Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biol Res ; 54(1): 7, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33653412

RESUMO

BACKGROUND: Leaves of the natural plant lotus are used in traditional Chinese medicine and tea production. They are rich in flavonoids. METHODS: In this study, lotus leaf flavonoids (LLF) were applied to human lung cancer A549 cells and human small cell lung cancer cells H446 in vitro to verify the effect of LLF on apoptosis in these cells through the ROS/p38 MAPK pathway. RESULTS: LLF had no toxic effect on normal cells at concentrations up to 500 µg/mL, but could significantly inhibit the proliferation of A549 cells and H446 cells. Flow cytometry showed that LLF could induce growth in A549 cells. We also found that LLF could increase ROS and MDA levels, and decrease SOD activity in A549 cells. Furthermore, qRT-PCR and western blot analyses showed that LLF could upregulate the expression of p38 MAPK (p-p38 MAPK), caspase-3, caspase-9, cleaved caspase-3, cleaved caspase-9 and Bax and downregulate the expression of Cu/Zn SOD, CAT, Nrf2, NQO1, HO-1, and Bcl-2 in A549 cells. Results of HPLC showed that LLF mainly contain five active substances: kaempferitrin, hyperoside, astragalin, phloridzin, and quercetin. The apoptosis-inducing effect of LLF on A549 cells came from these naturally active compounds. CONCLUSIONS: We have shown in this study that LLF is a bioactive substance that can induce apoptosis in A549 cells in vitro, and merits further research and development.


Assuntos
Apoptose/efeitos dos fármacos , Flavonoides/farmacologia , Lotus/química , Neoplasias Pulmonares/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células A549 , Proliferação de Células , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Compostos Fitoquímicos/farmacologia , Folhas de Planta/química , Transdução de Sinais/efeitos dos fármacos
2.
Molecules ; 25(2)2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31952105

RESUMO

Casticin was obtained from natural plants, and it has been shown to exert biological functions; however, no report concerns the induction of DNA damage and repair in human lung cancer cells. The objective of this study was to investigate the effects and molecular mechanism of casticin on DNA damage and repair in human lung cancer A549 cells. Cell viability was determined by flow cytometric assay. The DNA damage was evaluated by 4',6-diamidino-2-phenylindole (DAPI) staining and electrophoresis which included comet assay and DNA gel electrophoresis. The protein levels associated with DNA damage and repair were analyzed by western blotting. The expression and translocation of p-H2A.X were observed by confocal laser microscopy. Casticin reduced total viable cell number and induced DNA condensation, fragmentation, and damage in A549 cells. Furthermore, casticin increased p-ATM at 6 h and increased p-ATR and BRCA1 at 6-24 h treatment but decreased p-ATM at 24-48 h, as well as decreased p-ATR and BRCA1 at 48 h. Furthermore, casticin decreased p-p53 at 6-24 h but increased at 48 h. Casticin increased p-H2A.X and MDC1 at 6-48 h treatment. In addition, casticin increased PARP (cleavage) at 6, 24, and 48 h treatment, DNA-PKcs and MGMT at 48 h in A549 cells. Casticin induced the expressions and nuclear translocation of p-H2AX in A549 cells by confocal laser microscopy. Casticin reduced cell number through DNA damage and condensation in human lung cancer A549 cells.


Assuntos
Apoptose , Dano ao DNA , Reparo do DNA , Flavonoides/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Proteínas de Neoplasias/metabolismo , Células A549 , Sobrevivência Celular , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo
3.
Molecules ; 24(11)2019 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-31181779

RESUMO

Lung cancer is one of the most common malignancies and is an increasing cause of cancer-related deaths. In our previous study, a series of ferulic acid (FA) derivatives were designed and synthesized; they exhibited positive anti-cancer activities, especially for a compound labelled FXS-3. In this study, a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed, wherein it revealed the inhibitory effect of FXS-3 on the proliferation and metastasis of human lung cancer A549 cells. The further flow cytometry assay showed that FXS-3 induced apoptosis of A549 cells induced cell cycle arrest at the G0/G1 phase. The trans-well migration and Matrigel invasion assays revealed that FXS-3 inhibited the migration and invasion of A549 cells. By the western blotting analysis, FXS-3 increased the expression of B-cell lymphoma-2 (Bcl-2) associated X protein (Bax)/Bcl-2 ratio, inhibited matrix metalloproteinase (MMP)-2 and MMP-9, and regulated the extracellular signal-regulated kinase (ERK)/p38, c-Jun N-terminal kinase (JNK), protein kinase B (AKT)/mechanistic target of rapamycin (mTOR), as well as mitogen-activated protein kinase (MEK)/ERK signaling pathways. The subsequent A549 xenograft-bearing mouse model and tail vein injection of A549 cells induced pulmonary tumor metastasis model showed that FXS-3 significantly restrained the tumor growth and metastasis. In conclusion, FXS-3 might inhibit proliferation and metastasis of human lung cancer A549 cells by positively regulating JNK signaling pathway and negativly regulating ERK/p38, AKT/mTOR, and MEK/ERK signaling pathways, which provides important scientific basis for the development of anti-cancer drugs about FA derivatives.


