切换至 "中华医学电子期刊资源库"

中华临床医师杂志(电子版) ›› 2023, Vol. 17 ›› Issue (11) : 1181 -1191. doi: 10.3877/cma.j.issn.1674-0785.2023.11.008

基础研究

自噬及内质网应激在卡非佐米对MCF-7细胞的影响及作用机制
包文华, 塔拉()   
  1. 010020 内蒙古呼和浩特,内蒙古自治区人民医院乳腺肿瘤外科
    010020 内蒙古呼和浩特,内蒙古自治区人民医院甲状腺肿瘤外科
  • 收稿日期:2023-06-26 出版日期:2023-11-15
  • 通信作者: 塔拉
  • 基金资助:
    内蒙古自然科学基金(2018BS08006)

Role of autophagy and endoplasmic reticulum stress in pro-apoptosis effect of carfilzomib on MCF-7 cells

Wenhua Bao, La Ta()   

  1. Department of Breast Oncology Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010020, China
    Department of Thyroid Oncology Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010020, China
  • Received:2023-06-26 Published:2023-11-15
  • Corresponding author: La Ta
引用本文:

包文华, 塔拉. 自噬及内质网应激在卡非佐米对MCF-7细胞的影响及作用机制[J]. 中华临床医师杂志(电子版), 2023, 17(11): 1181-1191.

Wenhua Bao, La Ta. Role of autophagy and endoplasmic reticulum stress in pro-apoptosis effect of carfilzomib on MCF-7 cells[J]. Chinese Journal of Clinicians(Electronic Edition), 2023, 17(11): 1181-1191.

目的

探讨自噬及内质网应激在卡非佐米(Carfilzomib)对MCF-7细胞凋亡的影响及作用机制。

方法

体外培养MCF-7乳腺癌细胞,分为对照组及实验组,对照组进行传代培养,实验组卡非佐米(0.625、1.25、2.5、5、10、20、40、80、160、320 nmol/L)作用0 h、3 h、6 h、12 h、24 h、48 h,自噬抑制剂3-MA(0.5、1、5、10、20、40、80、160 nmol/L)作用48 h,内质网应激抑制剂Salubrinal(2.5、5、10、20、40、80、160、320 umol/L)作用48 h,采用MTT比色法检测卡非佐米、3-MA、Salubrinal对MCF-7细胞活力的影响.确定实验组的各药物浓度后,实验分为对照组,卡非佐米3-MA组,卡非佐米+Salubrinal组,流式细胞仪检测细胞凋亡,荧光定量PCR检测mRNA表达水平,Western blot法检测蛋白表达水平。

结果

MTT检测结果显示卡非佐米随着浓度的升高和作用时间的延长细胞活性抑制作用增强,呈浓度依赖性与时间依赖性,当卡非佐米浓度为10 nmol/L作用48 h,抑制率为20.03±0.34**。3-MA,Salubrinal对MCF-7细胞增值抑制作用随着浓度增加作用增强。流式细胞检测结果提示与对照组相比,卡非佐米能够诱导MCF-7的凋亡,凋亡率为3.54%。协同3-MA和Salubrinal处理后,凋亡率增加至5.04%和6.95%。荧光定量PCR结果显示与卡非佐米组相比,卡非佐米+3-MA和卡非佐米+Salubrinal作用后,Grp-78和Caspase-12转录的mRNA均减少,Salubrinal减少作用更显著,而GADD153的表达在3-MA组和Salubrinal组差异并无统计学意义。与 mRNA 检测结果一致,3-MA组和Salubrinal组Grp-78蛋白的表达水平显著降低,Salubrinal对蛋白的降低作用更明显,GADD153在3-MA组和Salubrinal组表达差异无统计学意义。Caspase-12的表达则3-MA组低于Salubrinal组。

结论

卡非佐米抑制MCF-7细胞增值,同时诱导细胞自噬,且部分通过内质网应激通路,抑制自噬与内质网应激信号通路能够增强卡非佐米对MCF-7细胞的促凋亡作用。

Objective

To investigate the effect of autophagy and endoplasmic reticulum (ER) stress on MCF-7 cells following carfilzomib treatment.

