1.上海中医药大学(上海 201203)
徐嘉若,女,硕士,主要从事中药新药及药理学研究
姚广涛,副研究员,硕士生导师;E-mail:yaoguangtao1969@126.com
扫 描 看 全 文
徐嘉若, 陈佳靓, 姚广涛. 基于PI3K/AKT信号通路探讨双氢青蒿素对人三阴性乳腺癌MDA-MB-231细胞增殖、凋亡的影响[J]. 上海中医药大学学报, 2021,35(6):49-57.
XU Jiaruo, CHEN Jialiang, YAO Guangtao. Effects of dihydroartemisinin on proliferation and apoptosis of human triple-negative breast cancer MDA-MB-231 cells based on PI3K/AKT signaling pathway[J]. Academic Journal of Shanghai University of Traditional Chinese Medicine, 2021,35(6):49-57.
徐嘉若, 陈佳靓, 姚广涛. 基于PI3K/AKT信号通路探讨双氢青蒿素对人三阴性乳腺癌MDA-MB-231细胞增殖、凋亡的影响[J]. 上海中医药大学学报, 2021,35(6):49-57. DOI: 10.16306/j.1008-861x.2021.06.008.
XU Jiaruo, CHEN Jialiang, YAO Guangtao. Effects of dihydroartemisinin on proliferation and apoptosis of human triple-negative breast cancer MDA-MB-231 cells based on PI3K/AKT signaling pathway[J]. Academic Journal of Shanghai University of Traditional Chinese Medicine, 2021,35(6):49-57. DOI: 10.16306/j.1008-861x.2021.06.008.
目的:,2,基于PI3K/AKT信号通路探讨双氢青蒿素(DHA)对人三阴性乳腺癌MDA-MB-231细胞增殖及凋亡的影响。,方法:,2,①不同浓度(25、50、100 μmol/L)DHA干预不同时间(12、24、48、72 h)后,MTT法检测细胞增殖。②不同浓度(0、30、60、90 μmol/L)DHA干预48 h后,流式细胞仪检测细胞凋亡和细胞周期,Hoechst 33258染色观察细胞凋亡。③划痕实验观察60 μmol/L DHA干预0、6、12、24 h后对细胞迁移能力的影响。④60 μmol/L DHA干预12 h后,PCR检测细胞内凋亡相关因子,Caspase-3,、,Caspase-9,、,Bcl-2,、,Bax,的mRNA表达水平。60 μmol/L DHA干预48 h后,Western blot检测细胞内Caspase-3、Caspase-9、Bcl-2、Bax以及PI3K、p-PI3K、AKT、p-AKT的蛋白表达水平。,结果:,2,①DHA能够抑制MDA-MB-231细胞增殖,且抑制作用随药物浓度增加而增强。②DHA可以促进MDA-MB-231细胞凋亡,且随着药物浓度增加,促凋亡作用增强。③30 μmol/L DHA作用可使细胞周期阻滞在G0/G1期;60、90 μmol/L DHA作用后细胞周期阻滞在G2/M期。④60 μmol/L DHA干预12、24 h后,细胞迁移率显著降低(,P,<,0.01)。⑤60 μmol/L DHA干预后,,Caspase-3,、,Caspase-9, mRNA表达水平以及,Bax,/,Bcl-2, mRNA表达比值显著上调(,P,<,0.01),cleaved Caspase-9/Caspase-9、Bax/Bcl-2蛋白表达比值显著升高(,P,<,0.05);同时PI3K蛋白表达水平显著升高(,P,<,0.05),而p-PI3K、p-AKT的蛋白表达水平显著降低(,P,<,0.01)。,结论:,2,DHA可能通过抑制PI3K/AKT信号通路,抑制MDA-MB-231细胞的增殖和迁移,并诱导细胞凋亡。
Objective:,2,To investigate the effects of dihydroartemisinin(DHA) on the proliferation and apoptosis of human triple-negative breast cancer MDA-MB-231 cells based on PI3K/AKT signaling pathway.,Methods:,2,①After treatment with DHA at different concentrations(25, 50, 100 μmol/L) for different times(12, 24, 48, 72 h), the cell proliferation was detected by MTT assay.②After treatment with DHA at different concentrations(0, 30, 60, 90 μmol/L) for 48 hours, the cell apoptosis and cell cycle were detected by flow cytometry, and the cell apoptosis was observed by Hoechst 33258 staining.③The effect of 60 μmol/L DHA on the cell migration was observed by scratch test after treatment for 0, 6, 12 and 24 hours respectively.④After treatment with DHA at 60 μmol/L for 12 hours, the mRNA expression levels of apoptosis related factors ,Caspase-3,Caspase-9,Bcl-2, and ,Bax, were detected by PCR.After treatment with DHA at 60 μmol/L for 48 hours, the protein expression levels of Caspase-3, Caspase-9, Bcl-2, Bax, PI3K, p-PI3K, AKT and p-AKT were detected by Western blot.,Results:,2,①DHA could inhibit the proliferation of MDA-MB-231 cells, and the inhibitory effect was increased with the increase of drug concentration.②DHA could promote the apoptosis of MDA-MB-231 cells, and this effect was increased with the increase of drug concentration.③DHA at 30 μmol/L could block the cell cycle in G0/G1 phase, and DHA at 60 and 90 μmol/L could block the cell cycle in G2/M phase.④After treatment with DHA at 60 μmol/L for 12 and 24 hours, the cell migration rate was decreased significantly(,P,<,0.01) .⑤After treatment with DHA at 60 μmol/L, the expression levels of ,Caspase-3, and ,Caspase-9, mRNA and the expression ratio of ,Bax/Bcl-2, mRNA were significantly up-regulated(,P,<,0.01), and the expression ratios of cleaved Caspase-9/Caspase-9 and Bax/Bcl-2 protein were significantly increased(,P,<,0.05) ; meanwhile, the protein expression level of PI3K was increased significantly(,P,<,0.05), and the protein expression levels of p-PI3K and p-AKT were decreased significantly(,P,<,0.01) .,Conclusion:,2,DHA may inhibit the proliferation and migration of MDA-MB-231 cells and induce the cell apoptosis by inhibiting PI3K/AKT signaling pathway.
