1.Dehydrodiisoeugenol resists H1N1 virus infection via TFEB/autophagy-lysosome pathway.
Zhe LIU ; Jun-Liang LI ; Yi-Xiang ZHOU ; Xia LIU ; Yan-Li YU ; Zheng LUO ; Yao WANG ; Xin JIA
China Journal of Chinese Materia Medica 2025;50(6):1650-1658
The present study delves into the cellular mechanisms underlying the antiviral effects of dehydrodiisoeugenol(DEH) by focusing on the transcription factor EB(TFEB)/autophagy-lysosome pathway. The cell counting kit-8(CCK-8) was utilized to assess the impact of DEH on the viability of human non-small cell lung cancer cells(A549). The inhibitory effect of DEH on the replication of influenza A virus(H1N1) was determined by real-time quantitative polymerase chain reaction(RT-qPCR). Western blot was employed to evaluate the influence of DEH on the expression level of the H1N1 virus nucleoprotein(NP). The effect of DEH on the fluorescence intensity of NP was examined by the immunofluorescence assay. A mouse model of H1N1 virus infection was established via nasal inhalation to evaluate the therapeutic efficacy of 30 mg·kg~(-1) DEH on H1N1 virus infection. RNA sequencing(RNA-seq) was performed for the transcriptional profiling of mouse embryonic fibroblasts(MEFs) in response to DEH. The fluorescent protein-tagged microtubule-associated protein 1 light chain 3(LC3) was used to assess the autophagy induced by DEH. Western blot was employed to determine the effect of DEH on the autophagy flux of LC3Ⅱ/LC3Ⅰ under viral infection conditions. Lastly, the role of TFEB expression in the inhibition of DEH against H1N1 infection was evaluated in immortalized bone marrow-derived macrophage(iBMDM), both wild-type and TFEB knockout. The results revealed that the half-maximal inhibitory concentration(IC_(50)) of DEH for A549 cells was(87.17±0.247)μmol·L~(-1), and DEH inhibited H1N1 virus replication in a dose-dependent manner in vitro. Compared with the H1N1 virus-infected mouse model, the treatment with DEH significantly improved the body weights and survival time of mice. DEH induced LC3 aggregation, and the absence of TFEB expression in iBMDM markedly limited the ability of DEH to counteract H1N1 virus replication. In conclusion, DEH exerts its inhibitory activity against H1N1 infection by activating the TFEB/autophagy-lysosome pathway.
Influenza A Virus, H1N1 Subtype/genetics*
;
Animals
;
Autophagy/drug effects*
;
Humans
;
Mice
;
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics*
;
Influenza, Human/metabolism*
;
Lysosomes/metabolism*
;
Orthomyxoviridae Infections/genetics*
;
Eugenol/pharmacology*
;
Antiviral Agents/pharmacology*
;
Virus Replication/drug effects*
;
A549 Cells
;
Male
2.A novel fully human LAG-3 monoclonal antibody LBL-007 combined with PD-1 antibody inhibits proliferation, migration and invasion of tumor cells via blocking NF-κB pathway.
