切换至 "中华医学电子期刊资源库"

中华肝脏外科手术学电子杂志 ›› 2022, Vol. 11 ›› Issue (01) : 92 -97. doi: 10.3877/cma.j.issn.2095-3232.2022.01.019

基础研究

circZKSCAN1抑制肝癌细胞侵袭转移的机制
邬杰忠1, 柯春连1, 陈少宏2, 刘波1, 姚志成1,()   
  1. 1. 510530 广州,中山大学附属第三医院岭南医院普通外科
    2. 510530 广州,中山大学附属第三医院岭南医院生物治疗中心
  • 收稿日期:2021-11-04 出版日期:2022-02-10
  • 通信作者: 姚志成
  • 基金资助:
    广东省自然科学基金(2018A030313641)

Mechanism of circZKSCAN1 inhibiting invasion and metastasis of hepatocellular carcinoma cells

Jiezhong Wu1, Chunlian Ke1, Shaohong Chen2, Bo Liu1, Zhicheng Yao1,()   

  1. 1. Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510530, China
    2. Biotherapy Center, Lingnan Hospital, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510530, China
  • Received:2021-11-04 Published:2022-02-10
  • Corresponding author: Zhicheng Yao
引用本文:

邬杰忠, 柯春连, 陈少宏, 刘波, 姚志成. circZKSCAN1抑制肝癌细胞侵袭转移的机制[J]. 中华肝脏外科手术学电子杂志, 2022, 11(01): 92-97.

Jiezhong Wu, Chunlian Ke, Shaohong Chen, Bo Liu, Zhicheng Yao. Mechanism of circZKSCAN1 inhibiting invasion and metastasis of hepatocellular carcinoma cells[J]. Chinese Journal of Hepatic Surgery(Electronic Edition), 2022, 11(01): 92-97.

目的

探讨circZKSCAN1抑制肝癌细胞侵袭转移的机制。

方法

利用前期构建的差异表达circZKSCAN1基因的HepG2肝癌细胞进行实验。采用克隆形成实验、划痕实验、Transwell细胞迁移和侵袭实验分别检测OE组(过表达circZKSCAN1的HepG2)、SH-1和SH-2组(沉默circZKSCAN1的HepG2)、NC组(阴性对照的HepG2)肝癌细胞的迁移和侵袭能力。采用生物信息学方法分析circZKSCAN1上富集RNA结合蛋白(RBP)的结合位点,并通过RNA免疫共沉淀(RIP)和RNA pull-down实验验证其靶标RBP,分析靶标RBP与肝癌生存预后的关系。4组克隆形成数、迁移和穿膜细胞数比较采用单因素方差分析和LSD-t检验。两组富集倍数比较采用t检验。生存分析采用Kaplan-Meier法和Log-rank检验。

结果

克隆形成实验显示,NC组、OE组、SH-1组、SH-2组克隆形成数分别为(10.5±1.1)、(4.7±0.8)、(21.7±1.8)、(22.6±2.6)个,与NC组相比,OE组克隆形成数明显减少,而SH-1组、SH-2组克隆形成数明显增多(LSD-t=-7.386,9.196,7.424;P<0.05)。划痕实验结果显示,与NC组相比,OE组24、48 h的迁移能力明显下降,而SH-1组、SH-2组24、48h的迁移能力则明显升高。Transwell细胞迁移实验表明,NC组、OE组、SH-1组、SH-2组迁移细胞数分别为(28.5±4.3)、(14.3±2.6)、(47.2±7.7)、(49.6±8.0)个,与NC组相比,OE组迁移细胞数明显减少,而SH-1组、SH-2组迁移细胞数明显增多(LSD-t=-4.895,3.673,3.474;P<0.05)。Transwell细胞侵袭实验显示,NC组、OE组、SH-1组、SH-2组穿膜细胞数分别为(21.3±2.1)、(7.6±1.5)、(38.6±5.8)、(41.3±7.1)个,与NC组相比,OE组穿膜细胞数明显减少,而SH-1组、SH-2组穿膜细胞数明显增多(LSD-t=-9.195,4.858,4.679;P<0.05) 。生物信息学分析显示,circZKSCAN1上富集RBP结合位点包括EIF4A3、FMR1、hnRNPC、U2AF65等。RIP实验显示circZKSCAN1可将细胞内的EIF4A3显著富集,与NC组相比,OE组富集倍数为(7.82±0.25)倍(t=1.057,P<0.05)。RNA pull-down实验显示EIF4A3可与circZKSCAN1结合。在线网站(http://gepia.cancer-pku.cn)分析显示,与癌旁组织相比,EIF4A3基因在肝癌组织中高表达,并主要出现在较晚分期的肝癌中(F=7.72,P<0.05),而高表达EIF4A3基因的肝癌患者具有更差的总体生存率(HR=1.90,P<0.05)。

