您的位置:山东大学 -> 科技期刊社 -> 《山东大学学报(医学版)》

山东大学学报 (医学版) ›› 2019, Vol. 57 ›› Issue (9): 74-82.doi: 10.6040/j.issn.1671-7554.0.2018.1267

• • 上一篇    

黄芩素通过抑制PI3K/AKT/mTOR通路诱导膀胱癌细胞凋亡

王健1,2,李健1,王勇2,朱耀丰2   

  1. 1. 乐陵市人民医院泌尿外科, 山东 乐陵 253600;2. 山东大学齐鲁医院泌尿外科, 山东 济南 250012
  • 发布日期:2022-09-27
  • 通讯作者: 朱耀丰. E-mail: feng2209@163.com
  • 基金资助:
    山东省重点研发计划(2016GSF201036)

Baicalein induces apoptosis of bladder cancer cells via inhibition of the PI3K/AKT/mTOR pathway

WANG Jian1,2, LI Jian1, WANG Yong2, ZHU Yaofeng2   

  1. 1. Department of Urology, Peoples Hospital of Laoling City, Laoling 253600, Shandong, China;
    2. Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
  • Published:2022-09-27

摘要: 目的 研究黄芩素对膀胱癌细胞的作用及机制。 方法 膀胱癌细胞T24和5637经黄芩素(处理组)或二甲基亚砜(对照组)处理后,3-(4,5-二甲基噻唑-2)-2,5-二苯基四氮唑溴盐(MTT)以及胸腺嘧啶核苷类似物(EdU)插入实验检测细胞增殖和DNA复制速率;划痕实验和Transwell实验检测细胞的迁移和侵袭能力;流式细胞技术检测细胞周期和细胞凋亡水平;Western blotting检测PI3K/AKT/mTOR信号通路蛋白、B淋巴细胞瘤-2(Bcl-2)和B淋巴细胞瘤-2相关X蛋白(Bax)等蛋白的表达水平;裸鼠成瘤实验检测黄芩素对于体内膀胱癌细胞增殖能力的影响。 结果 黄芩素可以在体内和体外抑制膀胱癌细胞的增殖速率,高浓度黄芩素体外抑制率可达80%以上,体内抑制率约50%。黄芩素可以减缓DNA复制并降低肿瘤细胞的迁移和侵袭能力,诱导细胞停滞在G0/G1期,发生细胞凋亡,与对照组相比差异有统计学意义。黄芩素处理后,PI3K的表达以及AKT和mTOR的磷酸化水平降低,Bax/Bcl-2的比值升高。 结论 黄芩素通过抑制PI3K/AKT/mTOR信号通路抑制膀胱癌细胞增殖,发生周期阻滞,诱导细胞凋亡。

关键词: 黄芩素, 膀胱癌, B淋巴细胞瘤-2, PI3K/AKT/mTOR, 凋亡

Abstract: Objective To explore the effects of baicalein on bladder cancer cells and the underlying mechanism. Methods Bladder cancer cells, T24 and 5637, were treated with baicalein(the treatment group)or dimethyl sulfoxide(the control group). The rate of cell proliferation and DNA replication were examined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide(MTT)assays and 5-Ethynyl-2'-deoxyuridine(EdU)incorporation assays. The migration and invasion of cancer cells were explored by Wound healing assays and transwell assay. The cell cycle and cell apoptosis were detected by flow cytometry. The protein levels of PI3K/AKT/mTOR, Bcl-2, Bax and so on were explored by Western blotting. The effects of baicalein on cell proliferation in vivo were explored by tumor formation on nude mice. Results The inhibition rate of baicalein on cell proliferation was above 80% in vitro and about 50% in vivo. Baicalein could interfere DNA replication and suppress the migration and invasion of bladder cancer cells. Baicalein could also induce G0/G1 phase arrest and apoptosis of bladder cancer cells. Treatment of baicalein down-regulated the expression of PI3K and inhibit the phosphorylation of AKT and mTOR, with an increase in Bax/Bcl-2 ratio. Conclusion Baicalein can inhibit cell proliferation, induce cell cycle arrest and apoptosis of bladder cancer cells via suppression of the PI3K/AKT/mTOR pathway.

