Effects of Intermittent Cisplatin Intervention on 6PGD Levels and Tumor Growth and Migration in A549 Lung Cancer Cells

2021 ◽  
Vol 8 ◽  
Author(s):  
WU Qiang ◽  
TANG Lin-dong ◽  
WU Yan-hui ◽  
LI Qiang ◽  
YU Li

Aim: To investigate the changes of A549 protein 6PGD expression, tumor growth and migration, NADPH and ROS levels in lung cancer cells after intermittent intervention with DDP. Methods: The sensitivity of A549 cells to DDP was detected by cck-8 method, A549 cells were treated by DDP intermitten, 6PGD protein expression was detected by Western blot method, and the ROS level of A549 cells was determined by ROS kit. NADPH levels of A549 cells were determined by NADPH kit. Result: After DDP intermittent intervention, the expression of 6PGD protein in lung cancer cells A549 was increased, the growth and migration of A549 cells were significantly inhibited, and ROS and NADPH levels were significantly increased. Conclusion: 6PGD were upregulated in after DDP intermittent intervention and affected the migration ability in lung cancer cells.

2017 ◽  
Vol 12 (1) ◽  
pp. 200-205 ◽  
Author(s):  
Bing Wang ◽  
Zhanjie Zuo ◽  
Fang Lv ◽  
Liang Zhao ◽  
Minjun Du ◽  
...  

AbstractAimsAccumulating evidence indicates that aberrant expression of miR-107 plays a crucial role in cancers. This study aims to display the function of miR-107 and its novel target genes in the progression of lung cancer.Methods and MaterialMiR-107 or miR-107 inhibitor was transfected into lung cancer cells A549. The levels of miR-107 and TP53 regulated inhibition of apoptosis 1 (TRIAP1) were examined by quantitative real-time Polymerase Chain Reaction (qRT-PCR) analysis and Western Blot. Functionally, MTT and colony formation assays were carried out to test the effect of miR-107 inhibitor and/or small interference RNA (siRNA) targeting TRIAP1 mRNA on proliferation of lung cancer cells. Levels of miR-107 or TRIAP1 were detected in clinical lung cancer samples by using qRT-PCR analysis.ResultsQRT-PCR analysis revealed that miR-107 inhibitor or miR-107 was successfully transfected into A549 cells. Western Blot indicated that miR-107 decreased the expression of TRIAP1 protein in the cells. In contrast, miR-107 inhibitor augmented the levels of TRIAP1 protein. Functionally, miR-107 inhibitor remarkably suppressed A549 cell proliferation, whereas, TRIAP1 siRNAs could abrogate the miR-107 inhibitor-induced proliferation of cells. Then, we validated that TRIAP1 was increased in clinical lung cancer samples. MiR-107 expression was negatively related to TRIAP1 expression in clinical lung cancer samples.ConclusionsMiR-107 suppresses cell proliferation by targeting TRIAP1 in lung cancer. Our finding allows new insights into the mechanisms of lung cancer that is mediated by miR-107.


2010 ◽  
Vol 29 (7) ◽  
pp. 607-614 ◽  
Author(s):  
Yong Hwan Han ◽  
Woo Hyun Park

Carbobenzoxy-Leu-Leu-leucinal (MG132) as a proteasome inhibitor has been shown to induce apoptotic cell death through formation of reactive oxygen species (ROS). In the present study, we evaluated the effects of MG132 on the growth of A549 lung cancer cells in relation to cell growth, ROS and glutathione (GSH) levels. Treatment with MG132 inhibited the growth of A549 cells with an IC50 of approximately 20 μM at 24 hours. DNA flow cytometric analysis indicated that 0.5 ∼ 30 μM MG132 induced a G1 phase arrest of the cell cycle in A549 cells. Treatment with 10 or 30 μM MG132 also induced apoptosis, as evidenced by sub-G1 cells and annexin V staining cells. This was accompanied by the loss of mitochondrial membrane potential (MMP; Δψm). The intracellular ROS levels including O2•- were strongly increased in 10 or 30 μM MG132-treated A549 cells but were down-regulated in 0.1, 0.5 or 1 μM MG132-treated cells. Furthermore, 10 or 30 μM MG132 increased mitochondrial O2•- level but 0.1, 0.5 or 1 μM MG132 decreased that. In addition, 10 or 30 μM MG132 induced GSH depletion in A549 cells. In conclusion, MG132 inhibited the growth of human A549 cells via inducing the cell cycle arrest as well as triggering apoptosis, which was in part correlated with the changes of ROS and GSH levels. Our present data provide important information on the anti-growth mechanisms of MG132 in A549 lung cancer cells in relation to ROS and GSH.