双氢青蒿素三阴性乳腺癌细胞增殖细胞凋亡PI3K/AKT信号通路MDA-MB-231细胞
dihydroartemisinintriple-negative breast cancercell proliferationcell apoptosisPI3K/AKT signaling pathwayMDA-MB-231 cell
SUNG H,FERLAY J,SIEGEL R L, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin,2021,71(3):209-249.
COSTA R L,HAN H S,GRADISHAR W J. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer:a review[J]. Breast Cancer Res Treat,2018,169(3):397-406.
SCHMADEKA R,HARMON B E,SINGH M. Triple-negative breast carcinoma:current and emerging concepts[J]. Am J Clin Pathol,2014,141(4):462-477.
BIANCHINI G,BALKO M J,MAYER A I, et al. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease[J]. Nat Rev Clin Oncol,2016,13(11):674-690.
PARK J H,AHN J H,KIM S B. How shall we treat early triple-negative breast cancer(TNBC):from the current standard to upcoming immuno-molecular strategies[J]. ESMO Open,2018,3(Suppl 1):e000357.
徐靖达,杨公利,杜井峰,等.双氢青蒿素通过调控PI3K/AKT通路增强肝癌H22细胞荷瘤小鼠放疗效果[J].中国病理生理杂志,2020,36(10):1782-1788.
LIANG R,CHEN W,CHEN X Y, et al. Dihydroartemisinin inhibits the tumorigenesis and invasion of gastric cancer by regulating STAT1/KDR/MMP9 and P53/BCL2L1/CASP3/7 pathways[J]. Pathol Res Pract,2021,218:153318.
ZHU L,CHEN X,ZHU Y, et al. Dihydroartemisinin Inhibits the Proliferation of Esophageal Squamous Cell Carcinoma Partially by Targeting AKT1 and p70S6K[J]. Front Pharmacol,2020,11:587470.
金红,杨岚,张黎,等.双氢青蒿素和吉非替尼联用对肺癌NCI-H1975细胞凋亡相关蛋白Bax与Bcl-2表达的影响[J].中国老年学杂志,2019,39(23):5806-5810.
李苒苒,陆文睿,陶敏,等.双氢青蒿素调控宫颈癌hela细胞的增殖迁移及其分子机制[J].皖南医学院学报,2020,39(4):315-318.
LIU Y,WANG W,XU J, et al. Dihydroartemisinin inhibits tumor growth of human osteosarcoma cells by suppressing Wnt/β-catenin signaling[J]. Oncol Rep,2013,30(4):1723-1730.
ZHU W H,LI Y W,ZHAO D H, et al. Dihydroartemisinin suppresses glycolysis of LNCaP cells by inhibiting PI3K/AKT pathway and downregulating HIF-1α expression[J]. Life Sci,2019,233:116730.
LI X G,BA Q,LIU Y L, et al. Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein[J]. Cell Discov,2017,3(1):17042.
李越,董炜,周晓艳,等.双氢青蒿素对三阴性乳腺癌MDA-MB-231细胞的抑制作用及其机制[J].西安交通大学学报(医学版),2021,42(5):791-796.
LI Y Q,WANG W,LI A X, et al. Dihydroartemisinin induces pyroptosis by promoting the AIM2/caspase-3/DFNA5 axis in breast cancer cells[J]. Chem Biol Interact,2021,340:109434.
