切换至 "中华医学电子期刊资源库"

中华普通外科学文献(电子版) ›› 2024, Vol. 18 ›› Issue (01) : 51 -55. doi: 10.3877/cma.j.issn.1674-0793.2024.01.009

论著

顺铂对肝细胞癌Hep3B细胞程序性死亡配体1表达及药物敏感性的研究
赵向阳, 刘双池, 张懿刚, 陶滔, 谈燚()   
  1. 233000 蚌埠医学院第一附属医院肝胆外科
    233000 蚌埠市第三人民医院肝胆外科
  • 收稿日期:2023-05-05 出版日期:2024-02-01
  • 通信作者: 谈燚
  • 基金资助:
    安徽省高校自然科学研究重点项目(KJ2018ZD022); 蚌埠医学院研究生科研创新项目(Byycx22089); 蚌埠医学院科技项目(2022byzd096)

Study on programmed death ligand 1 expression and drug sensitivity of cisplatin in hepatocellular carcinoma Hep3B cells

Xiangyang Zhao, Shuangchi Liu, Yigang Zhang, Tao Tao, Yi Tan()   

  1. Department of Hepatobiliary Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
    Department of Hepatobiliary Surgery, the Third People’s Hospital of Bengbu, Bengbu 233000, China
  • Received:2023-05-05 Published:2024-02-01
  • Corresponding author: Yi Tan
引用本文:

赵向阳, 刘双池, 张懿刚, 陶滔, 谈燚. 顺铂对肝细胞癌Hep3B细胞程序性死亡配体1表达及药物敏感性的研究[J]. 中华普通外科学文献(电子版), 2024, 18(01): 51-55.

Xiangyang Zhao, Shuangchi Liu, Yigang Zhang, Tao Tao, Yi Tan. Study on programmed death ligand 1 expression and drug sensitivity of cisplatin in hepatocellular carcinoma Hep3B cells[J]. Chinese Archives of General Surgery(Electronic Edition), 2024, 18(01): 51-55.

目的

探讨顺铂对肝细胞癌Hep3B细胞程序性死亡配体1(PD-L1)表达以及PD-L1对顺铂敏感性的影响。

方法

使用不同浓度的顺铂(0、5、10、20 mg/L)处理Hep3B细胞,利用qPCR、Western blotting法检测细胞中PD-L1的表达情况。设置对照组(加入PBS)、顺铂组(5 mg/L)、MK2206组(AKT抑制剂,5 μmol/L)和顺铂+MK2206组(5 mg/L顺铂处理前1 h给予5 μmol/L MK2206),分别干预Hep3B细胞,Western blotting检测AKT、p-AKT和PD-L1的表达情况。siRNA-PD-L1转染Hep3B细胞,Western blotting检测PD-L1和上皮-间质转化标志物(E-cadherin和N-cadherin)表达变化,CCK-8、细胞划痕实验和Transwell实验检测细胞功能变化。

结果

顺铂可上调Hep3B细胞中PD-L1的蛋白表达水平(P<0.05)。与对照组相比,5 mg/L的顺铂显著促进Hep3B细胞中PD-L1的mRNA水平(P<0.05)。MK2206可抑制顺铂对p-AKT和PD-L1的上调(P<0.05),而AKT的蛋白水平差异无统计学意义。siRNA-PD-L1可抑制顺铂对Hep3B细胞PD-L1表达的上调(P<0.05),增强顺铂对Hep3B细胞增殖、迁移和侵袭的抑制,以及对E-cadherin的表达上调和N-cadherin的表达下调(P<0.05)。

结论

顺铂可促进肝细胞癌Hep3B细胞PD-L1表达,其机制可能与上调了AKT通路蛋白的磷酸化水平相关;抑制PD-L1表达可增强肝细胞癌Hep3B细胞对顺铂的敏感性。

Objective

To investigate the effect of cisplatin on the expression of programmed death ligand 1 (PD-L1) in hepatocellular carcinoma Hep3B cells and the effect of PD-L1 on cisplatin sensitivity.