Huinan ZHOU ; Jianfei LIU ; Chenglin WU ; Kewei QIN ; Lijun ZHOU
Chinese Journal of Cellular and Molecular Immunology 2025;41(5):398-405
Objective To investigate the effects of LBL-007, a novel fully human lymphocyte activation gene 3 (LAG-3) monoclonal antibody, in combination with programmed cell death protein 1 (PD-1) antibody, on the invasion, migration and proliferation of tumor cells, and to elucidate the underlying mechanisms. Methods Human lymphocyte cells Jurkat were co-cultured with A549 and MGC803 tumor cell lines and treated with the isotype control antibody human IgG, LBL-007, anti-PD-1 antibody BE0188, or tumor necrosis factor-alpha (TNF-α, the NF-κB signaling pathway agonist). Tumor cell proliferation was assessed using a colony formation assay; invasion was measured by TranswellTM assay; migration was evaluated using a wound healing assay. Western blotting was employed to determine the expression levels of NF-κB pathway-related proteins: IκB inhibitor kinase alpha (Ikkα), phosphorylated Ikkα (p-IKKα), NF-κB subunit p65, phosphorylated p65 (p-p65), NF-κB Inhibitor Alpha (IκBα), phosphorylated IκBα (p-IκBα), matrix metalloproteinase 9 (MMP9), and MMP2. Results Compared with the control and IgG isotype groups, LBL-007 and BE0188 significantly reduced tumor cell proliferation, invasion, and migration. They also decreased the phosphorylation of p-IKKα, p-p65 and p-IκBα, and the expression of MMP9 and MMP2 of tumor cells in the co-culture system. The combined treatment of LBL-007 and BE0188 enhanced inhibitory effects. Treatment with the NF-κB signaling pathway agonist TNF-α reversed the suppressive effects of LBL-007 and BE0188 on tumor cell proliferation, invasion, migration, and NF-κB signaling. Conclusion LBL-007 and anti-PD-1 antibody synergistically inhibit the invasion, migration, and proliferation of A549 and MGC803 tumor cells by blocking the NF-κB signaling pathway.
Humans
;
Cell Proliferation/drug effects*
;
Cell Movement/drug effects*
;
Signal Transduction/drug effects*
;
NF-kappa B/metabolism*
;
Neoplasm Invasiveness
;
Antibodies, Monoclonal/pharmacology*
;
Programmed Cell Death 1 Receptor/antagonists & inhibitors*
;
Cell Line, Tumor
;
Antigens, CD/immunology*
;
Lymphocyte Activation Gene 3 Protein
;
A549 Cells
;
I-kappa B Kinase/metabolism*
;
Jurkat Cells
;
Matrix Metalloproteinase 9/metabolism*
3.Preliminary study on the role of TM9SF2 knockdown in promoting the activity of the type I interferon signaling pathway to inhibit vesicular stomatitis virus replication.
Kang LI ; Xinyu WANG ; Ran YE ; Lingyun GUO ; Linxu WANG ; Nuo XU ; Tong ZHANG ; Xiaotao DUAN
Chinese Journal of Cellular and Molecular Immunology 2025;41(6):481-487
Objective To explore the effect of the knockdown of transmembrane 9 superfamily protein member 2 (TM9SF2) on the replication of vesicular stomatitis virus (VSV), and investigate its role in the mechanism of antiviral innate immunity. Methods Small interfering RNA (siRNA) was used to knock down the TM9SF2 gene in human non-small cell lung cancer A549 cells. The CCK-8 method was used to assess cell proliferation. A VSV-green fluorescent protein (VSV-GFP) infected cell model was established. The plaque assay was used to measure the viral titer in the supernatant. RT-qPCR and Western blotting were employed to quantify the mRNA and protein levels of VSV genome replication in A549 cells following VSV infection, as well as the expression of interferon β (IFN-β) mRNA and interferon regulatory factor 3 (IRF3) protein phosphorylation following polyinosinic-polycytidylic acid (poly(I:C)) stimulation. Results Compared to the negative control, the knockdown of TM9SF2 exhibited a significant effect, with no observed impact on A549 cell proliferation. The VSV-GFP infected A549 cell model was successfully established. After viral stimulation, fluorescence intensity was reduced following TM9SF2 knockdown, and the mRNA and protein levels of VSV were significantly downregulated. The viral titer of VSV was decreased. After poly(I:C) stimulation, TM9SF2 knockdown significantly upregulated the mRNA level of IFN-β and the phosphorylation level of IRF3 protein. Conclusion The knockdown of TM9SF2 inhibits the replication of vesicular stomatitis virus, and positively regulates the type I interferon signaling pathway, thus enhancing the host's antiviral innate immune response.