Methods

MCF-7 breast cancer cells cultured in vitro were divided into a control group and an experimental group. The experimental group was treated with carfilzomib at 10, 20, 40, 80, 160, and 320 nmol /L for 0, 3, 6, 12, 24, and 48 h, 3-methyladenine (3-MA, an autophagy inhibitor) at 0.5, 1, 5, 10, 20, 40, 80, and 160 nmol/L for 48 h, and salubrinal (an ER stress inhibitor) at 2.5, 5, 10, 20, 40, 80, 160, and 320 μmol/L for 48 h. MCF-7 cell viability was detected by MTT assay to determine the optimal concentration of the drugs used. Cells were then treated with carfilzomib alone, carfilzomib+3-MA, and carfilzomib+salubrinal. Cell apoptosis was detected by flow cytometry, mRNA level was detected by real-time PCR, and protein expression level was detected by Western blot.

Results

Treatment with carfilzomib inhibited the viability of MCF-7 cells, and the inhibitory rate was increased significantly with the increase in the concentration and treatment duration. When cells were treated with carfilzomib at 10 nmol/L for 48 h, the inhibition rate for MCF-7 cells was 20.03±0.34. 3-MA and salubrinal inhibited the viability of MCF-7 cells in a dose-dependent manner. Compared with untreated control cells, carfilzomib induced the apoptosis of MCF-7 cells, with an apoptosis rate of 3.54%. Co-treatment of carfilzomib with either 3-MA or salubrinal resulted in enhanced cell apoptosis (apoptosis rate: 5.04% and 6.95%, respectively) compared with cells treated with carfilzomib alone. Compared with the carfilzomib alone group, cotreatment with 3-MA and salubrinal significantly decreased the mRNA expression levels of Grp-78 and caspase-12, but had no signifciant impact on the mRNA expression level of GADD153. In addition, consistent with mRNA detection results, the expression level of GRP-78 in the carfilzomib+3-MA group and the carfilzomib+salubrinal group was significantly protein reduced. Although salubrinal had an more obvious effect on the protein reduction, there was no significant difference in the expression of GADD153 between the carfilzomib+3-MA group and the carfilzomib+salubrinal group. However, caspase-12 expression was decreased significantly in the carfilzomib+3-MA group compared with the carfilzomib+salubrinal group.

Conclusion

Carfilzomib inhibits MCF-7 cell proliferation and induces autophagy partially through the ER stress pathway. Inhibiting autophagy and ER stress can enhance the pro-apoptosis effect of carfilzomib on MCF-7 cells.