Methods

Hep3B cells were treated with 0, 5, 10, 20 mg/L cisplatin, and the expression of PD-L1 was detected by qPCR and Western blotting. Hep3B cells were treated in control group (PBS only), cisplatin group (5 mg/L), MK2206 group (AKT inhibitor, 5 μmol/L) and cisplatin+MK2206 group (5 μmol/L MK2206 was administered 1 hour before cisplatin treatment), and the expressions of AKT, p-AKT and PD-L1 were detected by Western blotting. siRNA-PD-L1 was transfected into Hep3B cells. Western blotting was used to detect the expression of PD-L1 and epithelial-mesenchymal transition markers (E-cadherin and N-cadherin). CCK-8, scratch assay and Transwell test were used to detect the changes of cell function.

Results

Cisplatin could up-regulate the protein level of PD-L1 in Hep3B cells (P<0.05). Compared with the control group, 5 mg/L cisplatin significantly increased the mRNA level of PD-L1 in Hep3B cells (P<0.05). MK2206 could attenuate the promoting effect of cisplatin on p-AKT and PD-L1 expression in Hep3B cells (P<0.05). There was no significant difference in the protein level of AKT. siRNA-PD-L1 could attenuate the promoting effect of cisplatin on PD-L1 expression in Hep3B cells (P<0.05), enhance the inhibitory effect of cisplatin on proliferation, migration and invasion of Hep3B cells, enhance the promoting effect of cisplatin on E-cadherin expression and the inhibitory effect of cisplatin on N-cadherin expression in Hep3B cells (P<0.05).

Conclusions

Cisplatin can promote the expression of PD-L1 in hepatocellular carcinoma Hep3B cells, and its mechanism may be related to the up-regulation of phosphorylation level of AKT pathway proteins. Inhibition of PD-L1 expression may enhance the sensitivity of Hep3B cells to cisplatin.