Humans
;
Virus Replication/genetics*
;
Signal Transduction
;
Membrane Proteins/metabolism*
;
A549 Cells
;
Vesiculovirus/physiology*
;
Interferon-beta/metabolism*
;
Interferon Regulatory Factor-3/genetics*
;
Interferon Type I/metabolism*
;
Vesicular Stomatitis/immunology*
;
Gene Knockdown Techniques
;
Vesicular stomatitis Indiana virus/physiology*
;
RNA, Small Interfering/genetics*
4.TIPE2 inhibits the stemness of lung cancer cells by regulating the phenotypic polarization of tumor-associated macrophages.
Chinese Journal of Cellular and Molecular Immunology 2025;41(8):680-686
Objective To investigate the regulatory effect of tumor necrosis factor-α-induced protein-8-like factor 2 (TIPE2) on the phenotype of lung cancer tumor-associated macrophages (TAM) and its influence on the stemness of lung cancer cells. Methods Mouse macrophage cell line RAW264.7 was cultured and infected with either LV-TIPE2 lentivirus or negative control LV-NC lentivirus. The TIPE2 expression in infected cells was assessed by real-time quantitative PCR (RT-qPCR) and Western blotting to verify transfection efficiency. The infected RAW264.7 cells were co-cultured with lung cancer cell line A549, and were divided into four groups: control group (RAW264.7 cells or A549 cells cultured alone), TAM group (RAW264.7 cells co-cultured with A549 cells), LV-NC group (RAW264.7 cells infected with LV-NC and co-cultured with A549 cells), LV-TIPE2 group (RAW264.7 cells infected with LV- TIPE2 and co-cultured with A549 cells). The RAW264.7 cells were collected after co-culture, and the expression of mannose receptor (CD206) protein of M2 macrophages was detected by cellular immunofluorescence staining. The proportions of M1 and M2 macrophages were detected by flow cytometry. After co-culture, A549 cells were collected, and their activity was assessed by CCK-8 assay. Self-renewal ability was evaluated using tumor cell pelleting experiment. The expression of stemness marker proteins-including cluster of differentiation 133 (CD133), transmembrane adhesion molecule (CD44), sex-determining region Y-box protein 2 (SOX2) and octamer-binding transcription factor 4 (OCT4)-was detected by Western blot. Results Compared with the control group or LV-NC group, the relative mRNA and protein expression levels of TIPE2 in RAW264.7 cells from the LV-TIPE2 group were significantly upregulated. Compared with the control group, the fluorescence intensity of M2-type macrophage marker CD206 protein in RAW264.7 cells from the TAM group was significantly increased, the proportion of M1-type macrophages was significantly decreased, and the proportion of M2-type macrophages was significantly increased. In contrast, compared with the TAM group, the fluorescence intensity of CD206 protein in RAW264.7 cells from the LV-TIPE2 group was significantly decreased, the proportion of M1-type macrophages was significantly increased, and the proportion of M2-type macrophages was significantly decreased. Compared with the control group, the proliferation activity of A549 cells in TAM group was significantly increased, the number of tumor pellet formation was significantly increased, and the relative expression levels of CD133, CD44, SOX2 and OCT4 were significantly up-regulated. However, compared with the TAM group, the proliferation activity of A549 cells from the LV-TIPE2 group was significantly decreased, the number of tumor pellet formation was significantly decreased, and the relative expression levels of CD133, CD44, SOX2 and OCT4 were significantly decreased. Conclusion TIPE2 can suppress the stemness of lung cancer cells by inhibiting the polarization of macrophages to M2-type, thereby exerting an anticancer effect.
Animals
;
Mice
;
Humans
;
Tumor-Associated Macrophages/metabolism*
;
Lung Neoplasms/genetics*
;
Intracellular Signaling Peptides and Proteins/metabolism*
;
RAW 264.7 Cells
;
A549 Cells
;
Phenotype
;
Coculture Techniques
;
Receptors, Cell Surface/metabolism*
;
Neoplastic Stem Cells/metabolism*
;
Mannose Receptor
;
Mannose-Binding Lectins/metabolism*
;
Lectins, C-Type/metabolism*
;
Cell Polarity
;
Macrophages/metabolism*
5.Curcumin inhibits lipid metabolism in non-small cell lung cancer by downregulating the HIF-1α pathway.