Key words: Baicalein, Bladder cancer, B-cell lymphoma 2, PI3K/AKT/mTOR, Apoptosis

中图分类号: 

  • R737.14
[1] Morgan TM, Keegan KA, Clark PE. Bladder cancer[J]. Curr Opin Oncol, 2011, 23(3): 275-282.
[2] Schenk-Braat EA, Bangma CH. Immunotherapy for superficial bladder cancer[J]. Cancer Immunol Immunother. 2005, 54(5): 414-423.
[3] Griffiths TR, Action on Bladder Cancer. Current perspectives in bladder cancer management[J]. Int J Clin Pract, 2013, 67(5): 435-448.
[4] Fridlender M, Kapulnik Y, Koltai H. Plant derived substances with anti-cancer activity: from folklore to practice[J]. Front Plant Sci, 2015, 6: 799. doi: 10.3389/fpls.2015.00799.
[5] Cragg GM, Newman DJ. Plants as a source of anti-cancer agents[J]. J Ethnopharmacol, 2005, 100(1-2): 72-79.
[6] Li C, Lin G, Zuo Z. Pharmacological effects and pharmacokinetics properties of Radix Scutellariae and its bioactive flavones[J]. Biopharm Drug Dispos, 2011, 32(8): 427-445.
[7] Chen SF, Hsu CW, Huang WH, et al. Post-injury baicalein improves histological and functional outcomes and reduces inflammatory cytokines after experimental traumatic brain injury[J]. Br J Pharmacol, 2008, 155(8): 1279-1296.
[8] Boyle SP, Doolan PJ, Andrews CE, et al. Evaluation of quality control strategies in Scutellaria herbal medicines[J]. J Pharm Biomed Anal, 2011, 54(5): 951-957.
[9] He XL, Wang YH, Gao M, et al. Baicalein protects rat brain mitochondria against chronic cerebral hypoperfusion-induced oxidative damage[J]. Brain Res, 2009, 1249: 212-221. doi: 10.1016/j.brainres.2008.10.005.
[10] Wang L, Ling Y, Chen Y, et al. Flavonoid baicalein suppresses adhesion, migration and invasion of MDA-MB-231 human breast cancer cells[J]. Cancer Lett, 2010, 297(1): 42-48.
[11] Taniguchi H, Yoshida T, Horinaka M, et al. Baicalein overcomes tumor necrosis factor-related apoptosis-inducing ligand resistance via two different cell-specific pathways in cancer cells but not in normal cells[J]. Cancer Res, 2008, 68(21): 8918-8927.
[12] Chen CH, Huang LL, Huang CC, et al. Baicalein, a novel apoptotic agent for hepatoma cell lines: a potential medicine for hepatoma[J]. Nutr Cancer, 2000, 38(2): 287-295.
[13] Liu H, Dong Y, Gao Y, et al. The fascinating effects of baicalein on cancer: a review[J]. Int J Mol Sci, 2016, 17(10). pii: E1681.
[14] Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015[J]. CA Cancer J Clin, 2016, 66(2): 115-132.
[15] Shen Z, Shen T, Wientjes MG, et al. Intravesical treatments of bladder cancer: review[J]. Pharm Res, 2008, 25(7): 1500-1510.
[16] Yoshida T, Okuyama H, Nakayama M, et al. High-dose chemotherapeutics of intravesical chemotherapy rapidly induce mitochondrial dysfunction in bladder cancer-derived spheroids[J]. Cancer Sci, 2015, 106(1): 69-77.