Assuntos
Ácidos Cumáricos/farmacologia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células A549 , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Cumáricos/química , Humanos , Masculino , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Pharm ; 15(11): 4974-4984, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30207732

RESUMO

We jointly analyzed the changes in cell cycle arrest and distribution, the accumulation of subphase cells, apoptosis, and proliferation in A549 cells treated with Saikosaponin D (Ssd) and JNK inhibitor SP600125 alone or in combination. Our results indicated that cell cycle arrest at G0/G1, S, and G2/M phases was coupled with the accumulation of subG1, subS, and subG2 cells, corresponding to early apoptosis, DNA endoreplication, and later inhibitory proliferation, respectively. Analyzing the expression of 18 cell cycle regulatory genes and JNK and phosphorylated JNK (pJNK) levels revealed an enhancement in these factors by Ssd. Additional SP600125 weakened or eliminated the Ssd-induced increase of these factors except that p53/p21 and Rassfia levels were further improved. Ingenuity Pathway Analysis (IPA) of the interactions of these factors revealed a negative synergistic effect on apoptosis while a positive synergistic effect on proliferative inhibition of the two drugs: (1) Ssd induced apoptosis via the activation of two axes, TGFα-JNK-p53 and TGFα-Rassfia-Mst1. By eliminating the Ssd-induced increase of JNK/pJNK, additional SP600125 weakened the Ssd-induced apoptotic axis of TGFα-JNK-p53 and simultaneously abolished Ssd-induced apoptosis; (2) Ssd inhibited proliferation by the activation of two axes, TGFß-p53/p21/p27/p15/p16 and TGFα-Rassfia-cyclin D1. By improving the Ssd-induced increase of p53/p21 and Rassfia, additional SP600125 enhanced the two axes of Ssd-induced inhibitory proliferation. Analyzing JNK/pJNK, p53, phospho-p53, and TNF-α levels revealed an opposite association of JNK/pJNK with p53 while consistent with phospho-p53 and TNF-α, which supported the proposals that JNK/pJNK negatively regulated p53 level, while it mediated p53 phosphorylation to transcriptionally activate TNF-α expression of apoptotic gene and trigger apoptosis. With the multiple roles, JNK/pJNK forms a synergetic and antagonistic feedback loop with phospho-p53/p53. Within the feedback loop, (1) Ssd-induced apoptosis depended on JNK/pJNK activities mediating phospho-p53 that activated TNF-α expression; (2) by weakening the negative regulation of JNK/pJNK in p53, SP600125 enhanced p53 level and the Ssd-induced inhibitory proliferation axes of TGFß-p53/p21/p27/p15/p16. The results indicated the central coordinating roles of the feedback loop in the synergistic and antagonistic effects of the two drugs in A549 cells and provided a rationale for the combination of Ssd with SP600125 in the treatment of lung cancer.


Assuntos
Antracenos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Retroalimentação Fisiológica , Neoplasias Pulmonares/tratamento farmacológico , Ácido Oleanólico/análogos & derivados , Saponinas/farmacologia , Células A549 , Antracenos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Pulmonares/patologia , Ácido Oleanólico/antagonistas & inibidores , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Fosforilação/efeitos dos fármacos , Saponinas/antagonistas & inibidores , Saponinas/uso terapêutico , Proteína Supressora de Tumor p53/metabolismo
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 49(3): 404-407, 2018 May.
Artigo em Chinês | MEDLINE | ID: mdl-30014643