2021 ◽  
Vol 11 (7) ◽  
pp. 1339-1345
Author(s):  
Zhijie Shang ◽  
Yuxuan Wang ◽  
Lixun Chai ◽  
Gengpu Yang

The aim of the present study was to investigate the mechanism by which pumilio RNA binding family member 2 (PUM2), an RNA-binding protein (RBP) of C-X-C chemokine receptor type 4 (CXCR4), exerts its effects on the development of lung cancer. RT-qPCR and western blot analysis were utilized to measure the expression of PUM2 in several lung cancer cell lines. Cell Counting Kit-8 (CCK-8), colony formation assay, transwell- and wound healing assays were employed to determine the proliferation, invasion and migration of NCI-H520 cells, respectively. Next, the expression of CXCR4 was measured using western blot analysis, and the combination between PUM2 and CXCR4 was verified by RNA immunoprecipitation (RIP) assay and RNA pull down assay. Finally, whether the expression of PUM2 can affect the Ca2+ signaling pathway was confirmed by western blot assay. Results revealed that the expression level of PUM2 was notably upregulated in lung cancer cells, and knockdown of PUM2 significantly inhibited the proliferation, invasion and migration of NCI-H520 cells. PUM2 was confirmed to be the RBP of CXCR4, and PUM2 knockdown decreased the expression of CXCR4. In addition, PUM2 silencing inhibited the phosphorylation of CaMKII, ERK, and MEK. Taken together, these findings demonstrated that PUM2 could promote the proliferation and metastasis of lung cancer cells by regulating Ca2+ signaling pathway via targeting CXCR4, which may provide a novel insight for the future treatment of lung cancer.


2019 ◽  
Vol 14 (1) ◽  
pp. 262-274
Author(s):  
Shu Zhao ◽  
Peng Li ◽  
Peng Wang ◽  
Jing Yang ◽  
Peng Song ◽  
...  

AbstractObjectiveMitochondrial homeostasis is vital for the progression of lung cancer. Nurr1 has been identified as a novel mediator of mitochondrial homeostasis in several types of cancers. The aim of our study was to investigate whether Nurr1 modulates the viability of A549 lung cancer cells by inducing mitochondrial dysfunction, with a focus on the p53-Drp1 signaling pathway.Methodswestern blotting, ELISA and immunofluorescence assay was used to verify the alterations of cell death. siRNA was used to determine the role of p53-Drp1 pathway in lung cancer death.ResultsNurr1 was downregulated in A549 lung cancer cells compared to normal pulmonary epithelial cells. Interestingly, overexpression of Nurr1 reduced the viability of A549 lung cancer cells by activating apoptosis and mitochondrial stress. At the molecular level, we provide data to support the regulatory effects of Nurr1 on the p53-Drp1 signaling pathway. Blockade of the p53-Drp1 signaling pathway abolished the proapoptotic action of Nurr1 on A549 cells and sustained mitochondrial homeostasis.ConclusionTaken together, our results depict the tumor-suppressive role played by Nurr1 in A549 lung cancer in vitro and show that the anticancer effects of Nurr1 are executed via triggering of mitochondrial dysfunction and activation of the p53-Drp1 signaling pathway.


2004 ◽  
Vol 286 (1) ◽  
pp. L81-L86 ◽  
Author(s):  
S. Buckley ◽  
W. Shi ◽  
B. Driscoll ◽  
A. Ferrario ◽  
K. Anderson ◽  
...  

Lung cancer is the most common visceral malignancy in males, with rapidly increasing incidence in females, and a devastatingly poor prognosis. Transforming growth factor (TGF)-β has been shown to induce senescence in A549 lung cancer cells, and both TGF-β and bone morphogenetic protein (BMP) 2 can suppress the transformed phenotype of A549 cells in vitro. We examined the effects of BMP4, another member of the TGF-β superfamily, on specific oncogenic properties of A549 cancer cells. When A549 cancer cells were treated continuously with 100 ng/ml of BMP4, a senescent phenotype was observed after 2 wk of treatment. The BMP-treated cells appeared larger than untreated cells, grew more slowly, had more senescence-associated β-galactosidase activity, and had less telomerase activity, as measured by the telomeric repeat amplification protocol assay. Invasion through Engelbreth Holm-Swarm matrix was inhibited in the senescent cell population. Senescent BMP4-treated cells had lower ERK activation, VEGF expression, and Bcl2 expression than wild-type cells, consistent with a less proliferative, less angiogenic phenotype with increased susceptibility to death by apoptosis. BMP4 treatment also resulted in sustained elevation of Smad1. In vivo xenograft studies in the flanks of nude mice confirmed that the BMP-treated cells were significantly less tumorigenic than untreated cells. Direct overexpression of Smad1 using adenoviral constructs resulted in cell death within 5 days. These studies suggest that BMP4 pathway signaling can induce senescence and thus negatively regulate the growth of A549 lung cancer cells.


2016 ◽  
Vol 35 (4) ◽  
pp. 2053-2064 ◽  
Author(s):  
YANLI WANG ◽  
HUI KONG ◽  
XIAONING ZENG ◽  
WENRUI LIU ◽  
ZAILIANG WANG ◽  
...  

2021 ◽  
Vol 18 (9) ◽  
pp. 1867-1872
Author(s):  
Zhang Hu ◽  
Duan Jing

Purpose: To investigate the effect of Scutellaria barbata D. Don extract (SBDE) on apoptosis and proliferation in A549 human lung cancer cells. Methods: Inverted microscope was used to observe morphological changes in A549 cells after exposure to SBDE. Trypan blue staining of living cells was applied to construct cell growth curve after treatment with varying concentrations of SBDE. The influence of SBDE on cell proliferation, apoptosis and cell cycle was determined by MTT assay while protein expressions of key apoptosis-related enzymes were evaluated by immuno-cytochemical method. Results: SBDE inhibited the growth of A549 lung cancer cells at a concentration range of 20 - 160 μg/mL. Flow cytometry showed that SBDE induced apoptosis in the A549 cells. The proportion of cells in G0/G1-phase increased significantly (p < 0.01), while the proportion of cells in S-phase and G2/Mphase decreased correspondingly, indicating that the cells were in G0/G1-phase arrest. Cell cycle arrest and apoptosis-inducing effect gradually increased with increase in SBDE concentration. With increasing concentrations of SBDE, there were significant increases in the expressions of caspase-3 (p < 0.05), caspase-8 (p < 0.01) and caspase-9 (p < 0.05), and significant decreases in Ki-67 (p < 0.01) and p21 ras protein (p < 0.01). Conclusion: SBDE exerts significant inhibitory effect on the proliferation of A549 lung cancer cells, which can be developed for the treatment of lung cancer patients.


Molecules ◽  
2019 ◽  
Vol 24 (3) ◽  
pp. 407 ◽  
Author(s):  
Mixo Sibiya ◽  
Lerato Raphoko ◽  
Dikgale Mangokoana ◽  
Raymond Makola ◽  
Winston Nxumalo ◽  
...  

Despite major advancements in the development of various chemotherapeutic agents, treatment for lung cancer remains costly, ineffective, toxic to normal non-cancerous cells, and still hampered by a high level of remissions. A novel cohort of quinoxaline derivatives designed to possess a wide spectrum of biological activities was synthesized with promising targeted and selective anticancer drug activity. Hence, this study was aimed at determining in vitro anticancer activity effects of a newly synthesized class of 3-(quinoxaline-3-yl) prop-2-ynyl quinoxaline derivatives on A549 lung cancer cells. An assessment of the quinoxaline derivatives ferric reducing power, free radical scavenging activity, cytotoxic activity, and ability to induce reactive oxygen species (ROS) production was performed using the Ferric Reducing Antioxidant Power (FRAP), 2,2-diphenyl-1-picryl-hydrazyl (DPPH), 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and 2’,7’-dichlorodihydrofluorescein diacetate (H2DCFDA) assays, respectively. The ability of the quinoxaline derivatives to induce apoptosis in A549 cells was assessed using the Acridine Orange/Ethidium Bromide (AO/EB) and Annexin V-FITC/Dead Cell Assay. Of the four quinoxaline derivatives tested, 3-(quinoxaline-3-yl) prop-2-ynyl methanosulphate (LA-39B) and 3-(quinoxaline-3-yl) prop-2-yn-1-ol (LA-55) displayed a dose-dependent reducing power, free-radical scavenging activity, inhibition of cell viability, and stimulation of ROS production which was accompanied by induction of apoptosis in A549 lung cancer cells. None of the quinoxaline derivatives induced cell death or ROS production in non-cancerous Raw 267.4 macrophage cells. Cytotoxicity was observed in A549 lung cancer, HeLa cervical cancer, and MCF-7 breast cancer cells albeit inhibition was more pronounced in A549 cells. The results of the study suggest that 3-(quinoxaline-3-yl) prop-2-ynyl methanosulphate and 3-(quinoxaline-3-yl) prop-2-yn-1-ol induce apoptotic cell death in A549 lung cancer cells.


Sign in / Sign up

Export Citation Format

Share Document