结论

EIF4A3是circZKSCAN1的靶标RBP,circZKSCAN1可能通过与EIF4A3结合抑制肝癌细胞的侵袭转移能力。

Objective

To investigate the mechanism of circZKSCAN1 inhibiting the invasion and metastasis of hepatocellular carcinoma (HCC) cells.

Methods

HepG2 cells which differentially expressed circZKSCAN1 gene were utilized in this experiment. The migration and invasion capability of HCC cells in the OE group (HepG2 overexpressing circZKSCAN1), SH-1 and SH-2 groups (HepG2 silencing circZKSCAN1) and NC group (negative control) was assessed by colony formation assay, scratch test and Transwell chamber test, respectively. The binding sites of enriched RNA-binding protein (RBP) in circZKSCAN1 were identified by bioinformatics analysis. The target RBP was verified by RNA immunoprecipitation (RIP) and RNA pull-down experiment. The relationship between the target RBP and survival of HCC was analyzed. The number of colony formation, migration and number of membrane-penetrating cells among 4 groups were analyzed by single-factor analysis of variance and LSD-t test. The enrichment multiples between two groups were compared by t test. Survival analysis was performed by Kaplan-Meier method and Log-rank test.

Results

Colony formation assay showed that the number of colony formation in the NC, OE, SH-1 and SH-2 groups were 10.5±1.1, 4.7±0.8, 21.7±1.8 and 22.6±2.6, respectively. Compared with the NC group, the number of colony formation in the OE group was significantly reduced, whereas those in the SH-1 and SH-2 groups were significantly increased (LSD-t=-7.386, 9.196, 7.424; P<0.05). Scratch test demonstrated that compared with the NC group, the 24- and 48-h migration ability in the OE group was significantly decreased, whereas those in the SH-1 and SH-2 groups were significantly increased. Transwell chamber test found that the number of migrating cells in the NC, OE, SH-1 and SH-2 groups was 28.5±4.3, 14.3±2.6, 47.2±7.7 and 49.6±8.0, respectively. Compared with the NC group, the number of migrating cells in the OE group was significantly reduced, while those in the SH-1 and SH-2 groups were significantly elevated (LSD-t=-4.895, 3.673, 3.474; P<0.05). Transwell chamber test showed that the number of membrane-penetrating cells in the NC, OE, SH-1 and SH-2 groups was 21.3±2.1, 7.6±1.5, 38.6±5.8 and 41.3±7.1, respectively. Compared with the NC group, the number of membrane-penetrating cells in the OE group was significantly reduced, whereas those in the SH-1 and SH-2 groups were significantly increased (LSD-t=-9.195, 4.858, 4.679; P<0.05). Bioinformatics analysis revealed that enriched RBP-binding sites in circZKSCAN1 consisted of EIF4A3, FMR1, hnRNPC and U2AF65, etc. RIP experiment indicated that circZKSCAN1 could significantly enrich intracellular EIF4A3. Compared with the NC group, the enrichment multiple in the OE group was (7.82±0.25) times (t=1.057, P<0.05). RNA pull-down experiment showed that EIF4A3 could bind with circZKSCAN1. Online website (http://gepia.cancer-pku.cn) analysis demonstrated that compared with the adjacent tissues, EIF4A3 gene was highly expressed in HCC tissues, mainly in HCC of advanced stage (F=7.72, P<0.05). However, the overall survival rate of HCC patients with high expression level of EIF4A3 gene was even worse (HR=1.90, P<0.05).

