed with Wrighe-Giemsa staining; cell survival rate was examined by CCK-8; and colony formation capacity was measured by methylcellulose colony formation test; the flow cytometry was used to analyse the cell apoptosis rate and cell cycle; the protein expressions of BCL-2,caspase-3,BAX,SMO,Gli1 and Gli2 were detected by Western-blot.</p><p><b>RESULTS</b>The arsenic trioxide and itraconazole alone both could inhibit the KG1a cell proliferation in dose-and time-dependent manner. In comparison between single and combined drug-treatment group, both the cell survival rate and the colony number of the single drug-treatment group were significantly lower(P<0.05), and the apoptosis rate was higher in the combined drug-treatment group. In the combined-treatment group, the protein expression of Caspase-3 and BAX was upregulated, while the protein expression of BCL-2,SMO,Gli1 and Gli2 was downregulated.</p><p><b>CONCLUSION</b>Arsenic trioxide combined with itraconazole can inhibit the KG1a cell proliferation and induce apoptosis, which may be related with the inhibition of Hh signaling pathway and upregulation of both Caspase-3 and BAX protein expression, and provided experimental data of arsenic trioxide combined with itraconazole for the treatment of refractory AML.</p>

4.
Journal of Experimental Hematology ; (6): 1267-1272, 2014.
Article in Chinese | WPRIM | ID: wpr-340516

ABSTRACT

This study was aimed to explore the effect of arsenic trioxide combined with curcumin on proliferation and apoptosis of KG1a cells and its potential mechanism. The cell survival rate was mesured by MTT; colony formation capacity was examined by methylcellulose colony formation test; flow cytometry was used to analyse the cell surface molecules, cell apoptosis rate and cell cycle; the cell morphology was observed with Wright-Giemsa staining and the protein expression of BCL-2, BAX, PARP was detected by Western blot. The results showed that the phenotype of KG1a cells was CD34(+)CD38(-), while the phenotype of HL-60 cell was CD34(+)CD38(+). The former possessed a stronger colony ability than the latter. Effect of curcumin and arsenic trioxide alone on cell proliferation and inhibition was in dose-dependent manner. Compared with single drug-treatment group, the cell survival rate and colony number were lower, and the apoptosis rate was higher in combined drug-treatment group. Protein expression of BCL-2 and PARP was upregulated, while the protein expression of PARP was downregulated in the combined treatment group. It is concluded that compared with HL-60 cells, KG1a cells are the earlier leukemia stem/progenitor cells. Arsenic trioxide combined with curcumin can effectively inhibit the KG1a cell proliferation and induce apoptosis, which may be associated with the downregulation of BCL-2 and PARP protein expression and the upregulation of BAX protein expression.


Subject(s)
Humans , Apoptosis , Arsenicals , Pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Curcumin , Pharmacology , Oxides , Pharmacology , bcl-2-Associated X Protein
5.
Journal of Experimental Hematology ; (6): 1577-1583, 2014.
Article in Chinese | WPRIM | ID: wpr-340455

ABSTRACT

This study was aimed to investigate the effect of Honokiol (HNK) on proliferation and apoptosis of acute myeloid leukemia HL-60 cells and its potential mechanism. Inhibitory effect of HNK on the HL-60 cell proliferation was detected by MTT assay. Flow cytometry was used to detect the change of cell cycle and AnnexinV/PI staining was used to detect apoptosis. Western blot was applied to analyze the cell cycle protein (cyclins), cyclin-dependent kinase (CDK), P53, P21, P27, BCL-2, BCL-XL, Bax, caspase-3/9 and proteins for MAPK signal pathway. The results showed that HNK could inhibit the proliferation of HL-60 cells in time- and dose dependent ways. HNK arrested HL-60 cells in G0/G1 phase, and S phase cells decreased significantly (P < 0.05). The expression of cyclin D1, cyclin A, cyclin E and CDK2/4/6 were significantly down-regulated (P < 0.05), the expression of P53 and P21 was significantly upregulated after treating for 24 h with HNK (P < 0.05). After 24 h treatment with HNK, HL-60 cell apoptosis increased significantly with the upregulation of activated caspase-3, -9, BAX expression and the downregulation of BCL-2, BCL-XL expression. The MAPK subfamily, P38 and JNK were not significantly changed, but the expression of MEK1/2-ERK1/2 was significantly downregulated (P < 0.05). It is concluded that HNK arrestes the cells at G0/G1 phase and induces HL-60 cell apoptosis through the intervention of MEK1/2-ERK1/2 signaling pathway.


Subject(s)
Humans , Apoptosis , Biphenyl Compounds , Pharmacology , Caspase 3 , Cell Cycle , Cell Proliferation , Cyclin D1 , Cyclin E , Cyclin-Dependent Kinase 2 , HL-60 Cells , Lignans , Pharmacology , Oncogene Proteins , Signal Transduction , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL