1.Three new diterpenoids from whole herb of Carpesium cernuum.
Shu-Hui FENG ; Wei-Qing ZHANG ; Wei LIANG ; Chen YAN
China Journal of Chinese Materia Medica 2023;48(19):5244-5249
The study investigated the chemical constituents from the whole herb of Carpesium cernuum. Three new diterpenoids were isolated from the whole herb of C. cernuum by column chromatography on silica gel, Sephadex LH-20, and semi-preparative HPLC. Their structures were identified by MS, NMR and other spectral techniques. The isolates were identified as(5Z)-2-oxo-2, 10, 14-trimethylhexadeca-5, 13-diene-11α, 18-diol(1),(2E, 10E)-7-[(acetyloxy)methyl]-3, 11, 15-trimethylhexadeca-2, 10, 14-triene-1, 12α-diol(2),(2E, 6Z)-3, 11, 15-trimethylhexadeca-2, 6, 14-triene-1, 12α, 19-triol(3), respectively. The cytotoxic activity of compounds 1-3 were investigated with DU-145, MCF-7, and A549 cells by MTT. The results showed that compound 1 and 3 had certain inhibitory effects on MCF-7 cells, with the inhibition rates of 45.06% and 29.40%, respectively.
Humans
;
Asteraceae/chemistry*
;
MCF-7 Cells
;
Magnetic Resonance Spectroscopy
;
Chromatography, High Pressure Liquid
;
A549 Cells
2.Identification of Bulbocodin D and C as novel STAT3 inhibitors and their anticancer activities in lung cancer cells.
Xinyu HE ; Jiarui FU ; Wenyu LYU ; Muyang HUANG ; Jianshan MO ; Yaxin CHENG ; Yulian XU ; Lijun ZHENG ; Xiaolei ZHANG ; Lu QI ; Lele ZHANG ; Ying ZHENG ; Mingqing HUANG ; Lin NI ; Jinjian LU
Chinese Journal of Natural Medicines (English Ed.) 2023;21(11):842-851
Cancer stands as one of the predominant causes of mortality globally, necessitating ongoing efforts to develop innovative therapeutics. Historically, natural products have been foundational in the quest for anticancer agents. Bulbocodin D (BD) and Bulbocodin C (BC), two bibenzyls derived from Pleione bulbocodioides (Franch.) Rolfe, have demonstrated notable in vitro anticancer activity. In human lung cancer A549 cells, the IC50s for BD and BC were 11.63 and 11.71 μmol·L-1, respectively. BD triggered apoptosis, as evidenced by an upsurge in Annexin V-positive cells and elevated protein expression of cleaved-PARP in cancer cells. Furthermore, BD and BC markedly inhibited the migratory and invasive potentials of A549 cells. The altered genes identified through RNA-sequencing analysis were integrated into the CMap dataset, suggesting BD's role as a potential signal transducer and activator of transcription 3 (STAT3) inhibitor. SwissDock and MOE analyses further revealed that both BD and BC exhibited a commendable binding affinity with STAT3. Additionally, a surface plasmon resonance assay confirmed the direct binding affinity between these compounds and STAT3. Notably, treatment with either BD or BC led to a significant reduction in p-STAT3 (Tyr 705) protein levels, regardless of interleukin-6 stimulation in A549 cells. In addition, the extracellular signal-regulated kinase (ERK) was activated after BD or BC treatment. An enhancement in cancer cell mortality was observed upon combined treatment of BD and U0126, the MEK1/2 inhibitor. In conclusion, BD and BC emerge as promising novel STAT3 inhibitors with potential implications in cancer therapy.
Humans
;
Lung Neoplasms/metabolism*
;
STAT3 Transcription Factor/metabolism*
;
Antineoplastic Agents/chemistry*
;
A549 Cells
;
Apoptosis
;
Cell Line, Tumor
;
Cell Proliferation
4.LASS2/TMSG1 overexpression inhibits proliferation and promotes apoptosis of human lung cancer A549 cells possibly by upregulating ceramide and p38 MAPK to activate a signaling cascade.
Zheng Lu LIU ; Cheng Rui XUAN ; Xi Ran HAN ; Ze Ze ZHENG ; Rui XIAO ; Lu Ri BAO ; Xiao Yan XU
Journal of Southern Medical University 2023;43(2):166-174
OBJECTIVE:
To investigate the effects of LASS2/TMSG1 gene overexpression on proliferation and apoptosis of human lung cancer A549 cells and explore the possible mechanism.
METHODS:
We examined LASS2/TMSG1 expression level in a previously constructed A549 cell line overexpressing LASS2/TMSG1 using Western blotting. The proliferation and apoptosis of the cells were detected using colony-forming assay, CCK-8 assay, Hoechst/PI double staining and flow cytometry. Fourteen nude mice were randomized into 2 groups (n=7) to receive subcutaneous injection of A549 cells with or without LASS2/TMSG1 overexpression on the back of the neck, and the cell proliferation in vivo was observed. The expression levels of p38 MAPK protein and p-p38 MAPK protein in the xenografts were detected with Western blotting. ELISA was used to detect the levels of ceramide and p38 MAPK protein in cultured A549 cell supernatants and the xenografts in nude mice.
RESULTS:
Compared with the negative control cells, A549 cells with LASS2/TMSG1 overexpression had significantly lowered proliferation ability in vitro with increased early apoptosis rate (P < 0.05), and showed obvious growth inhibition after inoculation in nude mice(P < 0.05). Western blotting showed that in both cultured A549 cells and the xenografts in nude mice, LASS2/TMSG1 gene overexpression significantly increased the expression levels of p38 MAPK protein and p-p38 MAPK protein (P < 0.05); the results of ELISA also revealed significantly increased levels of ceramide and p38 MAPK protein in the cell supernatant andxenografts as well (P < 0.05).
CONCLUSION
Overexpression of LASS2/TMSG1 gene can significantly inhibit the proliferation and promote early apoptosis of human lung cancer A549 cells both in vitro and in vivo possibly by upregulating the expressions of ceramide and p38 MAPK protein to activate a signal transduction cascade.
Animals
;
Humans
;
Mice
;
A549 Cells
;
Apoptosis
;
Cell Line, Tumor
;
Cell Proliferation
;
Lung Neoplasms
;
Membrane Proteins/metabolism*
;
Mice, Nude
;
p38 Mitogen-Activated Protein Kinases/metabolism*
;
Signal Transduction
;
Tumor Suppressor Proteins/metabolism*
5.Naphthalene allyl trifluoromethyl benzocyclopentanone inhibits proliferation and induces apoptosis of lung cancer A549 cells in vitro.
Xu WU ; Jun Jie HE ; Ping LI ; Yi WANG ; Liang YAN ; Jin Zhu MA
Journal of Southern Medical University 2022;42(2):201-206
OBJECTIVE:
To investigate the molecular mechanism by which a novel naphthalene allyl trifluoromethyl benzocyclopentanone XX0335 inhibits the proliferation and induces apoptosis of lung cancer A549 cells.
METHODS:
Lung cancer A549 cells were treated with 0.1% DMSO (control) or different concentrations (6.25, 12.5, and 25 μg/mL) of XX0335, and the changes in cell viability, cell cycle, proliferation and apoptosis were assessed with CCK-8 assay, EdU experiment, and flow cytometry. The effects of different concentrations of XX0335 on phosphorylation levels of proliferation-related proteins Akt, mTOR, Akt/mTOR and the expressions of cleaved PARP and cyclin D1 were determined using Western blotting. We also assessed the effect of XX0335 on tumor growth in a mouse model bearing A945 cell xenograft.
RESULTS:
Treatment with XX0335 reduced the viability of A549 cells in a dose-dependent manner (P < 0.01) and significantly inhibited cell proliferation (P < 0.001). Flow cytometry showed that XX0335 treatment promoted apoptosis of the cells (P < 0.01) and caused an obvious increase of the number of G1-phase cells. Compared with DMSO, XX0335 significantly inhibited the phosphorylation of Akt and mTOR, increased the expression of cleaved PARP, and lowered the protein expression of cyclin D1. In the tumor-bearing mouse models, injection of XX0335 significantly decreased the tumor volume (P < 0.01).
CONCLUSION
XX0335 inhibits the proliferation, cycle and induces apoptosis of lung cancer A549 cells possibly by inhibiting the Akt/mTOR signal pathway.
A549 Cells
;
Animals
;
Apoptosis
;
Cell Proliferation
;
Humans
;
Lung Neoplasms/metabolism*
;
Mice
;
Naphthalenes/pharmacology*
6.miR-20a-5p inhibits proliferation of lung cancer A549 cells by down-regulating HOXB13.
Hai Jun LIU ; Jie WANG ; Jia Xin SHEN ; Xu WU ; Yu Lei LI
Journal of Southern Medical University 2022;42(4):568-574
OBJECTIVE:
To investigate the molecular mechanism by which miR-20a-5p regulates HOXB13 gene expression and inhibits lung cancer cell proliferation.
METHODS:
The expression levels of HOXB13 mRNA and protein in lung cancer A549 cells transfected with HOXB13 overexpression plasmid or HOXB13 siRNA were detected with real-time fluorescence quantitative PCR (qRT-PCR) and Western blotting. CCK-8 and EdU assays were used to examine the effect of modulation of HOXB13 expression on cell proliferation. We screened possible binding miRNAs of HOXB13 by bioinformatics analysis. In A549 cells transfected with miR-20a-5p mimic or miR-20a-5p inhibitor, the expression level of miR-20a-5p was detected by qRT-PCR and the protein expression of HOXB13 was determined with Western blotting. CCK-8 and EdU assays were used to assess the effect of miR-20a-5p overexpression on the proliferation of A549 cells. miR-20a-5p mimic and HOXB13 overexpression plasmids were co-transfected into A549 cells, and the changes in cell proliferation were evaluated with CCK-8 and EdU assays.
RESULTS:
HOXB13 overexpression obviously promoted the proliferation of A549 cells (P < 0.05). miR-20a-5p was identified as the potential binding miRNA of HOXB13. Overexpression of miR-20a-5p in A549 cells significantly decreased the expression of HOXB13 protein (P < 0.05), while interference of miR-20a-5p obviously increased HOXB13 expression (P < 0.05). The results of cell proliferation experiment showed that miR-20a-5p and HOXB13 had opposite effects on cell proliferation, and the cells overexpressing both miR-20a-5p and HOXB13 showed a lower proliferation activity than the cells overexpressing HOXB13 but higher than the cells overexpressing miR-20a-5p alone (P < 0.05).
CONCLUSION
miR-20a-5p inhibits proliferation of lung cancer cells by down-regulating the expression of HOXB13.
A549 Cells
;
Apoptosis
;
Cell Line, Tumor
;
Cell Proliferation
;
Homeodomain Proteins/genetics*
;
Humans
;
Lung Neoplasms/genetics*
;
MicroRNAs/genetics*
;
Sincalide
7.Research on the mechanism of hypoxia promoting the migration of lung adenocarcinoma A549 cells.
