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 (12): 46-51.doi: 10.6040/j.issn.1671-7554.0.2019.886

• • 上一篇    

乌苯美司对卵巢癌A2780细胞的生物学影响

关红卫,李娟,孙锐,刘婕,李长忠   

  1. 山东大学附属省立医院妇科, 山东 济南 250021
  • 发布日期:2022-09-27
  • 通讯作者: 李长忠. E-mail:15168888909@163.com
  • 基金资助:
    国家自然科学基金(81671434,81503298)

Biological effects of ubenimex on ovarian cancer A2780 cell

GUAN Hongwei, LI Juan, SUN Rui, LIU Jie, LI Changzhong   

  1. Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China
  • Published:2022-09-27

摘要: 目的 研究乌苯美司对卵巢癌A2780细胞增殖、细胞周期、凋亡和自噬以及迁移和侵袭能力的影响,探讨可能的作用机制。 方法 CCK-8实验检测乌苯美司对A2780细胞增殖的影响;Transwell实验检测乌苯美司对A2780细胞迁移及侵袭能力的影响;流式细胞术检测乌苯美司对A2780细胞凋亡和细胞周期的影响;Western blotting检测APN、AKT、p-AKT、LC3B、p62表达水平的变化。 结果 (1)乌苯美司能够抑制A2780细胞的增殖(F浓度=812.56,P<0.001;F时间=8.58,P=0.010;F交互=4.00,P=0.027)、迁移(t=56.9,P<0.001)和侵袭(t=11.8,P<0.001)、导致G2/M期细胞比例增高(t=7.9,P=0.016),G1期细胞比例减少(t=14.8,P=0.005),发生G2/M细胞周期阻滞。(2)乌苯美司能够诱导A2780细胞凋亡(P=0.002),且联合应用AKT抑制剂后诱导细胞凋亡的作用增强(P=0.007)。(3)乌苯美司可以导致APN表达降低,p-AKT、LC3-B和p62表达增加。 结论 乌苯美司可能是卵巢癌的一种治疗方法或者辅助治疗方法,与AKT抑制剂联用能够增强其治疗效果。

关键词: 卵巢癌, 乌苯美司, 增殖, 侵袭, 凋亡, 自噬

Abstract: Objective To investigate the effect of ubenimex on cell proliferation, cell cycle, apoptosis, autophagy, migration and invasion in ovarian cancer A2780 cell and its possible mechanism. Methods CCK-8 assay was used to analyze the effect of ubenimex on proliferation of A2780 cell. Transwell assay was performed to detect the effect of ubenimex on migration and invasion of A2780 cell. Flow cytometry was used to analyze the cell apoptosis and cell cycle. Western blotting was performed to investigate the protein expression levels of APN, AKT, p-AKT, LC3-B, and p62. Results (1)Ubenimex inhibited A2780 cell proliferation(Fconcentration=812.56,P<0.001;Ftime=8.58,P=0.010;Fconcentration×time=4.00,P=0.027), migration(t=56.9, P<0.001)and invasion(t=11.8, P<0.001), increased the cell proportion in G2/M phase (t=7.9, P=0.016)and decreased the cell proportion in G1 phase (t=14.8, P=0.005)which led to G2/M cell cycle arrest. (2)Ubenimex induced A2780 cell apoptosis(P=0.002), and cell apoptosis was increased when ubenimex was combined with AKT inhibitor(P=0.007). (3)Ubenimex reduced the APN expression, but increased p-AKT, LC3B and p62 expressions. Conclusion Ubenimex may be a treatment or adjuvant therapy for ovarian cancer, and its therapeutic effect can be enhanced in combination with AKT inhibitor.