Dandan LI ; Jiaxin CHU ; Yan YAN ; Wenjun XU ; Xingchun ZHU ; Yun SUN ; Haofeng DING ; Li REN ; Bo ZHU
Journal of Southern Medical University 2025;45(5):1039-1046
OBJECTIVES:
To investigate the effect of curcumin on lipid metabolism in non-small cell lung cancer (NSCLC) and its molecular mechanism.
METHODS:
The inhibitory effect of curcumin (0-70 μmol/L) on proliferation of A549 and H1299 cells was assessed using MTT assay, and 20 and 40 μmol/L curcumin was used in the subsequent experiments. The effect of curcumin on lipid metabolism was evaluated using cellular uptake assay, wound healing assay, triglyceride (TG)/free fatty acid (NEFA) measurements, and Oil Red O staining. Western blotting was performed to detect the expressions of PGC-1α, PPAR-α, and HIF-1α in curcumin-treated cells. Network pharmacology was used to predict the metabolic pathways, and the results were validated by Western blotting. In a nude mouse model bearing A549 cell xenograft, the effects of curcumin (20 mg/kg) on tumor growth and lipid metabolism were assessed by measuring tumor weight and observing the changes in intracellular lipid droplets.
RESULTS:
Curcumin concentration-dependently inhibited the proliferation of A549 and H1299 cells and significantly reduced TG and NEFA levels and intracellular lipid droplets. Western blotting revealed that curcumin significantly upregulated PGC-1α and PPAR‑α expressions in the cells. KEGG pathway enrichment analysis predicted significant involvement of the HIF-1 signaling pathway in curcumin-treated NSCLC, suggesting a potential interaction between HIF-1α and PPAR‑α. Western blotting confirmed that curcumin downregulated the expression of HIF-1α. In the tumor-bearing mice, curcumin treatment caused significant reduction of the tumor weight and the number of lipid droplets in the tumor cells.
CONCLUSIONS
Curcumin inhibits NSCLC cell proliferation and lipid metabolism by downregulating the HIF-1α pathway.
Curcumin/pharmacology*
;
Humans
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Animals
;
Lipid Metabolism/drug effects*
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
Lung Neoplasms/pathology*
;
Mice, Nude
;
Down-Regulation
;
Mice
;
Cell Proliferation/drug effects*
;
Cell Line, Tumor
;
Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
;
PPAR alpha/metabolism*
;
Signal Transduction/drug effects*
;
A549 Cells
6.Inhibitory effect of Fuzheng Huaji Decoction against non-small cell lung cancer cells in vitro and the possible molecular mechanism.
Lijun HE ; Xiaofei CHEN ; Chenxin YAN ; Lin SHI
Journal of Southern Medical University 2025;45(6):1143-1152
OBJECTIVES:
To investigate the inhibitory effect of Fuzheng Huaji Decoction against non-small cell lung cancer (NSCLC) cells in vitro and explore the underlying mechanism.
METHODS:
The active ingredients and targets of Fuzheng Huaji Decoction were identified using TCMSP and SwissTargetPrediction databases. NSCLC-related targets from GeneCards and PharmGKB were intersected with the targets of the Decoction, and a protein-protein interaction (PPI) network was constructed to identify the core targets, which were analyzed with GO and KEGG pathway enrichment analysis. Cultured A549 cells were treated with different concentrations of Fuzheng Huaji Decoction-medicated serum, and the changes in cell proliferation, apoptosis, and protein expressions were examined using CCK-8 assay, annexin V-FITC/PI staining and Western blotting.