RESUMO

OBJECTIVE: To investigate the relationship between expression of tumor suppressor gene p16 in non-small cell lung cancer (NSCLC) tissues and clinicopathological parameters,to further study on DNA methyltransferase inhibitors 5-nitrogen impurity-2'-deoxycytidine (5-Aza-CdR) in human lung cancer cell line A549 in regulating the expression of p16. METHODS: The expression of p16 protein in 76 cases of NSCLC tissues and normal tissue adjacent to carcinoma were detect by immunohistochemical SP method and the differences of p16 protein expression were analyzed. p16 gene promoter region of DNA methylation status were detect by MSP method in 5-Aza-CdR processing A549 cells,the expression of p16 in A549 lung cancer cell and effect of 5-Aza-CdR were detect by Western blot method. RESULTS: 32 cases (42.11%) of p16 protein expression was positive,significantly lower than that of the normal tissue adjacent to carcinoma (positive expression in 59 cases,77.63%) in 76 cases of NSCLC tissues; There were statistically significant differences (P<0.05) in the positive expression rates of p16 in NSCLC tissues with different pathological tissue grading,tumor differentiation degree,clinical TNM stage and lymph node metastasis. In A549 cells,p16 protein expression and non-methylated products were both in low expression states. After treated with 5-Aza-CdR,the expression of p16 protein and its non-methylated products were up-regulated,with the increase of 5-Aza-CdR concentration. CONCLUSION: The low expression of p16 in NSCLC tissues with squamous cell carcinomas,low differentiation,lymph node metastasis and phase Ⅲ-Ⅳ,which may prompt the deactivation and cause further progression of NSCLC,5-Aza-CdR could induce the expression of p16 protein and non-methylated products in A549 cells.


Assuntos
Azacitidina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Desoxicitidina/farmacologia , Neoplasias Pulmonares/patologia , Células A549 , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Regiões Promotoras Genéticas
6.
Drug Dev Ind Pharm ; 43(8): 1304-1313, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28402175

RESUMO

In this study, biodegradable chitosan hollow nanospheres (CHN) were fabricated using polystyrene nanospheres (PS) as templates. CHN were applied to increase the solubility of poorly water-soluble drugs. The lung cancer drug paclitaxel (PTX), which is used as a model drug, was loaded into CHN by the adsorption equilibrium method. The drug-loaded sample (PTX-CHN) offered sustained PTX release and good bioavailability. The state characterization of PTX by differential scanning calorimetry (DSC), X-ray diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR) showed that the PTX absorbed into CHN existed in an amorphous state. An in vitro toxicity experiment indicated that CHN were nontoxic as carriers of poorly water-soluble drugs. The PTX-CHN produced a marked inhibition of lung cancer A549 cells proliferation and encouraged apoptosis. A cell uptake experiment indicated that PTX-CHN was successfully taken up by lung cancer A549 cells. Furthermore, a degradation experiment revealed that CHN were readily biodegradable. These findings state clearly that CHN can be regarded as promising biomaterials for lung cancer treatment.


Assuntos
Células A549/efeitos dos fármacos , Antineoplásicos/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Pulmonares/tratamento farmacológico , Pulmão/metabolismo , Nanopartículas/química , Nanosferas/química , Paclitaxel/administração & dosagem , Células A549/química , Células A549/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Disponibilidade Biológica , Varredura Diferencial de Calorimetria , Humanos , Pulmão/química , Neoplasias Pulmonares/química , Paclitaxel/química , Paclitaxel/farmacologia , Difração de Raios X
7.
Environ Toxicol ; 29(7): 740-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-22848001

RESUMO

Chrysophanol (1,8-dihydroxy-3-methylanthraquinone) is one of the anthraquinone compounds, and it has been shown to induce cell death in different types of cancer cells. The effects of chrysophanol on human lung cancer cell death have not been well studied. The purpose of this study is to examine chrysophanol-induced cytotoxic effects and also to investigate such influences that involved apoptosis or necrosis in A549 human lung cancer cells in vitro. Our results indicated that chrysophanol decreased the viable A549 cells in a dose- and time-dependent manner. Chrysophanol also promoted the release of reactive oxygen species (ROS) and Ca(2+) and decreased the levels of mitochondria membrane potential (ΔΨm ) and adenosine triphosphate in A549 cells. Furthermore, chrysophanol triggered DNA damage by using Comet assay and DAPI staining. Importantly, chrysophanol only stimulated the cytocheome c release, but it did not activate other apoptosis-associated protein levels including caspase-3, caspase-8, Apaf-1, and AIF. In conclusion, human lung cancer A549 cells treated with chrysophanol exhibited a cellular pattern associated with necrotic cell death and not apoptosis in vitro. © 2012 Wiley Periodicals, Inc. Environ Toxicol 29: 740-749, 2014.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Neoplasias Pulmonares/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Trifosfato de Adenosina/metabolismo , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Humanos , Neoplasias Pulmonares/metabolismo , Necrose , Estresse Oxidativo/efeitos dos fármacos , Pontos de Checagem da Fase S do Ciclo Celular
8.
Front Vet Sci ; 10: 1208538, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37601754