Jia-Hao JIN ; Bao-Sheng ZHAO ; Yu-Zhen LIU
Chinese Journal of Applied Physiology 2022;38(1):68-74
Objective: To investigate the mechanism that hypoxia promotes the migration of lung adenocarcinoma A549 cells. Methods: A549 cells were cultured and cells that knockdown of acetyl-CoA carboxylase 1 (ACC1) were obtained by transfection with lentivirus, and cells that knockdown of sterol regulatory element-binding proteins-1 (SREBP-1) were obtained by treated with si-RNA. A549 cells were treated with hypoxia combined with hypoxia inducible factor-1α (HIF-1α) inhibitor PX-478 (25 μmol); Hypoxia combined with linoleic acid (LA) (20 μmol) treated A549 cells with ACC1 knockdown, and A549 cells with SREBP-1 knockdown were treated by hypoxia. Transwell migration assay was used to detect cell migration. Western blot was conducted to detect HIF-1α, ACC1 and epithelial mesenchymal transition (EMT) related proteins, Vimentin, E-Cadherin and SREBP-1; Real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) was performed to detect the changes of ACC1 and SREBP-1 mRNA in A549 cells after hypoxia and HIF-1α inhibitor PX-478 (25 μmol) treatment. Each experiment was repeated three times. Results: Compared with the normoxic control group, hypoxia promoted the migration of A549 cells (P<0.01), and up-regulated the expressions of ACC1, HIF-1α (all P<0.01) and SREBP-1 (P<0.05). PX-478 (25 μmol) inhibited the migration of A549 cells induced by hypoxia and down-regulated the expression of SREBP-1 (all P<0.05). ACC1 mRNA and SREBP-1 mRNA levels were increased after hypoxia treatment of A549 cells (all P<0.05). The levels of ACC1 mRNA and SREBP-1 mRNA were decreased after A549 cells treated with hypoxia combined with PX-478 (25 μmol) for 24 h (P<0.05, P<0.01). Knockdown of SREBP-1 in A549 cells was obtained by transfection with si-RNA. Transwell migration assay showed the number of cell migration in si-SREBP-1 group was less than that in normoxia control group (P<0.01). The si-SREBP-1 group and the si-NC group were treated with hypoxia. Compared with the control group, the number of cell migration in the si-SREBP-1 group was decreased (P<0.01), however, the difference was not statistically significant compared with the normoxia si-SREBP-1 group (P>0.05). Western blot showed that the expression of ACC1 in the si-SREBP-1 group was lower than that in the control group (P<0.01). Compared with the control group, the expression of ACC1 was decreased after si-SREBP-1 group treated with hypoxia (P<0.01). Knockdown of ACC1 inhibited the migration of A549 cells (P<0.05). After knockdown of ACC1, the migration number of A549 cells under normoxia and 5% O2 conditions had no significant difference (P>0.05). Application of LA under hypoxia condition rescued ACC1-knockdown induced inhibitory effect on hypoxia-promoted A549 cell migration (P<0.05). Conclusion: Hypoxia promotes migration of lung adenocarcinoma A549 cells by regulating fatty acid metabolism through HIF-1α/SREBP-1/ACC1 pathway.
A549 Cells
;
Acetyl-CoA Carboxylase
;
Adenocarcinoma of Lung
;
Cell Hypoxia/physiology*
;
Cell Line, Tumor
;
Humans
;
Hypoxia
;
Hypoxia-Inducible Factor 1, alpha Subunit
;
Lung Neoplasms
;
RNA/metabolism*
;
RNA, Messenger/metabolism*
;
Sterol Regulatory Element Binding Protein 1/metabolism*
8.The role and mechanism of autophagy in lipopolysaccharide-induced inflammatory response of A549 cells.
Jia SHI ; Hui-Xian TAO ; Yan GUO ; Yun-Su ZOU ; Mu-Zi WANG ; Zhi-Tao LU ; Yi-Fang DING ; Wei-Dong XU ; Xiao-Guang ZHOU
Chinese Journal of Contemporary Pediatrics 2022;24(10):1161-1170
OBJECTIVES:
To study the role and mechanism of autophagy in lipopolysaccharide (LPS)-induced inflammatory response of human alveolar epithelial A549 cells.
METHODS:
A549 cells were stimulated with LPS to establish a cell model of inflammatory response, and were then grouped (n=3 each) by concentration (0, 1, 5, and 10 μg/mL) and time (0, 4, 8, 12, and 24 hours). The A549 cells were treated with autophagy inhibitor 3-methyladenine (3-MA) to be divided into four groups (n=3 each): control, LPS, 3-MA, and 3-MA+LPS. The A549 cells were treated with autophagy agonist rapamycin (RAPA) to be divided into four groups (n=3 each): control, LPS, RAPA, and RAPA+LPS. The A549 cells were transfected with the Toll-like receptor 4 (TLR4) overexpression plasmid to be divided into four groups (n=3 each): TLR4 overexpression control, TLR4 overexpression, TLR4 overexpression control+LPS, and TLR4 overexpression+LPS. The A549 cells were transfected with TLR4 siRNA to be divided into four groups (n=3 each): TLR4 silencing control,TLR4 silencing, TLR4 silencing control+LPS, and TLR4 silencing+LPS. CCK-8 assay was used to measure cell viability. Western blot was used to measure the protein expression levels of inflammatory indicators (NLRP3, Caspase-1, and ASC), autophagic indicators (LC3B, Beclin-1, and P62), and TLR4.