RESULTS:
Fuzheng Huaji Decoction contained 140 active ingredients, and 707 drug-disease intersecting targets were identified. Among these targets, TP53, AKT1, HIF1A, GAPDH, ALB, EGFR, CTNNB1, and TNF were identified as the core targets which were involved in the biological processes related to kinases and receptors and the PI3K-AKT, Ras, calcium, and MAPK pathways. Molecular docking studies indicated strong binding affinity of the active ingredients with TP53, AKT1, and HIF1A. In cultured A549 cells, treatment with 2.5%, 5%, and 10% Fuzheng Huaji Decoction-medicated serum significantly inhibited cell proliferation, promoted cell apoptosis, and downregulated the expression levels of HIF1A, p-AKT (Thr308), and TP53 proteins.
CONCLUSIONS
Fuzheng Huaji Decoction inhibits proliferation of NSCLC cells possibly by downregulating the expressions of HIF1A, p-AKT (Thr308), and TP53.
Humans
;
Carcinoma, Non-Small-Cell Lung/metabolism*
;
Drugs, Chinese Herbal/pharmacology*
;
Cell Proliferation/drug effects*
;
Apoptosis/drug effects*
;
Lung Neoplasms/metabolism*
;
A549 Cells
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Protein Interaction Maps
;
Signal Transduction/drug effects*
;
Cell Line, Tumor
7.LncRNA SNHG15 promotes proliferation, migration and invasion of lung adenocarcinoma cells by regulating COX6B1 through sponge adsorption of miR-30b-3p.
Xiuying GONG ; Shunfu HOU ; Miaomiao ZHAO ; Xiaona WANG ; Zhihan ZHANG ; Qinghua LIU ; Chonggao YIN ; Hongli LI
Journal of Southern Medical University 2025;45(7):1498-1505
OBJECTIVES:
To explore the molecular mechanism by which lncRNA SNHG15 regulates proliferation, invasion and migration of lung adenocarcinoma cells.
METHODS:
The lncRNA microarray chip dataset GSE196584 and LncBase were used to predict the lncRNAs that interact with miR-30b-3p, and their association with patient prognosis were investigated using online databases, after which lncRNA nucleolar RNA host gene 15 (SNHG15) was selected for further analysis. The subcellular localization of lncRNA SNHG15 and its expression levels in normal human lung epithelial cells and lung adenocarcinoma cell lines were detected using fluorescence in situ hybridization and qRT-PCR. In cultured A549 cells, the changes in cell proliferation, migration, and invasion following transfection with a SNHG15 knockdown plasmid (sh-SNHG15), a miR-30b-3p inhibitor, or their co-transfection were assessed with EdU, wound healing, and Transwell assays. Bioinformatics analyses were used to predict the regulatory relationship between lncRNA SNHG15 and COX6B1, and the results were verified using Western blotting and rescue experiments in A549 cells transfected with sh-SNHG15, a COX6B1-overexpressing plasmid, or both.
RESULTS:
LncRNA SNHG15 was shown to target miR-30b-3p, and the former was highly expressed in lung adenocarcinoma, and associated with a poor patient prognosis. LncRNA SNHG15 was localized in the cytoplasm and expressed at higher levels in A549 and NCI-H1299 cells than in BEAS-2B cells. In A549 cells, lncRNA SNHG15 knockdown significantly inhibited cell migration, invasion and proliferation, and these changes were reversed by miR-30b-3p inhibitor. A regulatory relationship was found between lncRNA SNHG15 and COX6B1, and their expression levels were positively correlated (r=0.128, P=0.003). MiR-30b-3p knockdown obviously decreased COX6B1 expression in A549 cells, and COX6B1 overexpression rescued the cells from the inhibitory effects of lncRNA-SNHG15 knockdown.
CONCLUSIONS
LncRNA SNHG15 may compete with COX6B1 to bind miR-30b-3p through a ceRNA mechanism to affect proliferation, migration, and invasion of lung adenocarcinoma cells.