Key words: Ovarian cancer, Ubenimex, Proliferation, Invasion, Apoptosis, Autophagy

中图分类号: 

  • R737.31
[1] Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2018, 68(6): 394-424.
[2] Jin H, Yu Y, Zhang T, et al. Snail is critical for tumor growth and metastasis of ovarian carcinoma[J]. Int J Cancer, 2010, 126(9): 2102-2111.
[3] Cannistra SA. Cancer of the ovary[J]. New Engl J Med, 2004, 351(24): 2519-2529.
[4] Ffrench B, Gasch C, O’Leary JJ, et al. Developing ovarian cancer stem cell models: laying the pipeline from discovery to clinical intervention[J]. Mol Cancer, 2014, 13: 262. doi: 10.1186/1476-4598-13-262.
[5] Liu S, Xie F, Wang H, et al. Ubenimex inhibits cell proliferation, migration and invasion in renal cell carcinoma: the effect is autophagy-associated[J]. Oncol Rep, 2015, 33(3): 1372-1380.
[6] Hitzerd SM, verbrugge SE, Ossenkoppele G, et al. Positioning of aminopeptidase inhibitors in next generation cancer therapy[J]. Amino Acids, 2014, 46(4): 793-808.
[7] Wickstr(¨overo)m M, Larsson R, Nygren P, et al. Aminopeptidase N(CD13)as a target for cancer chemotherapy[J]. Cancer Sci, 2011, 102(3): 501-508.
[8] Wakita A, Ohtake S, Takada S, et al. Randomized comparison of fixed-schedule versus response-oriented individualized induction therapy and use of ubenimex during and after consolidation therapy for elderly patients with acute myeloid leukemia: the JALSG GML200 Study[J]. Int J Hematol, 2012, 96(1): 84-93.
[9] Ichinose Y, Genka K, Koike T, et al. NK421 Lung Cancer Surgery Group: randomized double-blind placebo-controlled trial of bestatin in patients with resected stage I squamous-cell lung carcinoma[J]. J Natl Cancer Inst, 2003, 95(8): 605-610.
[10] Xu JW, Li CG, Huang XE, et al. Ubenimex capsule improves general performance and chemotherapy related toxicity in advanced gastric cancer cases[J]. Asian Pac J Cancer Prev, 2011, 12(4): 985-987.
[11] Tsukamoto H, Shibata K, Kajiyama H, et al. Aminopeptidase N(APN)/CD13 inhibitor, Ubenimex, enhances radiation sensitivity in human cervical cancer[J]. BMC Cancer, 2008, 8:74. doi: 10.1186/1471-2407-8-74.
[12] Tang Z, Li C, Kang B, et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses[J]. Nucleic Acids Res, 2017, 45(W1): W98-W102.
[13] Terauchi M, Kajiyama H, Shibata K, et al. Inhibition of APN/CD13 leads to suppressed progressive potential in ovarian carcinoma cells[J]. BMC Cancer, 2007, 7: 140.
[14] 路金枝,陈爱平.顺铂联合乌苯美司对卵巢癌SKOV3细胞体外生长的作用[J].山东医药, 2012, 52(25): 50-52.
[15] Wang X, Liu Y, Wu R, et al. Role of ubenimex as an anticancer drug and its synergistic effect with akt inhibitor in human A375 and A2058 cells[J]. Onco Targets Ther, 2018, 11: 943-953. doi: 10.2147/OTT.S157480.
[16] Aozuka Y, Koizumi K, Saitoh Y, et al. Anti-tumor angiogenesis effect of aminopeptidase inhibitor bestatin against B16-BL6 melanoma cells orthotopically implanted into syngeneic mice[J]. Cancer Lett, 2004, 216(1): 35-42.
[17] Ishii K, Usui S, Sugimura Y, et al. Inhibition of aminopeptidase N(AP-N)and urokinase-type plasminogen activator(uPA)by zinc suppresses the invasion activity in human urological cancer cells[J]. Biol Pharm Bull, 2001, 24(3): 226-230.
