scholarly journals Metformin promotes apoptosis of A549 cells via regulation of p-AMPK protein expression, bax/bcl-2 ratio and ROS levels

2021 ◽  
Vol 20 (9) ◽  
pp. 1827-1832
Author(s):  
Ziying Yu ◽  
Fengtao Liu ◽  
Xiaoli Zhang

Purpose: To investigate the influence of metformin on apoptosis of pulmonary carcinoma cells (A549), and the associated mode of action.Methods: Pulmonary carcinoma cells in logarithmic growth phase were treated with graded concentrations of metformin, and the anti-proliferative and apoptotic effects of the drug were measured using MTT assay and flow cytometry, respectively. The levels of reactive oxygen species (ROS) in A549 cell suspension were determined with 2, 7- dihydrodichlorofluorescein diacetate (DCFH-DA) assay. The expression levels of phosphorylated AMP-activated protein kinase (p-AMPK), mammalian target of rapamycin (mTOR), and bax/bcl-2 ratio were measured using Western blotting and real-time fluorescence quantitative polymerase chain reaction (qRT-PCR).Results: Metformin significantly promoted A549 cell apoptosis, but suppressed its proliferative potential in a dose- and time-based fashion. The levels of ROS, superoxide anion and MDA in A549 cells were significantly and dose-dependently increased by metformin (p < 0.05). Moreover, metformin markedly upregulated the mRNA and protein expressions of p-AMPK as well as bax/bcl-2 ratio, but had no impact on the expression level of mTOR (p < 0.05).Conclusion: Metformin promotes apoptosis in A549 cells via regulation of p-AMPK protein expression, bax/bcl-2 ratio, and ROS levels, and hence may play a role in lung cancer therapy.

Author(s):  
Deqing Zhu ◽  
Xuan Li ◽  
Hao Gong ◽  
Jing Li ◽  
Xike Lu ◽  
...  

Background: The effects of transthyretin (TTR) over-expression on the proliferation and cell cycle of nonsmall cell lung cancer (NSCLC) A549 cells and its possible mechanism were verified. Methods: A total of 196 LC patients and 20 healthy controls were enrolled at Tianjin Hospital, Tianjin, China between Apr 2017 and Oct 2017. The serum TTR content was detected by ELISA. Through lentiviral transfection method, NSCLC cells were divided into non-transfected group (group A), negative control group (group B) transfected with empty vector and experimental group (group C) transfected with TTR overexpression. Cell proliferation was detected by CCK-8 method, TTR mRNA expression was detected by realtime quantitative polymerase chain reaction (RT-qPCR), and TTR protein expression was tested by Western blot (WB). Cell cycle was detected by flow cytometry, Wnt3a/β-catenin protein expression was detected by WB, and mRNA expression was detected by RT-qPCR. Results: The serum TTR content in early, middle and late LC group was remarkably lower than that in healthy group (P<0.05). Compared with late stage, TTR content in early and middle stages of LC group was higher, and the difference was statistically marked (P < 0.05). The absorbance value of group C was lower than that of groups A and B, indicating that the cell proliferation activity dramatically decreased, with statistically marked difference (P<0.05). LC A549 cells in group C were obviously blocked in G2M, with statistical significance (P<0.05). Conclusion: TTR over-expression can inhibit the proliferation of NSCLC A549 cells, and the expression is related to Wnt3a/β-catenin pathway. TTR in serum of patients was helpful for diagnosing LC and has certain clinical value.  


2019 ◽  
Vol 14 (1) ◽  
pp. 392-399
Author(s):  
Rong-Li Sun ◽  
Feng-Juan Liu ◽  
Xiao Wu ◽  
Li-Sheng Wang ◽  
Peng-Fei Wang ◽  
...  

AbstractObjectiveThe objective of this research is to investigate the expression and function of SKA3 in lung adenocarcinoma.MethodsThe mRNA expression level of SKA3 in lung adenocarcinoma and its association with clinic-pathological factors were analyzed using data obtained from the TCGA database. Small interfering RNA (siRNA) for SKA3 (si-SKA3) was used to down-regulate SKA3 in A549 cells. pcDNA3.1- SKA3 was used to overexpress SKA3 in A549 cells. The proliferation ability of A549 cells was determined via MTT assay and colony formation assay. A wound healing assay was performed to examine the migration ability of A549 cells. The protein expression of p-MEK, MEK, p-ERK and ERK were determined by western blot.ResultsWe found that SKA3 is up-regulated in lung adenocarcinoma compared to the normal lung tissues. Kaplan-Meier analysis showed that high SKA3 expression is markedly associated with poor prognosis in lung adenocarcinoma patients. SKA3 expression is significantly correlated with age, gender, pathologic-stage, pathologic-N and pathologic-M. Moreover, depleting SKA3 obviously inhibited A549 cell proliferation and migration in vitro, while overexpression of SKA3 notably increased A549 cell proliferation and migration. Western blot analysis showed that the protein expression ratio of p-MEK/MEK and p-ERK/ERK decreased noticeably after depleting SKA3.ConclusionSKA3 expression was enhanced and associated with poor prognosis in lung adenocarcinoma patients, and it might play a facilitating role in cell growth and motility by regulating the MAPK signaling pathway.