Humans
;
MicroRNAs/metabolism*
;
RNA, Long Noncoding/genetics*
;
Cell Proliferation
;
Cell Movement
;
Lung Neoplasms/genetics*
;
Adenocarcinoma of Lung
;
Neoplasm Invasiveness
;
A549 Cells
;
Adenocarcinoma/genetics*
;
Gene Expression Regulation, Neoplastic
;
Cell Line, Tumor
8.Effects of Electromagnetic Pulses on Exosomes Secretion by A549 Cells.
Qingxia HOU ; Yingmei WANG ; Meng CAO ; Jiangzheng LIU ; Deqin KONG ; Qian ZHANG ; Weihua YU ; Guangzhou AN
Chinese Journal of Lung Cancer 2024;27(12):885-893
BACKGROUND:
Numerous researches indicated that electromagnetic pulses (EMP) possessed advantages such as strong targeting, minimal side-effects and low treatment cost in tumor therapy, but its optimum parameters for treatment and the relationship between EMP and tumor-derived exosomes remains unclear. This study aims to clarify the effects of EMP with different parameters on the quantity and miRNA (microRNA) of exosomes secreted by human non-small cell lung cancer A549 cells, providing beneficial reference for the clinical application of EMP and related research.
METHODS:
A549 cells were randomly divided into control group and different EMP radiation groups with respective intensity of 400, 600 and 800 kV/m. EMP was performed with 2000 pulses once, 20 Hz of repetition frequency and 120 ns of pulse width. A549 cells were radiated once per day for continuous 3 days. After radiation, exosomes were collected and identified; cell number was measured by trypan blue staining; the concentration of exosomes was measured by nanoparticle tracking analysis (NTA); the abundance of miRNAs was determined by miRNA sequencing.
RESULTS:
Compared with control group, the morphology and cell viability of A549 cells in radiation group was not different, but the quantity of exosomes in 400 or 800 kV/m radiation group was significantly decreased (P<0.05), in contrast with obvious increase in 600 kV/m radiation group (P<0.05). The abundance of exosomal miRNAs between control group and each EMP group was obviously different (P<0.05) and target genes of differentially abundant miRNAs enriched in different pathways.
CONCLUSIONS
Under the experimental condition, the quantity and miRNA abundance of exosomes could be changed by EMP radiation, which could further influence the function of tumor-derived exosomes.
Humans
;
Exosomes/genetics*
;
A549 Cells
;
MicroRNAs/metabolism*
;
Lung Neoplasms/pathology*
;
Cell Survival/radiation effects*
;
Electromagnetic Fields
9.Network Pharmacology Based Elucidation of Molecular Mechanisms of Laoke Formula for Treatment of Advanced Non-Small Cell Lung Cancer.
Yu-Yu FENG ; Jin-Feng LIU ; Yang XUE ; Dan LIU ; Xiong-Zhi WU
Chinese journal of integrative medicine 2024;30(11):984-992
OBJECTIVE:
To explore the specific pharmacological molecular mechanisms of Laoke Formula (LK) on treating advanced non-small cell lung cancer (NSCLC) based on clinical application, network pharmacology and experimental validation.
METHODS:
Kaplan-Meier method and Cox regression analysis were used to evaluate the survival benefit of Chinese medicine (CM) treatment in 296 patients with NSCLC in Tianjin Medical University Cancer Institute and Hospital from January 2011 to December 2015. The compounds of LK were screened using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, and the corresponding targets were performed from Swiss Target Prediction. NSCLC-related targets were obtained from Therapeutic Target Database and Comparative Toxicogenomics Database. Key compounds and targets were identified from the compound-target-disease network and protein-protein interaction (PPI) network analysis, respectively. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analysis were used to predict the potential signaling pathways involved in the treatment of advanced NSCLC with LK. The binding affinities between key ingredients and targets were further verified using molecular docking. Finally, A549 cell proliferation and migration assay were used to evaluate the antitumor activity of LK. Western blot was used to further verify the expression of key target proteins related to the predicted pathways.