HAN P,LUAN Y,LIU Y, et al. Small interfering RNA targeting Rac1 sensitizes colon cancer to dihydroartemisinin-induced cell cycle arrest and inhibited cell migration by suppressing NFκB activity[J]. Mol Cell Biochem,2013,379(1-2):171-180.
CHEN Y,MI Y,ZHANG X, et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells[J]. J Exp Clin Cancer Res,2019,38(1):402.
YUAN B,LIAO F,SHI Z Z, et al. Dihydroartemisinin Inhibits the Proliferation, Colony Formation and Induces Ferroptosis of Lung Cancer Cells by Inhibiting PRIM2/SLC7A11 Axis[J]. Onco Targets Ther,2020,13:10829-10840.
LIU Y,GAO S,ZHU J, et al. Dihydroartemisinin induces apoptosis and inhibits proliferation, migration, and invasion in epithelial ovarian cancer via inhibition of the hedgehog signaling pathway[J]. Cancer Med,2018,7(11):5704-5715.
MAGENTA D,SANGIOVANNI E,BASILICO N, et al. Inhibition of metalloproteinase-9 secretion and gene expression by artemisinin derivatives[J]. Acta Trop,2014,140:77-83.
LIN R Y,ZHANG Z H,CHEN L F, et al. Dihydroartemisinin(DHA)induces ferroptosis and causes cell cycle arrest in head and neck carcinoma cells[J]. Cancer Lett,2016,381(1):165-175.
QIN G Q,ZHAO C B,ZHANG L L, et al. Dihydroartemisinin induces apoptosis preferentially via a Bim-mediated intrinsic pathway in hepatocarcinoma cells[J]. Apoptosis,2015,20(8):1072-1086.
LIAO K,LI J,WANG Z L. Dihydroartemisinin inhibits cell proliferation via AKT/GSK3β/cyclinD1 pathway and induces apoptosis in A549 lung cancer cells[J]. Int J Clin Exp Pathol,2014,7(12):8684-8691.
CHEN S S,HU W,WANG Z, et al. p8 attenuates the apoptosis induced by dihydroartemisinin in cancer cells through promoting autophagy[J]. Cancer Biol Ther,2015,16(5):770-779.
LU M,SUN L H,ZHOU J, et al. Dihydroartemisinin-Induced Apoptosis is Associated with Inhibition of Sarco/Endoplasmic Reticulum Calcium ATPase Activity in Colorectal Cancer[J]. Cell Biochem Biophys,2015,73(1):137-145.
SHAO Y Y,ZHANG T L,WU L X, et al. AKT Axis, miR-21, and RECK Play Pivotal Roles in Dihydroartemisinin Killing Malignant Glioma Cells[J]. Int J Mol Sci,2017,18(2):350.
ZHANG H,ZHOU F,WANG Y Y, et al. Eliminating Radiation Resistance of Non-Small Cell Lung Cancer by Dihydroartemisinin Through Abrogating Immunity Escaping and Promoting Radiation Sensitivity by Inhibiting PD-L1 Expression[J]. Front Oncol,2020,10:595466.
ONTIKATZE T,RUDNER J,HANDRICK R, et al. Dihydroartemisinin is a Hypoxia-Active Anti-Cancer Drug in Colorectal Carcinoma Cells[J]. Front Oncol,2014,4:116.
WU C Y,ZHANG Y X,SUN Z R, et al. Molecular evolution of Cide family proteins: Novel domain formation in early vertebrates and the subsequent divergence[J]. BMC Evol Biol,2008,8(1):159.
SINGH N,HASSAN A,BOSE K. Molecular basis of death effector domain chain assembly and its role in caspase-8 activation[J]. FASEB J,2016,30(1):186-200.
YANG Y Y,YU Y B,WANG J H, et al. Silica nanoparticles induced intrinsic apoptosis in neuroblastoma SH-SY5Y cells via CytC/Apaf-1 pathway[J]. Environ Toxicol Pharmacol,2017,52:161-169.
LU W N,ZHENG F P,LAI D W, et al. Xuezhikang(血脂康)reduced renal cell apoptosis in streptozocin-induced diabetic rats through regulation of Bcl-2 family[J]. Chin J Integr Med,2016,22(8):611-618.
LARSEN B D,RAMPALLI S,BURNS L E, et al. Caspase 3/Caspase-Activated DNase Promote Cell Differentiation by Inducing DNA Strand Breaks[J]. Proc Natl Acad Sci U S A,2010,107(9):4230-4235.
SONG G,OUYANG G L,BAO S D. The activation of Akt/PKB signaling pathway and cell survival[J]. J Cell Mol Med,2005,9(1):59-71.
XIN M G,DENG X M. Nicotine Inactivation of the Proapoptotic Function of Bax through Phosphorylation[J]. J Biol Chem,2005,280(11):10781-10789.
0
浏览量
631
下载量
0
CSCD
3
CNKI被引量
关联资源
相关文章
相关作者
相关机构