Conclusions

EIF4A3 is the target RBP of circZKSCAN1. circZKSCAN1 may suppress the invasion and metastasis of HCC cells by binding with EIF4A3.

表1 RT-PCR引物序列
图1 克隆形成实验检测HepG2肝癌细胞克隆数注:NC为阴性对照组,OE为过表达circZKSCAN1组,SH-1和SH-2为沉默circZKSCAN1组
图2 划痕实验检测HepG2肝癌细胞迁移能力注:NC为阴性对照组,OE为过表达circZKSCAN1组,SH-1和SH-2为沉默circZKSCAN1组
图3 生物信息学检测EIF4A3在肝癌中的表达及其与生存预后的关系注:a为EIF4A3基因在肝癌中的表达情况,LIHC为肝细胞肝癌,T为肝癌组,N为正常组;b为EIF4A3基因与肝癌分期的关系;c为EIF4A3水平与肝癌患者总体生存的Kaplan-Meier生存分析
[1]
Forner A, Reig M, Bruix J.Hepatocellular carcinoma[J].Lancet,2018,391(10127):1301-1314.
[2]
Sun H, Tang W, Rong D,et al.Hsa_circ_0000520, a potential new circular RNA biomarker, is involved in gastric carcinoma[J].Cancer Biomark,2018,21(2):299-306.
[3]
Li A, Wang WC, McAlister V,et al.Circular RNA in colorectal cancer[J].J Cell Mol Med,2021,25(8):3667-3679.
[4]
Wei X, Shi Y, Dai Z,et al.Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review)[J].Int J Oncol,2021,58(3):289-297.
[5]
Zhao X, Cai Y, Xu J.Circular RNAs: biogenesis, mechanism, and function in human cancers[J].Int J Mol Sci,2019,20(16):3926.
[6]
Yao Z, Luo J, Hu K,et al.ZKSCAN1 gene and its related circular RNA (circZKSCAN1) both inhibit hepatocellular carcinoma cell growth, migration, and invasion but through different signaling pathways[J].Mol Oncol,2017,11(4):422-437.
[7]
Zhang KM, Pan XY, Yang Y,et al.CRIP: predicting circRNA-RBP interaction sites using a codon-based encoding and hybrid deep neural networks[J].RNA,2019,25(12):1604-1615.
[8]
Kazimierczyk M, Kasprowicz MK, Kasprzyk ME,et al.Human long noncoding RNA interactome: detection, characterization and function[J].Int J Mol Sci,2020,21(3):1027.
[9]
Li C, Zhou H.Circular RNA hsa_circRNA_102209 promotes the growth and metastasis of colorectal cancer through miR-761-mediated Ras and Rab interactor 1 signaling[J].Cancer Med,2020,9(18):6710-6725.
[10]
Viralippurath Ashraf J, Sasidharan Nair V, Saleh R,et al.Role of circular RNAs in colorectal tumor microenvironment[J].Biomed Pharmacother,2021(137):111351.
[11]
Zhang C, Yang Q, Li W,et al.Roles of circRNAs in prostate cancer: expression, mechanism, application and potential[J].Int J Biochem Cell Biol,2021(134):105968.
[12]
Long F, Lin Z, Li L,et al.Comprehensive landscape and future perspectives of circular RNAs in colorectal cancer[J].Mol Cancer,2021,20(1):26.
[13]
Gebauer F, Schwarzl T, Valcárcel J,et al.RNA-binding proteins in human genetic disease[J].Nat Rev Genet,2021,22(3):185-198.
[14]
Zhu Y, Ren C, Yang L.Effect of eukaryotic translation initiation factor 4A3 in malignant tumors[J].Oncol Lett,2021,21(5):358.
[15]