RESUMO

Trichinella spiralis (T. spiralis) muscle-larva excretory/secretory products (ML-ESPs) is a complex array of proteins with antitumor activity. We previously demonstrated that ML-ESPs inhibit the proliferation of A549 non-small cell lung cancer (NSCLC) cell line. However, the mechanism of ML-ESPs against A549 cells, especially on the transcriptional level, remains unknow. In this study, we systematically investigated a global profile bioinformatics analysis of transcriptional response of A549 cells treated with ML-ESPs. And then, we further explored the transcriptional regulation of genes related to glucose metabolism in A549 cells by ML-ESPs. The results showed that ML-ESPs altered the expression of 2,860 genes (1,634 upregulated and 1,226 downregulated). GO and KEGG analysis demonstrated that differentially expressed genes (DEGs) were mainly associated with pathway in cancer and metabolic process. The downregulated genes interaction network of metabolic process is mainly associated with glucose metabolism. Furthermore, the expression of phosphofructokinase muscle (PFKM), phosphofructokinase liver (PFKL), enolase 2 (ENO2), lactate dehydrogenase B (LDHB), 6-phosphogluconolactonase (6PGL), ribulose-phosphate-3-epimerase (PRE), transketolase (TKT), transaldolase 1 (TALDO1), which genes mainly regulate glycolysis and pentose phosphate pathway (PPP), were suppressed by ML-ESPs. Interestingly, tricarboxylic acid cycle (TCA)-related genes, such as pyruvate dehydrogenase phosphatase 1 (PDP1), PDP2, aconitate hydratase 1 (ACO1) and oxoglutarate dehydrogenase (OGDH) were upregulated by ML-ESPs. In summary, the transcriptome profiling of A549 cells were significantly altered by ML-ESPs. And we also provide new insight into how ML-ESPs induced a transcriptional reprogramming of glucose metabolism-related genes in A549 cells.

9.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 37(5): 506-510, 2021 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-34816662

RESUMO

Objective: To investigate the synergistic effects of magnolol and gefitinib on non-small cell lung cancer A549 cells. Methods: A549 cells were treated with Magnolol (6.25~500 µmol/L) or gefitinib (6.25~500 µmol/L) for 24 h, respectively, and the cell viability was detected by cell counting Kit-8 (CCK-8) experiment (n=3). Magnolol 100 µmol/L and gefitinib 5 µmol/L were selected in the following experiments (n=3, 24 h). Control group, magnolol group, gefitinib group and magnolol+gefitinib group were set up for factorial analysis. Colony formation experiment was applied to detect the cell proliferation. Western blot was used to detect protein expressions. Flow cytometry was applied to test cell apoptosis and sorting CD44+ and CD133+ cells. Results: Compared with the control group, the colony formation rate of Magnolol or Gefitinib groups was decreased significantly (P<0.05); the apoptosis rate was increased significantly (P<0.05); the number of CD44+ and CD133+ cells was reduced significantly (P<0.05); the expressions of Ki67, PCNA, and stem cell marker proteins SOX2 and OCT4 were down-regulated (P<0.05); and the ratio of Bax/Bcl-2 was increased significantly (P<0.05). Compared with the Magnolol group or Gefitinib group, the Magnolol+Gefitinib group further promoted the above changes (P<0.05), and the apoptosis rate, the ratio of Bax/Bcl-2, SOX2 and OCT4 all showed interactions between magnolol and gefitinib (P<0.05). Conclusion: Magnolol and gefitinib promote the apoptosis of A549 cells and inhibit its stem cell-like properties, and the effect of the two combined is better than separated administration. Magnolol and gefitinib have interactive effects on A549 cells.


Assuntos
Compostos de Bifenilo/farmacologia , Carcinoma Pulmonar de Células não Pequenas , Gefitinibe , Lignanas/farmacologia , Neoplasias Pulmonares , Células A549 , Apoptose , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Gefitinibe/farmacologia , Humanos , Neoplasias Pulmonares/patologia
10.
Artigo em Chinês | WPRIM | ID: wpr-1030951