2021 ◽  
Author(s):  
Yun Gui ◽  
Jing Cheng ◽  
Zhiguo Chen

Abstract Oridonin, a compound from Rabdosia rubescens, has been shown to have a potency for the improvement of the antitumor effect of lentinan (LNT). In this study, we tested the effect of oridonin, LNT, and the combination of them on a normal human fetal lung fibroblast cell line MRC-5 and non-small cell lung cancer cell line A549. Then we tested their effects on metastasis and survival with a lung cancer mice model. The effects of them on the mRNA and protein expression of several regulatory factors in A549 and lung tissue were determined by QPCR and western blotting. Results showed that the viability of MRC-5 and A549 were not affected by 0-20 µg/ml oridonin. 0-300 µg/ml LNT did not affect the viability of MRC-5, but 50-400 µg/ml LNT inhibited the viability of A549. 20 µg/ml oridonin and 100 or 300 µg/ml LNT were used in the subsequent study. The oridonin, LNT, or the combination of both had no effect on MRC-5 cell viability. The oridonin had no effect on A549 cell viability but LNT suppressed A549 cell viability, and oridonin promoted the suppression of LNT on A549 cells. In vivo study showed that oridonin alone had no effect on metastasis and survival but LNT decreased metastasis and survival in mice. Oridonin improved the suppression of LNT against metastasis and further improved the survival rate. In both A549 and lung tissues, LNT increased the mRNA and protein expression of caspase-3, caspase-8, caspase-9, Bax, p53, p21, and IκB-α, reduced mRNA and protein expressions of Bcl-2 and NF-κB. Oridonin enhanced all the effects of LNT on cells. Our study demonstrated that oridonin enhanced the antitumor effects of LNT and is conducive to the development of oridonin and LNT as a novel cancer drug regimen.


2021 ◽  
Vol 12 (2) ◽  
Author(s):  
Yuqing Lou ◽  
Jianlin Xu ◽  
Yanwei Zhang ◽  
Wei Zhang ◽  
Xueyan Zhang ◽  
...  

AbstractEpidermal growth factor receptor (EGFR) is a key oncogene in lung adenocarcinoma (LUAD). Resistance to EGFR tyrosine kinase inhibitors is a major obstacle for EGFR-mutant LUAD patients. Our gene chip array, quantitative polymerase chain reaction validation, and shRNA-based high-content screening identified the Akt kinase lanthionine synthetase C-like protein 2 (LANCL2) as a pro-proliferative gene in the EGFR-mutant LUAD cell line PC9. Therefore, we investigated whether LANCL2 plays a role in promoting cell proliferation and drug resistance in EGFR-mutant LUAD. In silico clinical correlation analysis using the Cancer Genome Atlas Lung Adenocarcinoma dataset revealed a positive correlation between LANCL2 and EGFR expression and an inverse relationship between LANCL2 gain-of-function and survival in LUAD patients. The EGFR-mutant LUAD cell lines PC9 and HCC827 displayed higher LANCL2 expression than the non-EGFR-mutant cell line A549. In addition, LANCL2 was downregulated following gefitinib+pemetrexed combination therapy in PC9 cells. LANCL2 knockdown reduced proliferation and enhanced apoptosis in PC9, HCC827, and A549 cells in vitro and suppressed murine PC9 xenograft tumor growth in vivo. Notably, LANCL2 overexpression rescued these effects and promoted gefitinib + pemetrexed resistance in PC9 and HCC827 cells. Pathway analysis and co-immunoprecipitation followed by mass spectrometry of differentially-expressed genes in LANCL2 knockdown cells revealed enrichment of several cancer signaling pathways. In addition, Filamin A and glutathione S-transferase Mu 3 were identified as two novel protein interactors of LANCL2. In conclusion, LANCL2 promotes tumorigenic proliferation, suppresses apoptosis, and promotes gefitinib+pemetrexed resistance in EGFR-mutant LUAD cells. Based on the positive association between LANCL2, EGFR, and downstream Akt signaling, LANCL2 may be a promising new therapeutic target for EGFR-mutant LUAD.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shao-Yuan Chen ◽  
Koichi Tsuneyama ◽  
Mao-Hsiung Yen ◽  
Jiunn-Tay Lee ◽  
Jiun-Liang Chen ◽  
...  