[17] Bie B, Sun J, Guo Y, et al. Baicalein: a review of its anti-cancer effects and mechanisms in Hepatocellular Carcinoma[J]. Biomed Pharmacother, 2017, 93: 1285-1291. doi: 10.1016/j.biopha.2017.07.068.
[18] Yu X, Yang Y, Li Y, et al. Baicalein inhibits cervical cancer progression via downregulating long noncoding RNA BDLNR and its downstream PI3K/Akt pathway[J]. Int J Biochem Cell Biol, 2018, 94: 107-118. doi: 10.1016/j.biocel.2017.11.009.
[19] Ma X, Yan W, Dai Z, et al. Baicalein suppresses metastasis of breast cancer cells by inhibiting EMT via downregulation of SATB1 and Wnt/beta-catenin pathway[J]. Drug Des Devel Ther, 2016, 10: 1419-1441. doi: 10.2147/DDDT.S102541.
[20] Liu P, Feng J, Sun M, et al. Synergistic effects of baicalein with gemcitabine or docetaxel on the proliferation, migration and apoptosis of pancreatic cancer cells[J]. Int J Oncol, 2017, 51(6): 1878-1886.
[21] Malaguarnera L. Implications of apoptosis regulators in tumorigenesis[J]. Cancer Metastasis Rev, 2004, 23(3-4): 367-387.
[22] Goswami A, Ranganathan P, Rangnekar VM. The phosphoinositide 3-kinase/Akt1/Par-4 axis: a cancer-selective therapeutic target[J]. Cancer Res, 2006, 66(6): 2889-2892.
[23] Chen M, Gu J, Delclos GL, et al. Genetic variations of the PI3K-AKT-mTOR pathway and clinical outcome in muscle invasive and metastatic bladder cancer patients[J]. Carcinogenesis, 2010, 31(8): 1387-1391.
[24] Jiang BH, Liu LZ. PI3K/PTEN signaling in angiogenesis and tumorigenesis[J]. Adv Cancer Res, 2009, 102: 19-65. doi: 10.1016/S0065-230X(09)02002-8.
[25] Hartmann W, Kuchler J, Koch A, et al. Activation of phosphatidylinositol-3'-kinase/AKT signaling is essential in hepatoblastoma survival[J]. Clin Cancer Res, 2009, 15(14): 4538-4545.
[26] Fahmy M, Mansure JJ, Brimo F, et al. Relevance of the mammalian target of rapamycin pathway in the prognosis of patients with high-risk non-muscle invasive bladder cancer[J]. Hum Pathol, 2013, 44(9): 1766-1772.
[27] Chaux A, Comperat E, Varinot J, et al. High levels of phosphatase and tensin homolog expression are associated with tumor progression, tumor recurrence, and systemic metastases in pT1 urothelial carcinoma of the bladder: a tissue microarray study of 156 patients treated by transurethral resection[J]. Urology, 2013, 81(1): 116-122.
[28] Gonzalez-Roibon ND, Chaux A, Al-Hussain T, et al. Dysregulation of mammalian target of rapamycin pathway in plasmacytoid variant of urothelial carcinoma of the urinary bladder[J]. Hum Pathol, 2013, 44(4): 612-622.