RESUMO

ObjectiveTo explore the interaction between bruceoside B and gut microbiota and the inhibitory activity of its metabolites on human lung cancer A549 cells, and to explore the value of bruceoside B in the treatment of non-small cell lung cancer(NSCLC). MethodBruceoside B was co-incubated with the human gut microbiota under anoxic conditions in vitro, and ultra high performance liquid chromatography-quadrupole-time-of-flight mass spectrometry(UPLC-Q-TOF-MS) was used to analyze the metabolic transformation products. Cell counting kit-8(CCK-8) assay was performed to determine the effects of bruceoside B and its metabolites on the proliferation of human lung cancer A549 cells and the half inhibitory concentration(IC50) was calculated. Five healthy male rats were gavaged with bruceoside B(2 mg·kg-1) for 7 days after adaptive feeding. The feces of rats were collected before and after administration. 16S rRNA sequencing was used to assess gut microbiota. ResultBruceoside B was mainly metabolized to brusatol by human gut microbiota, the IC50 of bruceoside B and the conversion product to A549 cells were 1 755.50, 19.57 μmol·L-1, respectively, and the conversion product had a better activity at inhibiting A549 cells proliferation than bruceoside B. Additionally, The results of intestinal flora analysis showed no significant differences in α diversity and β diversity of gut microbiota after administration. In terms of species abundance, at the phylum level, bruceoside B decreased the relative abundance of Actinobacteriota and Proteobacteria, increased the relative abundance of Firmicutes, Patescibacteria and Cyanobacteria. At the genus level, bruceoside B decreased the relative abundance of Staphylococcus, Aerococcus and Psychrobacter, increased the relative abundance of Romboutsia, Lactobacillus, Clostridium sensu stricto 1, Norank-f-norank-o-Clostridia-UCG-014, Turicibacter, Allobaculum and Candidatus Saccharimonas. The results of functional prediction showed that the gut microbiota functional compositions were relatively stable. ConclusionBruceoside B can be deglycosylated by intestinal flora and converted into brusatol, with a significant increase in antitumor activity. The administration of bruceoside B will not cause significant changes in the structure and function of the intestinal flora, resulting in intestinal microecological balance disorders, and the administration appears to be beneficial to the intestinal flora of NSCLC patients.

11.
Chinese Journal of Biologicals ; (12): 1368-1372, 2023.
Artigo em Chinês | WPRIM | ID: wpr-998392

RESUMO

@#Objective To isolate,extract and identify exosomes from the culture supernatant of non-small cell lung cancer A549 cells. Methods Exosomes from supernatant of non-small cell lung cancer A549 cells were isolated by ultracentrifugation(Ult)and modified method[Ult combined with Exo Quick~(TM)(Ult-Exo Quick~(TM),U-EQ)],which were determined for the concentration by BCA,measured for the particle size distribution by nanoparticle tracking analysis(NTA),observed for the morphology by fluorescence staining and transmission electron microscope(TEM),and detected for the expression of CD63 and HSP70 proteins on the surface of exosomes by Western blot. Results The concentrations of exosomes isolated by U-EQ and Ult were 1. 355 and 0. 909 mg/mL,respectively;The total number of particles was 4. 65 × 10~9 and 4. 06 × 10~9 particles/mL,the peak sizes ranged from 81. 5 to 165. 5 and 59. 5 to 176. 5 nm,and the average size was(152. 3 ± 8. 8)and(94. 3 ±9. 8)nm,respectively;The exosomes were all 30~150 nm in diameter,while the morphology of exosomes extracted by U-EQ method was more uniform;CD63 and HSP70 proteins were expressed in all of them,while the exosomes extracted by U-EQ method showed higher expression. Conclusion Both U-EQ and Ult methods can isolate exosomes from the supernatant of non-small cell lung cancer A549 cells,while the concentration of exosomes extracted by U-EQ method is higher.