[18] Malaguarnera L. Implication of apoptosis regulation in tumorigenesis[J]. Cancer Metastasis Rev, 2004, 23(3-4): 367-387.
[19] Han L, Zhang Y, Liu S, et al. Autophagy flux inhibition, G2/M cell cycle arrest and apoptosis induction by ubenimex in glioma cell lines[J]. Oncotarget, 2017, 8(64): 107730-107743.
[20] Klionsky DJ. Autophagy from phenomenology to molecular understanding in less than a decade[J]. Nat Rev Mol Cell Biol, 2007, 8(11): 931-937.
[21] Eum KH, Lee M. Crosstalk between autophagy and apoptosis in the regulation of paclitaxel-induced cell death in v-Ha-ras-transformed fibroblasts[J]. Mol Cell Biochem, 2011, 348(1-2): 61-68.
[22] Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research[J]. Cell, 2010, 140(3): 313-326.
[23] Han L, Zhao Q, Liang X, et al. Ubenimex enhances Brd4 inhibition by suppressing HEXIM1 autophagic degradation and suppressing the Akt pathway in glioma cells[J]. Oncotarget, 2017, 8(28): 45643-45655.
[24] Krajarng A, Chulasiri M, Watanapokasin R. Etlingera elatior Extract promotes cell death in B16 melanoma cells via down-regulation of ERK and Akt signaling pathways[J]. BMC Complement Altern Med, 2017, 17(1):415. doi: 10.1186/s12906-017-1921-y.
[25] Díaz M, González R, Plano D, et al. A diphenyldiselenide derivative induces autophagy via JNK in HTB-54 lung cancer cells[J]. J Cell Mol Med, 2018, 22(1): 289-301.
[26] Tang R, Xu X, Yang W, et al. MED27 promotes melanoma growth by targeting AKT/MAPK and NF-κB/iNOS signaling pathways[J]. Cancer Lett, 2016, 373(1): 77-87.
[27] Wu J, Hu G, Dong Y, et al. Matrine induces Akt/mTOR signalling inhibition-mediated autophagy and apoptosis in acute myeloid leukaemia cells[J]. J Cell Mol Med, 2017, 21(6): 1171-1181.
[28] Wang C, Zhang X, Teng Z, et al. Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice[J]. Eur J Pharmacol, 2014, 740: 312-320. doi: 10.1016/j.ejphar.2014.06.051.
[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] 王晓磊 张海涛 张辉 郭成浩. 舒血宁注射液对高碘致培养血管内皮细胞损伤的保护作用[J]. 山东大学学报(医学版), 2209, 47(6): 38-.
[3] 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-.
[4] 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.
[5] 张振伟,李佳,陈克明. IGF2BP2/m6A/ITGA5信号轴调控肾透明细胞增殖和迁移[J]. 山东大学学报 (医学版), 2022, 60(9): 74-84.
[6] 张艺馨,赵玉立,封丽. 超声特征及术前CA-125联合对51例卵巢交界性及Ⅰ期恶性肿瘤的鉴别诊断[J]. 山东大学学报 (医学版), 2022, 60(7): 104-109.
[7] 孙丽娜,杜晓晓,张红娟,孟金来. 人类白细胞抗原G调控蜕膜自然杀伤细胞促进滋养细胞侵袭[J]. 山东大学学报 (医学版), 2022, 60(6): 41-45.
[8] 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.
[9] 申晓畅,孙一卿,颜磊,赵兴波. 芳基烃受体核转位因子样蛋白2在子宫内膜癌中的表达[J]. 山东大学学报 (医学版), 2022, 60(5): 74-80.
[10] 陈兆波,方敏,薛浩然,刘春艳. 去泛素化酶USP35促进非小细胞肺癌细胞迁移和侵袭[J]. 山东大学学报 (医学版), 2022, 60(4): 30-37.
[11] 钟黎黎,盛莹,郭江虹,阳双健,何宜静. LncRNA-UCA1通过靶向调控miR-182-5p对滋养细胞侵袭与转移的影响[J]. 山东大学学报 (医学版), 2022, 60(3): 76-82.
[12] 宋甜,付琳琳,王秋敏,杨晓,王莹,边月红,石玉华. 脂肪酸转运蛋白1在多囊卵巢综合征患者颗粒细胞中的表达[J]. 山东大学学报 (医学版), 2022, 60(2): 22-26.
[13] 亓梦雨,周敏然,孙洺山,李世洁,陈春燕. T大颗粒淋巴细胞白血病合并原发性骨髓纤维化1例[J]. 山东大学学报 (医学版), 2022, 60(2): 118-120.
[14] 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.
[15] 哈春芳,李茹月. 卵巢癌耐药机制与靶向治疗策略的研究进展[J]. 山东大学学报 (医学版), 2021, 59(9): 117-123.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!