Kwon OS, Mishra R, Safieddine A,et al.Exon junction complex dependent mRNA localization is linked to centrosome organization during ciliogenesis[J].Nat Commun,2021,12(1):1351.
[16]
Singh K, Lin J, Lecomte N,et al.Targeting eIF4A-dependent translation of KRAS signaling molecules[J].Cancer Res,2021,81(8):2002-2014.
[17]
Sanghvi VR, Mohan P, Singh K,et al.NRF2 activation confers resistance to eIF4A inhibitors in cancer therapy[J].Cancers,2021,13(4):639.
[18]
Sun HD, Xu ZP, Sun ZQ,et al.Down-regulation of circPVRL3 promotes the proliferation and migration of gastric cancer cells[J].Sci Rep,2018,8(1):10111.
[19]
Tang W, Wang D, Shao L,et al.LINC00680 and TTN-AS1 stabilized by EIF4A3 promoted malignant biological behaviors of glioblastoma cells[J].Mol Ther Nucleic Acids,2020(19):905-921.
[20]
Xu B, Yang N, Liu Y,et al.Circ_cse1l inhibits colorectal cancer proliferation by binding to eIF4A3[J].Med Sci Monit,2020(26):e923876.
[1] 张生军, 赵阿静, 李守博, 郝祥宏, 刘敏丽. 高糖通过HGF/c-met通路促进结直肠癌侵袭和迁移的实验研究[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 21-24.
[2] 叶晓琳, 刘云飞, 庞明泉, 王海久, 任利, 侯立朝, 于文昊, 王志鑫, 樊海宁. 肝再生细胞来源及调控机制的研究进展[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 96-99.
[3] 晏晴艳, 雍晓梅, 罗洪, 杜敏. 成都地区老年转移性乳腺癌的预后及生存因素研究[J]. 中华普外科手术学杂志(电子版), 2023, 17(06): 636-638.
[4] 王兴, 张峰伟. 腹腔镜肝切除联合断面射频消融治疗伴微血管侵犯肝细胞癌的临床研究[J]. 中华普外科手术学杂志(电子版), 2023, 17(05): 580-583.
[5] 杜锡林, 谭凯, 贺小军, 白亮亮, 赵瑶瑶. 肝细胞癌转化治疗方式[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 597-601.
[6] 魏小勇. 原发性肝癌转化治疗焦点问题探讨[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 602-607.
[7] 张其坤, 商福超, 李琪, 栗光明, 王孟龙. 联合脾切除对肝癌合并门静脉高压症患者根治性切除术后的生存获益分析[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 613-618.
[8] 严庆, 刘颖, 邓斐文, 陈焕伟. 微血管侵犯对肝癌肝移植患者生存预后的影响[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 624-629.
[9] 张文华, 陶焠, 胡添松. 不同部位外生型肝癌临床病理特点及其对术后肝内复发和预后影响[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 651-655.
[10] 韩宇, 张武, 李安琪, 陈文颖, 谢斯栋. MRI肝脏影像报告和数据系统对非肝硬化乙肝患者肝细胞癌的诊断价值[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 669-673.
[11] 张维志, 刘连新. 基于生物信息学分析IPO7在肝癌中的表达及意义[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 694-701.
[12] 陈安, 冯娟, 杨振宇, 杜锡林, 柏强善, 阴继凯, 臧莉, 鲁建国. 基于生物信息学分析CCN4在肝细胞癌中表达及其临床意义[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 702-707.
[13] 叶文涛, 吴忠均, 廖锐. 癌旁组织ALOX15表达与肝癌根治性切除术后预后的关系[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 708-712.
[14] 吴晨瑞, 廖锐, 贺强, 潘龙, 黄平, 曹洪祥, 赵益, 王永琛, 黄俊杰, 孙睿锐. MDT模式下肝动脉灌注化疗联合免疫靶向治疗肝细胞癌多处转移一例[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 713-716.
[15] 李田, 徐洪, 刘和亮. 尘肺病的相关研究进展[J]. 中华临床医师杂志(电子版), 2023, 17(08): 900-905.
阅读次数
全文


摘要