RESULTS:
After stimulation with 1 μg/mL LPS for 12 hours, the levels of inflammatory indicators (NLRP3, Caspase-1, and ASC), autophagic indicators (LC3B, Beclin-1, and P62), and TLR4 increased and reached the peak (P<0.05). Compared with the LPS group, the 3-MA+LPS group had reduced expression of autophagy-related proteins and increased expression of inflammation-related proteins and TLR4, while the RAPA+LPS group had increased expression of autophagy-related proteins and reduced inflammation-related proteins and TLR4 (P<0.05). The TLR4 overexpression+LPS group had reduced autophagy-related proteins and increased inflammation-related proteins compared with the TLR4 overexpression control+LPS group, and the TLR4 silencing+LPS group had increased autophagy-related proteins and reduced inflammation-related proteins compared with the TLR4 silencing control+LPS group (P<0.05).
CONCLUSIONS
In the LPS-induced inflammatory response of human alveolar epithelial A549 cells, autophagic flux has a certain protective effect on A549 cells. TLR4-mediated autophagic flux negatively regulates the LPS-induced inflammatory response of A549 cells.
Humans
;
A549 Cells
;
Autophagy
;
Beclin-1/metabolism*
;
Caspase 1/metabolism*
;
Inflammation
;
Lipopolysaccharides/pharmacology*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Toll-Like Receptor 4/metabolism*
9.miR-367-3p Regulates Cells Proliferation and Invasion in NSCLC by Targeting ZEB2.
Chinese Journal of Lung Cancer 2022;25(11):782-788
BACKGROUND:
microRNAs play an important role in the development and biological phenotype of lung cancer. The present study was to investigate miR-367-3p level in non-small cell lung cancer (NSCLC) patients and its biological function of NSCLC cells.
METHODS:
Twenty-two patients with NSCLC (13 cases of adenocarcinoma and 9 cases of squamous carcinoma) admitted to our hospital and treated by surgery were included. During the operation, cancer tissue, paracancerous tissue and 5 mL peripheral blood were collected. Meanwhile, 22 healthy controls were selected and 5 mL peripheral blood was taken. Real-time PCR was applied to detected the expression of miR-367-3p in cancer tissues, peripheral blood of patients with NSCLC and healthy controls. miR-367-3p was detected in lung cancer cell lines (A549) and normal bronchial epithelial cells (BEAS-2B). The proliferation and invasion ability of A549 cells before and after infection were detected by MTT and Transwell assay after transfection with exogenous miR-367-3p. The downstream target gene of miR-367-3p was analyzed by bioinformatics. Zinc finger E-box binding homeobox 2 (ZEB2) was detected by Real-time PCR and Western blot.
RESULTS:
miR-367-3p in cancer tissues of 22 NSCLC patients was lower than corresponding normal tissues (P<0.05), and the serum miR-367-3p level in healthy subjects was higher than NSCLC subjects (P<0.05). The area under the receiver operating characteristic (ROC) curve of NSCLC was 0.95 (95%CI: 0.89-1.00) and 0.85 (95%CI: 0.74-0.97) respectively; The proliferation and migration ability of lung cancer cell line A549 transfected with exogenous miR-367-3p decreased significantly (P<0.05); Bioinformatics predicted that the downstream target of miR-367-3p was ZEB2 and up-regulating miR-367-3p expression, ZEB2 gene was decreased (P<0.05). The Cancer Genome Atlas (TCGA) data analysis showed that there was no significant difference in overall survival (OS) and disease free survival (DFS) between ZEB2 high expression group and low expression group (P>0.05). ZEB2 expression was positively correlated with infiltration of B lymphocytes (r=0.32, P<005), CD8⁺ T cells (r=0.44, P<005), CD4⁺ T cells (r=0.46, P<005), macrophages (r=0.65, P<005), neutrophils (r=0.73, P<005) and dendritic cells (r=0.71, P<005) in NSCLC.
CONCLUSIONS
The expression of miR-367-3p is down regulated in NSCLC patients and participates in the biological process of proliferation and invasion of NSCLC by targeting ZEB2 gene.
Humans
;
Carcinoma, Non-Small-Cell Lung/genetics*
;
Lung Neoplasms/genetics*
;
Cell Proliferation
;
MicroRNAs/genetics*
;
A549 Cells
;
Zinc Finger E-box Binding Homeobox 2/genetics*
10.Effects of arsenic and its main metabolites on A549 cell apoptosis and the expression of pro-apoptotic genes Bad and Bik.
Qian ZHOU ; Jin Yao YIN ; Jing Wen TAN ; Shu Ting LI ; Cheng Lan JIANG ; Yue Feng HE
Chinese Journal of Industrial Hygiene and Occupational Diseases 2022;40(9):661-667
Objective: To investigate the effect of arsenic and its main metabolites on the apoptosis of human lung adenocarcinoma cell line A549 and the expression of pro-apoptotic genes Bad and Bik. Methods: In October 2020, A549 cells were recovered and cultured, and the cell viability was detected by the cell counting reagent CCK-8 to determine the concentration and time of sodium arsenite exposure to A549. The study was divided into NaAsO(2) exposure groups and metobol: le expoure groups: the metabolite comparison groups were subdivided into the control group, the monomethylarsinic acid exposure group (60 μmol/L) , and the dimethylarsinic acid exposure group (60 μmol/L) ; sodium arsenite dose groups were subdivided into 4 groups: control group (0) , 20, 40, 60 μmol/L sodium arsenite NaAsO(2). Hoechst 33342/propidium iodide double staining (Ho/PI) was used to observe cell apoptosis and real-time quantitative polymerase chain reaction (qRT-PCR) was used to detect the expression levels of Bad and Bik mRNA in cells after exposure. Western blotting was used to detect the protein expressions of Bad, P-Bad-S112, Bik, cleaved Bik and downstream proteins poly ADP-ribose polymerase PARP1 and cytochrome C (Cyt-C) , using spectrophotometry to detect the activity changes of caspase 3, 6, 8, 9. Results: Compared with the control group, the proportion of apoptotic cells in the 20, 40, and 60 μmol/L NaAsO(2) dose groups increased significantly (P<0.01) , and the expression levels of Bad, Bik mRNA, the protein expression levels of Bad, P-Bad-S112, Bik, cleaved Bik, PARP1, Cyt-C were increased (all P<0.05) , and the activities of Caspase 3, 6, 8, and 9 were significantly increased with significantly differences (P<0.05) . Compared with the control group, the expression level of Bad mRNA in the DMA exposure group (1.439±0.173) was increased with a significant difference (P=0.024) , but there was no significant difference in the expression level of Bik mRNA (P=0.788) . There was no significant differences in the expression levels of Bad and Bik mRNA in the poison groups (P=0.085, 0.063) . Compared with the control group, the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to MMA were 0.696±0.023, 0.707±0.014, 0.907±0.031, 1.032±0.016, and there was no significant difference between the two groups (P=0.469, 0.669, 0.859, 0.771) ; the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to DMA were 0.698±0.030, 0.705±0.022, 0.908±0.015, 1.029±0.010, and there was no difference between the two groups (P=0.479, 0.636, 0.803, 0.984) . Conclusion: Sodium arsenite induces the overexpression of Bad and Bik proteins, initiates the negative feedback regulation of phosphorylated Bad and the degradation of Bik, activates the downstream proteins PARP1, Cyt-C and Caspase pathways, and mediates the apoptosis of A549 cells.
A549 Cells
;
Adenosine Diphosphate Ribose/pharmacology*
;
Apoptosis
;
Apoptosis Regulatory Proteins
;
Arsenic
;
Arsenites
;
Cacodylic Acid/pharmacology*
;
Caspase 3
;
Caspases/pharmacology*
;
Cytochromes c/pharmacology*
;
Humans
;
Mitochondrial Proteins/pharmacology*
;
Poisons
;
Propidium/pharmacology*
;
RNA, Messenger
;
Sincalide/pharmacology*
;
Sodium Compounds
;
bcl-Associated Death Protein/metabolism*

Result Analysis
Print
Save
E-mail