RESULTS:
Kaplan-Meier survival analysis showed that the overall survival of the CM group was longer than that of the non-CM group (36 months vs. 26 months), and COX regression analysis showed that LK treatment was an independent favorable prognostic factor (P=0.027). Next, 97 components and 86 potential targets were included in the network pharmacology, KEGG and GO analyses, and the results indicated that LK was associated with proliferation and apoptosis. Moreover, molecular docking revealed a good binding affinity between the key ingredients and targets. In vitro, A549 cell proliferation and migration assay showed that the biological inhibition effect was more obvious with the increase of LK concentration (P<0.05). And decreased expressions of nuclear factor κB1 (NF-κB1), epidermal growth factor receptor (EGFR) and AKT serine/threonine kinase 1 (AKT1) and increased expression of p53 (P<0.05) indicated the inhibitory effect of LK on NSCLC by Western blot.
CONCLUSION
LK inhibits NSCLC by inhibiting EGFR/phosphoinositide 3-kinase (PI3K)/AKT signaling pathway, NFκB signaling pathway and inducing apoptosis, which provides evidence for the therapeutic mechanism of LK to increase overall survival in NSCLC patients.
Humans
;
Carcinoma, Non-Small-Cell Lung/metabolism*
;
Lung Neoplasms/pathology*
;
Drugs, Chinese Herbal/therapeutic use*
;
Network Pharmacology
;
Female
;
Male
;
Molecular Docking Simulation
;
Middle Aged
;
Protein Interaction Maps/drug effects*
;
A549 Cells
;
Cell Proliferation/drug effects*
;
Signal Transduction/drug effects*
;
Cell Movement/drug effects*
;
Kaplan-Meier Estimate
;
Aged
;
ErbB Receptors/metabolism*
10.Luteolin inhibits proliferation of lung cancer A549 cells by increasing ROS production and inhibiting the AKT/mTOR signaling pathway and HO-1 expression.
Huan LI ; Zixin QIU ; Wenjie XU ; Xue CHEN ; Diandian WEI ; Yun WANG
Journal of Southern Medical University 2024;44(12):2367-2374
OBJECTIVES:
To investigate the mechanism of luteolin for inhibiting proliferation of lung cancer A549 cells.
METHODS:
A549 cells treated with different concentrations of luteolin for 48 h were evaluated for changes in cell viability, proliferation, reactive oxygen species (ROS) production and apoptosis using MTT assay, plate cloning assay, EdU staining, DCFH-DA assay and Hoechst33258 staining. The changes in cell autophagy were examined with MDC staining, and the expressions of apoptosis-related proteins (Bax, Bcl-2, and cleaved caspase-9), autophagy-related proteins (LC3B, Beclin 1, and P62), AKT/mTOR pathway proteins, and HO-1 protein were detected using Western blotting.
RESULTS:
Treatment with luteolin dose-dependently inhibited the viability and proliferation of A549 cells, increased intracellular ROS levels, up-regulated the expressions of Bax, cleaved caspase-9, and Beclin 1, increased the LC3B-II/LC3B-I ratio, down-regulated the expressions of Bcl-2 and P62, and induced cell apoptosis and autophagy. Luteolin also significantly inhibited the phosphorylation of AKT and mTOR and down-regulated the expression of HO-1 protein in the cells.
CONCLUSIONS
Luteolin induces apoptosis and autophagy to inhibit proliferation of A549 cells by increasing ROS production, inhibiting the AKT/mTOR pathway and down-regulating HO-1 protein expression.
Humans
;
TOR Serine-Threonine Kinases/metabolism*
;
A549 Cells
;
Reactive Oxygen Species/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Cell Proliferation/drug effects*
;
Signal Transduction/drug effects*
;
Lung Neoplasms/pathology*
;
Apoptosis/drug effects*
;
Luteolin/pharmacology*
;
Autophagy/drug effects*
;
Heme Oxygenase-1/metabolism*
;
Cell Survival/drug effects*

Result Analysis
Print
Save
E-mail