12.
Artigo em Chinês | WPRIM | ID: wpr-907796

RESUMO

Objective:To investigate the effects of long non-coding RNA (lncRNA) CDKN2B-AS1 targeting miR-98-5p on proliferation and invasion of lung cancer A549 cells.Methods:A549 cells cultured in vitro were divided into control group, pcDNA group (transfected with pcDNA) , CDKN2B-AS1 group (transfected with pcDNA CDKN2B-AS1) and double transfection group (transfected with pcDNA CDKN2B-AS1 and pcDNA miR-98-5p) . The expression of lncRNA CDKN2B-AS1, miR-98-5p and the protein expression of PCNA, MMP-9 in A549 cells were detected. The activity, clone number, cloning efficiency, and the number of invasive cells of A549 cells were detected.Results:Compared with pcDNA group, the expression level of lncRNA CDKN2B-AS1 [ (2.14±0.14) vs (1.03±0.10) ], OD value in each time points, clone number [ (314.60±18.13) vs (220.08±12.46) ], cloning efficiency [ (85.81±3.06) % vs (60.03±2.85) %], invasive cell number [ (233.30±18.98) vs (140.84±12.30) ], expression levels of PCNA [ (0.78±0.08) vs (0.48±0.07) ] and MMP-9 [ (0.75±0.06) vs (0.38±0.06) ] proteins in A549 cells in CDKN2B-AS1 group were significantly increased ( P<0.05) ; the expression level of miR-98-5p [ (0.23±0.03) vs (0.99±0.09) ] was significantly decreased ( P<0.05) ; compared with CDKN2B-AS1 group, there was no significant difference in the expression level of lncRNA CDKN2B-AS1 in A549 cells in double transfection group ( P>0.05) , while the expression level of miR-98-5p in A549 cells was significantly increased ( P<0.05) . The OD value in each time points, clone number, cloning efficiency, invasive cell number, expression levels of PCNA and MMP-9 proteins were significantly decreased ( P<0.05) . Conclusion:LncRNA CDKN2B-AS1 can promote the proliferation and invasion of lung cancer A549 cells by targetingly inhibiting the expression of miR-98-5p.

13.
Biol. Res ; 54: 7-7, 2021. tab, graf, ilus
Artigo em Inglês | LILACS | ID: biblio-1505800

RESUMO

BACKGROUND: Leaves of the natural plant lotus are used in traditional Chinese medicine and tea production. They are rich in flavonoids. METHODS: In this study, lotus leaf flavonoids (LLF) were applied to human lung cancer A549 cells and human small cell lung cancer cells H446 in vitro to verify the effect of LLF on apoptosis in these cells through the ROS/p38 MAPK pathway. RESULTS: LLF had no toxic effect on normal cells at concentrations up to 500 µg/mL, but could significantly inhibit the proliferation of A549 cells and H446 cells. Flow cytometry showed that LLF could induce growth in A549 cells. We also found that LLF could increase ROS and MDA levels, and decrease SOD activity in A549 cells. Furthermore, qRT-PCR and western blot analyses showed that LLF could upregulate the expression of p38 MAPK (p-p38 MAPK), caspase-3, caspase-9, cleaved caspase-3, cleaved caspase-9 and Bax and downregulate the expression of Cu/Zn SOD, CAT, Nrf2, NQO1, HO-1, and Bcl-2 in A549 cells. Results of HPLC showed that LLF mainly contain five active substances: kaemp-feritrin, hyperoside, astragalin, phloridzin, and quercetin. The apoptosis-inducing effect of LLF on A549 cells came from these naturally active compounds. CONCLUSIONS: We have shown in this study that LLF is a bioactive substance that can induce apoptosis in A549 cells in vitro, and merits further research and development.


Assuntos
Humanos , Flavonoides/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Lotus/química , Neoplasias Pulmonares/patologia , Transdução de Sinais/efeitos dos fármacos , Folhas de Planta/química , Proliferação de Células , Compostos Fitoquímicos/farmacologia , Células A549 , Neoplasias Pulmonares/tratamento farmacológico
14.
Artigo em Chinês | WPRIM | ID: wpr-906421

RESUMO

Objective:To investigate the molecular mechanism of Qiyu Sanlong prescription (QYSL) in inhibiting the "addiction" of lung cancer A549 cells to miRNA21. Method:The human lung cancer A549 cells were routinely passaged and divided into the blank group, blank serum group, QYSL-containing serum group, and siRNA group. The prepared QYSL-containing serum was used for intervention, with the optimal concentration and action time determined in previous studies. The protein and mRNA expression levels of miRNA21 and related molecules in its target phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphatidylinositol 3-kinase (PI3K) signaling pathway were detected by real-time polymerase chain reaction (Real-time PCR) and Western blot assay. Result:The comparison with the blank serum group revealed that the mRNA expression levels of miRNA21 in the QYSL-containing serum group and the siRNA group were decreased, while the PTEN mRNA expression in the QYSL-containing serum group was increased, showing significant differences (<italic>P</italic><0.01). Compared with the blank serum, the QYSL-containing serum and siRNA significantly down-regulated PI3K and mammalian target of rapamycin (mTOR) mRNA expression (<italic>P</italic><0.01), whereas the QYSL-containing serum did not change the mRNA expression of protein kinase B (Akt). The protein expression levels of PTEN in the QYSL-containing serum group and the siRNA group were obviously elevated in contrast to that in the blank serum group (<italic>P</italic><0.05). Meanwhile, the protein expression levels of phosphorylated Akt (p-Akt) and phosphorylated mTOR (p-mTOR) evidently declined in the QYSL-containing serum group (<italic>P</italic><0.05), but there was no significant reduction in total Akt and mTOR protein expression. The PI3K protein expression was slightly down-regulated, with no statistical significance. Conclusion:QYSL inhibits the transcription of miRNA21, increases the expression of PTEN, and reduces the expression of key molecules in PI3K/Akt/mTOR signaling pathway, thus mildly inhibiting the "addiction" of lung cancer cells to oncogenes and blocking their proliferation.