图1 顺铂通过AKT通路促进Hep3B细胞PD-L1表达 A、B为Western blotting检测PD-L1的表达变化;C为qPCR检测PD-L1的表达变化;D、E为Western blotting检测p-AKT和AKT的表达变化;F、G为Western blotting检测PD-L1的表达变化;与0 mg/L组比较,*P<0.05;与顺铂组比较,#P<0.05
图2 抑制PD-L1表达增强Hep3B细胞顺铂敏感性 A为Western blotting检测PD-L1的表达变化;B为CCK-8检测Hep3B细胞的增殖变化;C为划痕实验检测Hep3B细胞的迁移变化;D为Transwell检测Hep3B细胞的侵袭变化(×200);E为Western blotting检测E-cadherin和N-cadherin的表达;与siRNA-NC组比较,*P<0.05;与顺铂+siRNA-NC组比较,#P<0.05
[1]
王乙钦, 李顺, 曾小芳, 等. 环状RNA circEIF6在肝细胞癌中的表达及功能研究[J/CD]. 中华普通外科学文献(电子版), 2021, 15(4): 252-257.
[2]
Finn RS, Zhu AX, Farah W, et al. Therapies for advanced stage hepatocellular carcinoma with macrovascular invasion or metastatic disease: A systematic review and meta-analysis[J]. Hepatology, 2018, 67(1): 422-435.
[3]
Ghosh S. Cisplatin: the first metal based anticancer drug[J]. Bioorg Chem, 2019, 88: 102925.
[4]
Li P, Song R, Yin F, et al. circMRPS35 promotes malignant progression and cisplatin resistance in hepatocellular carcinoma[J]. Mol Ther, 2022, 30(1): 431-447.
[5]
Spranger S, Gajewski TF. Impact of oncogenic pathways on evasion of antitumour immune responses[J]. Nat Rev Cancer, 2018, 18(3): 139-147.
[6]
Twomey JD, Zhang B. Cancer Immunotherapy update: FDA-approved checkpoint inhibitors and companion diagnostics[J]. AAPS J, 2021, 23(2): 39.
[7]
Glorieux C, Xia X, You X, et al. Cisplatin and gemcitabine exert opposite effects on immunotherapy with PD-1 antibody in K-ras-driven cancer[J]. J Adv Res, 2022, 40: 109-124.
[8]
Zhu H, Liu D, Deng S. Cisplatin promotes PD-L1 expression in A549 human lung adenocarcinoma cells via activating the ERK pathway[J]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi, 2021, 37(10): 891-896.
[9]
Shen B, Huang D, Ramsey AJ, et al. PD-L1 and MRN synergy in platinum-based chemoresistance of head and neck squamous cell carcinoma[J]. Br J Cancer, 2020, 122(5): 640-647.
[10]
Sheng Q, Zhang Y, Wang Z, et al. Cisplatin-mediated down-regulation of miR-145 contributes to up-regulation of PD-L1 via the c-Myc transcription factor in cisplatin-resistant ovarian carcinoma cells[J]. Clin Exp Immunol, 2020, 200(1): 45-52.
[11]
Hao X, Sun G, Zhang Y, et al. Targeting immune cells in the tumor microenvironment of HCC: new opportunities and challenges[J]. Front Cell Dev Biol, 2021, 9: 775462.
[12]
Xu L, Zhang K, Ma X, et al. Boosting cisplatin chemotherapy by nanomotor-enhanced tumor penetration and DNA adducts formation[J]. J Nanobiotechnology, 2022, 20(1): 429.
[13]
Muraro E, Vinante L, Fratta E, et al. Metronomic chemotherapy: anti-tumor pathways and combination with immune checkpoint inhibitors[J]. Cancers (Basel), 2023, 15(9): 2471.
[14]
Zhang P, Liu J, Li W, et al. Lactoferricin B reverses cisplatin resistance in head and neck squamous cell carcinoma cells through targeting PD-L1[J]. Cancer Med, 2018, 7(7): 3178-3187.
[15]
Wangpaichitr M, Kandemir H, Li YY, et al. Relationship of metabolic alterations and PD-L1 expression in cisplatin resistant lung cancer[J]. Cell Dev Biol, 2017, 6(2): 183.