[29] Schultz L, Chaux A, Albadine R, et al. Immunoexpression status and prognostic value of mTOR and hypoxia-induced pathway members in primary and metastatic clear cell renal cell carcinomas[J]. Am J Surg Pathol, 2011, 35(10): 1549-1556.
[30] Danial NN. BCL-2 family proteins: critical checkpoints of apoptotic cell death[J]. Clin Cancer Res, 2007, 13(24): 7254-7263.
[31] Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked[J]. Science, 1997, 275(5303): 1129-1132
[32] Skommer J, Brittain T, Raychaudhuri S. Bcl-2 inhibits apoptosis by increasing the time-to-death and intrinsic cell-to-cell variations in the mitochondrial pathway of cell death[J]. Apoptosis, 2010, 15(10): 1223-1233.
[33] Zhao Y, Zhang CL, Zeng BF, et al. Enhanced chemosensitivity of drug-resistant osteosarcoma cells by lentivirus-mediated Bcl-2 silencing[J]. Biochem Biophys Res Commun, 2009, 390(3): 642-627.
[34] Reagan-Shaw S, Nihal M, Ahsan H, et al. Combination of vitamin E and selenium causes an induction of apoptosis of human prostate cancer cells by enhancing Bax/Bcl-2 ratio[J]. Prostate, 2008, 68(15): 1624-1634.
[35] Katiyar SK, Roy AM, Baliga MS. Silymarin induces apoptosis primarily through a p53-dependent pathway involving Bcl-2/Bax, cytochrome c release, and caspase activation[J]. Mol Cancer Ther, 2005, 4(2): 207-216.
[1] 张士宝 刘庆勇 阮喜云 陈杰 张建军 李宗武 杨广笑 王全颖. NT4-SAC-HA2-TAT融合基因表达载体的构建及鉴定[J]. 山东大学学报(医学版), 2209, 47(6): 15-19.
[2] 鹿向东 杨伟 徐广明 曲元明. 脑膜瘤中PPAR-γ的表达及曲格列酮对脑膜瘤培养细胞生长的影响[J]. 山东大学学报(医学版), 2209, 47(6): 65-.
[3] 赵舸,邹存华,宋冬冬,赵淑萍. 丹参酮IIA对子宫内膜癌细胞增殖与凋亡的影响[J]. 山东大学学报 (医学版), 2022, 60(9): 53-58.
[4] 刘敏,张玉超,马小莉,刘昕宇,孙露,左丹,刘元涛. 孤核受体NR4A1在H2O2诱导小鼠肾脏足细胞损伤中的作用[J]. 山东大学学报 (医学版), 2022, 60(5): 16-21.
[5] 李卉,姜朝阳,刘岩,张曼. 组蛋白去乙酰化酶SIRT1调控氧化低密度脂蛋白诱导巨噬细胞凋亡的表达[J]. 山东大学学报 (医学版), 2022, 60(1): 6-12.
[6] 张倩,秦明明,何学佳,蔡秋景,张亚民,李庆苏,朱薇薇. 骨化三醇对哮喘中TGF-β1所诱导上皮间充质转化的调控作用[J]. 山东大学学报 (医学版), 2021, 59(7): 10-18.
[7] 米琦,史爽,李娟,李培龙,杜鲁涛,王传新. 膀胱癌circRNAs介导的ceRNA网络及预后评估模型的构建[J]. 山东大学学报 (医学版), 2021, 59(6): 94-102.
[8] 卢游,且迪,伍晋辉,杨凡. 干预Sonic Hedgehog信号通路对宫内发育迟缓新生大鼠学习记忆能力的影响[J]. 山东大学学报 (医学版), 2021, 59(5): 82-89.
[9] 南莉,杨凯转,张一帆. 室内照明白色发光二极管对大鼠视网膜的影响[J]. 山东大学学报 (医学版), 2021, 59(4): 56-62.
[10] 刘淑丹,张飞燕,郭松林,梁雪云,陈冬梅. 氧化苦参碱改善缺氧缺血引起的HaCaT细胞氧化应激损伤[J]. 山东大学学报 (医学版), 2021, 59(3): 26-34.
[11] 薛源,林雪艳,徐歌,田永杰. 低氧诱导因子-1α在子宫内膜异位症患者血清中的表达和对在位子宫内膜间质细胞上皮-间质转化的影响[J]. 山东大学学报 (医学版), 2021, 59(2): 41-47.
[12] 张晓璐,王丽莉,陈凯明,娄宪芝,张曼. 组蛋白去乙酰化酶SIRT1经Toll样受体4途径对巨噬细胞凋亡的调控[J]. 山东大学学报 (医学版), 2020, 58(12): 8-14.
[13] 史丽,马静,赵喜娃,关英霞,赵连梅,单保恩. miR-25-3p在40例子宫内膜腺癌组织中的表达及对KLE细胞生物学功能的影响[J]. 山东大学学报 (医学版), 2020, 58(12): 86-91.
[14] 罗梦颖,吴蓓,王叶,骆衡,曹煜. 芳姜黄酮对SiHa细胞增殖、迁移及侵袭的影响[J]. 山东大学学报 (医学版), 2019, 57(6): 68-74.
[15] 万秀霞,孙秀萍,佟晶洁. 大黄酚对结肠癌的抗肿瘤作用[J]. 山东大学学报 (医学版), 2019, 57(5): 74-79.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!