AbstractTumor cells have long been recognized as a relative contraindication to hyperbaric oxygen treatment (HBOT) since HBOT might enhance progressive cancer growth. However, in an oxygen deficit condition, tumor cells are more progressive and can be metastatic. HBOT increasing in oxygen partial pressure may benefit tumor suppression. In this study, we investigated the effects of HBOT on solid tumors, such as lung cancer. Non-small cell human lung carcinoma A549-cell-transferred severe combined immunodeficiency mice (SCID) mice were selected as an in vivo model to detect the potential mechanism of HBOT in lung tumors. HBOT not only improved tumor hypoxia but also suppressed tumor growth in murine xenograft tumor models. Platelet endothelial cell adhesion molecule (PECAM-1/CD31) was significantly increased after HBOT. Immunostaining of cleaved caspase-3 was demonstrated and apoptotic tumor cells with nuclear debris were aggregated starting on the 14th-day after HBOT. In vitro, HBOT suppressed the growth of A549 cells in a time-dependent manner and immediately downregulated the expression of p53 protein after HBOT in A549 cells. Furthermore, HBOT-reduced p53 protein could be rescued by a proteasome degradation inhibitor, but not an autophagy inhibitor in A549 cells. Our results demonstrated that HBOT improved tissue angiogenesis, tumor hypoxia and increased tumor apoptosis to lung cancer cells in murine xenograft tumor models, through modifying the tumor hypoxic microenvironment. HBOT will merit further cancer therapy as an adjuvant treatment for solid tumors, such as lung cancer.


2000 ◽  
Vol 192 (4) ◽  
pp. 460-469 ◽  
Author(s):  
Ben Davidson ◽  
Aasmund Berner ◽  
Jahn M. Nesland ◽  
Bj�rn Risberg ◽  
Heidi S. Berner ◽  
...  

2007 ◽  
Vol 22 (4) ◽  
pp. 543-550 ◽  
Author(s):  
Qing-Feng Xiong ◽  
Yue Chen ◽  
Ling He ◽  
Cun-Liang Deng ◽  
Zhan-Wen Huang ◽  
...  

1997 ◽  
Vol 16 (11) ◽  
pp. 636-644 ◽  
Author(s):  
Christopher D Lindsay ◽  
Joy L Hambrook ◽  
Alison F Lailey

1 The A549 cell line was used to assess the toxicity of sulphur mustard (HD), using gentian violet (GV) and neutral red (NR) dyes as indicators of cell viability. It was found that exposure to concentrations in excess of 40 ?M HD resulted in a rapid onset of toxicity. 2 The ability of monoisopropylglutathione ester (MIPE) to protect A549 cells against the effects of a 100 ?M challenge dose ofHD was determined using the NR and GV assays. It was found that MIPE (8 mM) could protect cells against the effects ofHD though MIPE had to be present at the time of HD challenge. Cultures protected with MIPE were two times more viable than HD exposed cells 48 h after HD challenge when using the GV and NR assays to assess viability. Observations by phase contrast microscopy of NR and GV stained cultures confirmed these findings. Addition of MIPE after previously exposing the A549 cultures to HD (for up to 5 min) maintained cell viability at 72% compared to 37% for unprotected cultures, after which time viability fell significantly so that at 10 min there was no difference in viability between the MIPE treated and untreated cultures. 3 Pretreating A549 cultures with MIPE for 1 h followed by its removal prior to HD challenge did not maintain cell viability. Treatment of cultures with HD for 1 h followed by addition of MIPE did not maintain the viability of the cultures, thus the window within which it was possible for MIPE to rescue cell cultures from the effects of HD was of short duration. 4 High performance liquid chromatography was used to determine the biochemical basis of the actions of MIPE. It was found that whilst intracellular levels of cysteine were increased up to 40-fold following treatment of A549 cell cultures with MIPE, levels of reduced glutathione did not rise. The lack of protection seen in cultures pretreated with MIPE for 1 h prior to HD exposure suggests that raising intracellular cysteine levels was not an effective strategy for protecting cells from the effects of HD. The protection observed is probably due to extra cellular inactivation of HD by MIPE.


2021 ◽  
Vol 17 ◽  
Author(s):  
Om Prakash ◽  
Shazia Usmani ◽  
Amresh Gupta ◽  
Asif Jafri ◽  
Mohammad Fahad Ullah ◽  
...  

Background: In recent years, novel metabolites isolated from botanical sources have attracted much attention for traditional and therapeutic significance. The ethnopharmacological studies suggest that Ziziphus mauritiana is a common remedy against several kinds of ailments. Objective: The current study has evaluated the MeOH extract of Ziziphus mauritiana leaves (ZME) through physicochemical, phytochemical, and chromatographic fingerprinting analysis, which displayed an array of biometabolites of pharmacological significance including flavonoids. Methods: The extract was further examined for anticancer activities which revealed promising anticancer properties against human lung epithelial carcinoma cells (A549) and induction of apoptosis impart by ROS. The oxidative stress was evaluated in terms of production and accumulation of cytosolic extent of ROS whereas anticancer perspective was determined by MTT assay, cell morphology analysis, followed by nuclear condensation for the examination of apoptosis induction. Results: Finding suggests that the MeOH extract of ZME markedly exhibited promising anticancer activity against the A549 lung epithelial carcinoma cell. The ZME was found to be most active in the MTT assay against A549 cells while it was less toxic to normal cells. The intracellular ROS generation was remarkably induced by ZME, which correlated with the ability of the flavonoid-rich fractions in the MeOH extract to inhibit cell growth and might induce apoptosis. Conclusion: The present study provides useful insight concerning the promising anticancer potential of ZME against A549 lung carcinomas. However, the clinical correlation will be required for its authorization and in the discovery of significant and least noxious novel agents against lung carcinomas.


Sign in / Sign up

Export Citation Format

Share Document