[16]
Li Y, Zhai Y, Chen Y. GATA1-induced upregulation of LINC01503 promotes carboplatin resistance in ovarian carcinoma by upregulating PD-L1 via sponging miR-766-5p[J]. J Ovarian Res, 2021, 14(1): 108.
[17]
Wu M, Huang Q, Xie Y, et al. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation[J]. J Hematol Oncol, 2022, 15(1): 24.
[18]
周红建, 李军, 夏建国. 顺铂诱导脑胶质瘤U87细胞的PD-L1表达及耐药机制的研究[J]. 中国现代应用药学, 2022, 39(9): 1155-1161.
[19]
Amornsupak K, Thongchot S, Thinyakul C, et al. HMGB1 mediates invasion and PD-L1 expression through RAGE-PI3K/AKT signaling pathway in MDA-MB-231 breast cancer cells[J]. BMC Cancer, 2022, 22(1): 578.
[20]
Gao Y, Sun Z, Gu J, et al. Cancer-associated fibroblasts promote the upregulation of PD-L1 expression through Akt phosphorylation in colorectal cancer[J]. Front Oncol, 2021, 11: 748465.
[21]
Zhang H, Yuan X, Yang Y, et al. Cathelicidin LL-37 promotes EMT, migration and metastasis of hepatocellular carcinoma cells in vitro and mouse model[J]. Cell Adh Migr, 2023, 17(1): 20-34.
[22]
Du L, Wang L, Yang H, et al. Sex comb on midleg like-2 accelerates hepatocellular carcinoma cell proliferation and metastasis by activating Wnt/β-catenin/EMT signaling[J]. Yonsei Med J, 2021, 62(12): 1073-1082.
[23]
熊玮, 刘慧敏, 刘平, 等. lncRNA LSINCT5通过miR-451调控肺癌细胞侵袭、迁移及顺铂敏感性的实验研究[J]. 中国免疫学杂志, 2022, 38(11): 1349-1354.
[1] 李淼, 朱连华, 韩鹏, 姜波, 费翔. 高帧频超声造影评价肝细胞癌血管形态与风险因素的研究[J]. 中华医学超声杂志(电子版), 2023, 20(09): 911-915.
[2] 丁建民, 秦正义, 张翔, 周燕, 周洪雨, 王彦冬, 经翔. 超声造影与普美显磁共振成像对具有高危因素的≤3 cm肝结节进行LI-RADS分类诊断的前瞻性研究[J]. 中华医学超声杂志(电子版), 2023, 20(09): 930-938.
[3] 张华, 孙宇, 乡世健, 李樱媚, 王小群. 循环肿瘤细胞预测晚期胃肠癌患者化疗药物敏感性的研究[J]. 中华普通外科学文献(电子版), 2023, 17(06): 422-425.
[4] 董杰, 杨松, 杨浩, 陈翔, 张万里. 乙酰辅酶A羧化酶2基因高甲基化与肝细胞癌临床病理因素和生存期的关系[J]. 中华普通外科学文献(电子版), 2023, 17(06): 433-437.
[5] 王得晨, 杨康, 杨自杰, 归明彬, 屈莲平, 张小凤, 高峰. 结直肠癌微卫星稳定状态和程序性死亡、吲哚胺2,3-双加氧酶关系的研究进展[J]. 中华普通外科学文献(电子版), 2023, 17(06): 462-465.
[6] 黄威, 刘启, 陈流华, 滕茶香, 区喆建, 刘韩笑, 陈健聪, 张昆松. 新定义的可预测肝癌预后的焦亡相关lncRNA模型[J]. 中华普通外科学文献(电子版), 2023, 17(05): 357-365.
[7] 王荣昌, 欧奇峰, 黄晋杰, 王彩琴, 汪谦, 黄晓卉. miR-145-5p在肝细胞癌中的表达及其临床意义[J]. 中华普通外科学文献(电子版), 2023, 17(03): 197-202.
[8] 毛永欢, 奚玲, 陆晨, 刘理想, 喻春钊, 沈晓菲. PI3K/Akt信号通路通过Plk1影响胰腺癌细胞PANC-1对吉西他滨的化疗敏感性[J]. 中华普外科手术学杂志(电子版), 2023, 17(02): 135-138.
[9] 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.
[10] 孟俊杰, 李晓辉, 武晋荣, 郭光伟. 卡瑞丽珠单抗联合TP方案对晚期肺癌患者的疗效及作用机制分析[J]. 中华肺部疾病杂志(电子版), 2023, 16(05): 724-726.
[11] 曹亚娟, 黎兵华, 余德才. 转化治疗联合Laennec入路机器人右半肝切除治疗进展期肝癌[J]. 中华腔镜外科杂志(电子版), 2023, 16(02): 116-119.
[12] 王甜甜, 温媛, 李振, 叶美红, 郭影, 马双. 和厚朴酚调控Nrf2/ARE通路对胃癌细胞的顺铂化疗敏感性的影响[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 202-209.
[13] 胡宝茹, 尚乃舰, 高迪. 中晚期肝细胞癌的DCE-MRI及DWI表现与免疫治疗预后的相关性分析[J]. 中华消化病与影像杂志(电子版), 2023, 13(06): 399-403.
[14] 吴凤芸, 滕鑫, 刘连娟. 高帧频超声造影与增强磁共振对不同直径原发性高分化肝细胞癌的诊断价值[J]. 中华消化病与影像杂志(电子版), 2023, 13(06): 404-408.
[15] 刘中百, 任勇军. 肝细胞癌的介入治疗现状及进展[J]. 中华消化病与影像杂志(电子版), 2023, 13(02): 111-115.
阅读次数
全文


摘要