表1 Real Time PCR反应体系
表2 引物序列
表3 卡非佐米对乳腺癌MCF-7细胞的抑制检测
图1 卡非佐米对MCF-7细胞的增殖抑制作用。抑制作用6 h开始出现,8 h开始明显,作用48 h抑制效果最显著,并且随着浓度增加抑制作用增强,当卡非佐米浓度为10 nmol/L作用48 h,细胞增殖开始明显受抑制,抑制率为20.03±0.34a,卡非佐米在48 h的IC20值约为10 nmol/L。
表4 3-MA对乳腺癌MCF-7细胞作用的MTT检测结果
图2 3-MA作用下MCF-7乳腺癌细胞抑制率曲线.随着3-MA浓度变大,MCF-7细胞抑制率明显变大。(与对照组相比P<0.05)
表5 Salubrinal对乳腺癌MCF-7细胞作用的MTT检测结果
图3 Salubrinal对乳腺癌MCF-7细胞的增殖抑制作用曲线。随着Salubrinal浓度变大,MCF-7细胞抑制率明显变大(与对照组相比P<0.05)
图4 图a为各组作用12 h的OD值;图b为为各组作用24 h的OD值;图c为各组作用48 h的OD值
图5 流式细胞仪检测MCF-7细胞凋亡。图A为空白对照组;图B为卡非佐米组;图C为卡非佐米+3-MA组;图D为卡非佐米+Salubrinal组。
图6 3-MA和Salubrinal对卡非佐米诱导的MCF-7细胞自噬相关蛋白表达的影响。图a为自噬相关蛋白LC3蛋白电泳图;图b为自噬相关蛋白LC3-I蛋白灰度图;图c为自噬相关蛋白LC3-II蛋白灰度图
图7 3-MA和Salubrinal对卡非佐米诱导的MCF-7细胞凋亡相关蛋白表达的影响。图a为凋亡相关蛋白Bax、caspase-3及Bcl-2蛋白电泳图;图b为凋亡相关蛋白Bax蛋白灰度图;图c为凋亡相关蛋白caspase-3蛋白灰度图;图d为凋亡相关蛋白Bcl-2蛋白灰度图
图8 3-MA和Salubrinal对内质网应激相关蛋白表达的影响。图a为内质网应激相关蛋白GRP-78,GADD153和,Caspase-12的蛋白电泳图;图b为内质网应激相关蛋白GRP-78蛋白灰度图;图c为内质网应激相关蛋白GADD153蛋白灰度图;图d为内质网应激相关蛋白Caspase-12蛋白灰度图
图9 3-MA和Salubrinal对Grp-78,GADD153与Caspase-12 mRNA表达的影响
1
Global Burden of Disease Cancer Collaboration; Fitzmaurice C, Allen C, Barber RM, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study [J]. JAMA Oncol, 2017, 3(4): 524-548.
2
Torre LA, Siegel RL, Ward EM, et al. Global cancer incidence and mortality rates and trends--an update [J]. Cancer Epidemiol Biomarkers Prev, 2016, 25(1): 16-27.
3
El Yaagoubi OM, Oularbi L, Bouyahya A, et al. The role of the ubiquitin-proteasome pathway in skin cancer development: 26S proteasome-activated NF-κB signal transduction [J]. Cancer Biol Ther, 2021, 22(10-12): 479-492.
4
Storz P. Targeting the alternative NF-κB pathway in pancreatic cancer: a new direction for therapy? [J]. Expert Rev Anticancer Ther, 2013, 13(5): 501-504.
5
Andela VB, Schwarz EM, O'Keefe RJ, et al. A genome-wide expression profile and system-level integration of nuclear factor kappa B regulated genes reveals fundamental metabolic adaptations during cell growth and survival [J]. FEBS Lett, 2005, 579(30): 6814-6820.
6
马李洁, 李莉娟, 张连生, 等. 多发性骨髓瘤治疗现状及前景 [J]. 临床荟萃, 2015, 30(4): 476-480
7
马勇, 汪兴洪. 多发性骨髓瘤治疗进展 [J]. 亚太传统医药, 2012, 8(4): 201-202.
8
Genin E, Reboud-Ravaux M, Vidal J. Proteasome inhibitors: recent advances and new perspectives in medicinal chemistry [J]. Curr Top Med Chem, 2010, 10(3): 232-256.
9
邢志佳, 张叶叶, 马宇衡, 等. 针对 20S 蛋白酶体的抗肿瘤药物研究进展 [J]. 天津药学, 2015, 27(1): 55-57.
10
Liu L, Zhao N, Xu W, et al. Pooled analysis of the reports of carfilzomib, panobinostat, and elotuzumab combinations in patients with refractory/relapsed multiple myeloma [J]. J Hematol Oncol, 2016, 9(1): 54.
11
Wang H, Guan F, Chen D, et al. An analysis of the safety profile of proteasome inhibitors for treating various cancers [J]. Expert Opin Drug Saf, 2014, 13(8): 1043-1054.
12
Lisa Z, Myriem B, Ross L, et al. Carflzomib potentiates CUDC-101-nduced apoptosis in anaplastic thyroid cancer [J]. Oncotarget, 2016, 7(13): 16517-16528.