15.
Artigo em Chinês | WPRIM | ID: wpr-855913

RESUMO

AIM: To investigate the effects of silencing carbonic anhydrase 1 (CA1) on proliferation, apoptosis, invasion and migration of human lung cancer A549 cells. METHODS: CA1-specific siRNA (si-CA1 group) and negative control (si-NC group) were transfected into lung cancer A549 cells by lipofection. The A549 cells transfected with empty liposome were used as blank control group. Real-time quantitative PCR (qPCR) and Western blot (Western blot) were used to detect the expression of CA1 mRNA and protein. Cell counting kit method (CCK-8), flow cytometry and Transwell assay were used to detect proliferation and apoptosis of A549 cells, invasion and migration capabilities. RESULTS:qPCR and Western blot showed that the expression levels of CA1 mRNA and protein in A549 cells transfected with CA1 siRNA were significantly down-regulated (P0.05). CONCLUSION: Silencing CA1 can inhibit the proliferation, invasion and migration of lung cancer A549 cells and promote cell apoptosis.

16.
Practical Oncology Journal ; (6): 228-232, 2019.
Artigo em Chinês | WPRIM | ID: wpr-752844

RESUMO

Objective The objective of this study was to investigate the role and molecular mechanism of AtractylenolideⅠin the progression of lung cancer. Methods qRT-qPCR and immunohistochemistry were used to detect the expression of TLR4 and MyD88 at levels of mRNA and protein in lung cancer and adjacent tissues. Transwell and MTT assays were used to detect effects of at-ractylenolide I(100 μM)on the invasion,migration and proliferation of A549 cells. Western blot was also used to detect the effect of atractylenolide I on the expression of TLR4 and MyD88 proteins. Results The expression of TLR4 and MyD88 at levels of mRNA and protein was highly expressed in lung cancer tissues when compared to adjacent tissues(P<0. 05). Compared with the control group,the invasion ability of A549 cells in the Atractylenolide I group was significantly decreased,and the proliferative activity was in-hibited(P<0. 05). Atractylenolide I inhibited the expression of TLR4 and MyD88 protein in A549 cells( P<0. 05). Conclusion

17.
Chinese Pharmacological Bulletin ; (12): 181-186, 2019.
Artigo em Chinês | WPRIM | ID: wpr-857279

RESUMO

Aim: To study the induction of apoptotic effect of sodium selenite on human lung cancer A549 cells and its mechanisms. Methods: A549 cells were exposed to different concentrations of sodium selenite for 24 h. MTT assay was applied to determine A549 cell proliferation. Inverted fluorescence microscope was used to investigate the morphological changes in A549 cells. Flow cytometry analysis was applied to assess the apoptotic rates of A549 cells. Laser confocal microscope was employed to measure the reactive oxygen species (ROS) fluorescence intensity. A multi-detection reader was used to determine the antioxidant parameter. Western blot was utilized to detect the expression of Keapl, Nrf2, HO-1 and Nrf2 in cytoplasm and nucleus. Results: MTT results showed that sodium selenite inhibited the proliferation of A549 cells in a concentration-dependent manner. After treatment with sodium selenite for 24 h, the apoptotic rate of A549 cells was markedly increased through Hoechst 33342 staining and flow cytometry measurement. Sodium selenite significantly up-regulated ROS and malondialdehyde (MDA) content and down-regulated the levels of superoxide dismutase (SOD) and glutathione (GSH). Meanwhile, sodium selenite treatment also reduced the expressions of Keapl, Nrf2 and HO-1 at protein levels and inhibited Nrf2 protein nuclear translocation in A549 cells. Conclusions: Treatment with sodium selenite induces A549 cells apoptosis, which may contribute to the anti-proliferation activity, induction of apoptosis and regulation of oxidative stress reaction and Keapl/Nrf2/ARE antioxidative signaling pathway expression.