13
Areeb Z, Stylli SS, Ware TM, et al. Inhibition of glioblastoma cell proliferation, migration and invasion by the proteasome antagonist carfilzomib [J]. Med Oncol, 2016, 33(5): 53.
14
Alanazi A, Algfeley SG, Al-Hosaini KA, et al. Therapeutic potential of carfilzomib, an irreversible proteasome inhibitor, against acetaminophen-induced hepatotoxicity in mice [J]. J Biochem Mol Toxicol, 2017, 31(4).
15
Jeon YJ, Khelifa S, Ratnikov B, et al. Regulation of glutamine carrier proteins by RNF5 determines breast cancer response to ER stress-inducing chemotherapies [J]. Cancer Cell, 2015, 27(3): 354-369.
16
Li Y, Li J, Li S, et al. Curcumin attenuates glutamate neurotoxicity in the hippocampus by suppression of ER stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on AMPK [J]. Toxicol Appl Pharmacol, 2015, 286(1): 53-63.
17
Høyer-Hansen M, Jäättelä M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium [J]. Cell Death Differ, 2007, 14(9): 1576-1582.
18
Ding WX, Ni HM, Gao W, et al. Linking of autophagy to ubiquitin-proteasome system is important for the regulation of endoplasmic reticulum stress and cell viability [J]. Am J Pathol, 2007, 171(2): 513-524.
19
Moretti L, Cha YI, Niermann KJ, et al. Switch between apoptosis and autophagy: radiation-induced endoplasmic reticulum stress?[J]. Cell Cycle, 2007, 6(7): 793-798.
[1] 李康, 冀亮, 赵维, 林乐岷. 自噬在乳腺癌生物学进展中的双重作用[J]. 中华乳腺病杂志(电子版), 2023, 17(04): 195-202.
[2] 孔莹莹, 谢璐涛, 卢晓驰, 徐杰丰, 周光居, 张茂. 丁酸钠对猪心脏骤停复苏后心脑损伤的保护作用及机制研究[J]. 中华危重症医学杂志(电子版), 2023, 16(05): 355-362.
[3] 张晓燕, 肖东琼, 高沪, 陈琳, 唐发娟, 李熙鸿. 转录因子12过表达对脓毒症相关性脑病大鼠大脑皮质的保护作用及其机制[J]. 中华妇幼临床医学杂志(电子版), 2023, 19(05): 540-549.
[4] 刘星辰, 刘娟, 魏宝宝, 刘洁, 刘辉. XIAP与XAF1异常表达与卵巢癌的相关性分析[J]. 中华妇幼临床医学杂志(电子版), 2023, 19(04): 419-427.
[5] 周子慧, 李恭驰, 李炳辉, 王知, 刘慧真, 王卉, 邹利军. 细胞自噬在创面愈合中作用的研究进展[J]. 中华损伤与修复杂志(电子版), 2023, 18(06): 542-546.
[6] 张永博, 张亮, 陈浏阳, 戴睿, 孙华, 杨盛, 孟博, 彭晴. 线粒体与椎间盘退变[J]. 中华损伤与修复杂志(电子版), 2023, 18(03): 265-269.
[7] 刘硕儒, 王功炜, 张斌, 李书豪, 胡成. 新型溶瘤病毒M1激活内质网应激致前列腺癌细胞凋亡的机制[J]. 中华腔镜泌尿外科杂志(电子版), 2023, 17(04): 388-393.
[8] 邵浩仁, 郭佳. 铁死亡的分子机制及其在前列腺癌治疗中的研究进展[J]. 中华腔镜泌尿外科杂志(电子版), 2023, 17(03): 294-298.
[9] 许娟, 张党锋. 尼达尼布对肺纤维化小鼠肺功能及内质网应激反应的影响[J]. 中华肺部疾病杂志(电子版), 2023, 16(05): 673-675.
[10] 于迪, 于海波, 吴焕成, 李玉明, 苏彬, 陈馨. 发状分裂相关增强子1差异表达对胆固醇刺激下血管内皮细胞的影响[J]. 中华脑科疾病与康复杂志(电子版), 2023, 13(05): 264-270.
[11] 王小红, 钱晶, 翁文俊, 周国雄, 朱顺星, 祁小鸣, 刘春, 王萍, 沈伟, 程睿智, 秦璟灏. 巯基丙酮酸硫基转移酶调控核因子κB信号介导自噬对重症急性胰腺炎大鼠的影响及机制[J]. 中华消化病与影像杂志(电子版), 2023, 13(06): 422-426.
[12] 邓世栋, 刘凌志, 郭大勇, 王超, 黄忠欣, 张晖辉. 沉默SNHG1基因对膀胱癌细胞增殖、凋亡、迁移和铁死亡的影响[J]. 中华临床医师杂志(电子版), 2023, 17(07): 804-811.
[13] 张敏洁, 张小杉, 段莎莎, 施依璐, 赵捷, 白天昊, 王雅晳. 氢气治疗心肌缺血再灌注损伤的作用机制及展望[J]. 中华临床医师杂志(电子版), 2023, 17(06): 744-748.
[14] 郭如烨, 孟黎明, 陈楠, 宋玉莹, 尹海燕, 郭岩. Apelin/APJ系统对帕金森病模型的神经保护作用及机制研究进展[J]. 中华诊断学电子杂志, 2023, 11(04): 276-282.
[15] 邱甜, 杨苗娟, 胡波, 郭毅, 何奕涛. 亚低温治疗脑梗死机制的研究进展[J]. 中华脑血管病杂志(电子版), 2023, 17(05): 518-521.
阅读次数
全文


摘要