18.
China Pharmacy ; (12): 906-909, 2016.
Artigo em Chinês | WPRIM | ID: wpr-504326

RESUMO

OBJECTIVE:To study the effects of Thymalfasin for injection on the apoptosis of human lung cancer A549 cells. METHODS:After treated with 0(blank control),25,50,100,200 and 400 mg/L Thymalfasin for injection for 24,48 and 72 h, the cell proliferation inhibitory rate was analyzed with MTT and calculated. After treated with 0(blank control),50 and 100 mg/L Thymalfasin for injection for 48 h,cell apoptosis was detected by flow cytometry,and the expression of Caspase-3,Bcl-2 and Bax and the phosphorylation level of Akt were deteced by Western blot. RESULTS:Compared with blank control group,proliferation in-hibitory rate of A549 cells increased after treated with Thymalfasin for injection,in concentration and time-dependent manner(P<0.05). The apoptotic rate of A549 cells increased after treated with Thymalfasin for injection 50,100 mg/L for 48 h (P<0.05). The expression of Caspase-3 increased while the Bcl-2/Bax and phosphorylation level of Akt decreased in A549 cells after treated with Thymalfasin for injection 100 mg/L (P<0.05). CONCLUSIONS:Thymalfasin for injection can inhibit the proliferation of A549 cells by activating Caspase-3,decreasing Bcl-2/Bax ratio,inhibiting Akt signal pathway and induce the apoptosis of A549 cells.

19.
China Pharmacy ; (12): 1346-1349, 2016.
Artigo em Chinês | WPRIM | ID: wpr-504405

RESUMO

OBJECTIVE:To study the inhibitory mechanism of timosaponin B-Ⅱ(TB-Ⅱ) on the proliferation and migration of human lung cancer A549 cells. METHODS:A549 cells were treated with TB-Ⅱ [0(blank control),1,10 and 100 μg/ml] for 48 h,and total RNA and total protein were extracted respectively. Real time fluorescence quantitative-PCR and Western blot were used to detect mRNA and protein levels of IL-18. IL-18 in A549 cells was silenced by transfection;the expression of IL-18 mRNA and protein were compared among untransfection group,negative control group and transfection group;and then human lung can-cer A549 cells with silenced gene were treated with 10 μg/ml TB-Ⅱ for 24,48 and 72 h. The activity of cell proliferation was de-tected with CCK-8,and the change of cell migration ability was observed by streak method. RESULTS:Compared with blank con-trol,the expression of IL-18 mRNA and protein in A549 cells all increased after treated with TB-Ⅱ(P<0.05 or P<0.01),and were positively correlated with concentration. Compared with untransfection group,the expression of IL-18 mRNA and protein de-creased in transfection group(P<0.01). Compared with untransfected cell treated with TB-Ⅱ,the viability and migration ability of A549 cells with transfection gene increased after treated with TB-Ⅱ for 72 h(P<0.01). CONCLUSIONS:TB-Ⅱ can inhibit the proliferation and migration of A549 cells by up-regulating IL-18 gene expression.

20.
China Pharmacy ; (12): 2690-2692,2693, 2016.
Artigo em Chinês | WPRIM | ID: wpr-605202

RESUMO

OBJECTIVE:To prepare folic acid(FA)-loaded vincristine(VCR)nano liposome(VCR-nLip-FA)and to study its effects on human liver and lung cancer cells. METHODS:VCR-nLip-FA was prepared by ammonium sulfate gradient method,and particle size,Zeta-potential,encapsulation rate and release rate were investigated. Taking human liver cancer HepG2 cells and lung cancer A549 cells as example,uptake rate and inhibitory effect in vitro (5-80 μg/ml) were compared between VCR-nLip-FA and VCR-nLip. RESULTS:The particle size distribution,average particle size,average Zeta-potential,average encapsulation rate and 24 h accumulative release rate of VCR-nLip-FA were 98.1-159.0 nm,132.2 nm,-40.1 mV,(86.6±3.5)%(n=4)and(42.2± 2.6)%. Compared with VCR-nLip,there was no statistical significance in uptake rate of A549 cells to VCR-nLip-FA and inhibitory effect of VCR-nLip-FA on A549 cell viability (P>0.05);uptake rate of HepG2 cells to VCR-nLip-FA and inhibitory effect of VCR-nLip-FA on HepG2 cell viability increased significantly (P<0.01),in dose-dependent manner. CONCLUSIONS:Prepared VCR-nLip-FA can target anti-tumor drug to HepG2 cells efficiently,and highly inhibit the growth of HepG2 cells. But it has no higher effects on A549 cells.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa