·实验研究·

沉默FOXC1对宫颈癌HeLa细胞侵袭和转移能力的影响

张婕 汪露 韩世愈

基金项目:2019年哈尔滨医科大学创新基金

作者单位:俄罗斯莫斯科,莫斯科国立第一医科大学(张婕);哈尔滨医科大学附属第四医院妇产科(汪露,韩世愈)

通讯作者:韩世愈(shiyu962@163.com)

【摘要】 目的 研究小干扰RNA(siRNA)沉默人宫颈癌细胞中叉头框蛋白C1(FOXC1)基因对宫颈癌细胞侵袭及转移能力的影响。方法研究NC104、宫颈癌 CaSki、ME-180、HeLa细胞中FOXC1基因的表达,筛选出高表达FOXC1的HeLa细胞株,构建特异性慢病毒干扰载体FOXC1-shRNA,感染细胞株,通过Western blot和Real-time PCR等实验方法检测转染效率。具体分组为:感染阴性组(HeLa)、空白对照组(HeLa-GFP)、感染FOXC1-shRNA组(HeLa-FOXC1(-))。通过Transwell小室及Matrigel基质胶鉴定转染后三组细胞的迁移和侵袭性。并通过Western Blot实验方法检测转染后三组细胞中Vimentin、E-cadherin及β-catenin与EMT相关蛋白表达的变化,进一步验证转染后三种细胞迁移和侵袭性的变化。 结果RT-PCR以及Western blot结果分别提示,HeLa-FOXC1(-)组,FOXC1 mRNA以及FOXC1蛋白表达均被抑制。Transwell迁移及侵袭实验显示,HeLa-FOXC1(-)组细胞的迁移及侵袭能力明显降低,Western blot检测三组细胞中Vimentin、E-cadherin及β-catenin的表达,发现HeLa-FOXC1(-)组细胞系内Vimentin及β-catenin蛋白的表达明显降低,E-cadherin蛋白的表达有增高,以上结果差异均有统计学意义。结论敲除人宫颈癌HeLa细胞系中的FOXC1的表达,上皮间质转化相关β-catenin及Vimentin蛋白表达也随之降低。E-cadherin蛋白的表达增高,能够影响细胞的生物学行为,使其侵袭及转移能力降低。

【关键词】 宫颈癌; FOXC1; 侵袭及转移

Knockdown of FOXC1 in a cervical cancer Hela cell line and its effects on cell invasion and metastasis

ZHANG Jie, WANG Lu, HAN Shiyu.

Moscow State First Medical University, Moscow, Russia

[Abstract] Objective To investigate the effect of invasion and metastasis ability of cervical cancer cells when their forhead box C1 (FOXC1) gene was knocked down by small interfering RNA (siRNA).MethodsThe expression of FOXC1 gene in NC104, CaSki, me-180 and HeLa cells of cervical cancer was detected, and the HeLa cell lines with high FOXC1 expression were screened out. The specific lentiviral interference vector foxc1-shrna was constructed to infect cell lines, and the transfection efficiency was detected by Western blot and real-time PCR. The samples were divided into three groups:infection negative group (HeLa), blank control group (hela-gfp), and infection foxc1-shrna group (hela-foxc1 (-)). The migration and invasiveness of the transfected cells were determined by Transwell assay and Matrigel. To further verify the changes of migration and invasion of the three types of cells after transfection, the changes of emt-related proteins such as Vimentin, e-cadherin and β-catenin in the three groups of cells after transfection were detected by Western Blot.ResultsReal-time PCR and Western blot results indicated that the expression of FOXC1 mRNA and FOXC1 protein were inhibited in the hela-foxc1 (-) group. The Transwell migration experiment showed that the migration ability of cells in the hela-foxc1 (-) group was significantly reduced. The Transwell and Matrigel invasion experiment showed that the invasion ability of cells in the hela-foxc1 (-) group was significantly reduced. Western blot analysis showed that the expressions of Vimentin, e-cadherin and β-catenin in the three groups were significantly decreased. The expression of e-cadherin protein was increased. These differences among the three groups were considered statistically significant (P<0.05).ConclusionWe found that knockdown of FOXC1 gene suppressed the expression of compound β-catenin and Vimentin. The increased expression of e-cadherin protein can affect biological behavior of cervical cancer cells and decrease their invasion and metastasis ability.

Key words] cervical cancer; FOXC1; invasion and metastasis

宫颈癌(cervical cancer,CESC)属于女性生殖系统恶性肿瘤,发病率及死亡率较高,对女性生命和健康存在极大威胁。据统计,全球每年新发病例约53万人,死亡人数高达27万人[1]。临床统计资料显示,80%以上有夫妻生活经历的女性存在程度不等的宫颈疾病困扰,而处于17岁~28岁阶段的女性更易患宫颈疾病,一些女性未及30岁即发生了早期宫颈病变,有的甚至出现晚期宫颈病变,在发病年龄方面相较于以往提早了近十年[2]。在恶性肿瘤的迁移与侵袭活动中,上皮间质转化(epithelial mesenchymal transition,EMT)发挥着关键性影响。肿瘤的形成与发展过程涉及诸多基因以及步骤,其中EMT对于源于上皮的恶性肿瘤迁移与侵袭行为发挥着关键性影响[3]。EMT,即在一定生理与病理环境中上皮细胞转分化为间充质细胞的行为。此行为发生期间,上皮细胞的细胞极性、细胞间的黏附性逐渐丧失,游走性能与侵入能力显著提升[4]。叉头框C1蛋白 (forkhead box protein C1, FOXC1)是一种新型转录因子,研究显示,FOXC1在恶性肿瘤发病、进展过程中发挥着重要作用,例如,在鼻咽癌中检测到FOXC1的高表达,而FOXC1通过上调波形蛋白、纤维连接蛋白和N-cadherin的表达[5],在EMT中起到关键作用。FOXC1在乳腺癌中是EGFR功能的关键调节因子,目前已发现FOXC1的显著上调可通过激活Hedgehog信号通路来调控癌症干细胞特性的丰富[6]。在肝细胞癌中,FOXC1通过调节EMT参与微血管浸润[7]。有报道表明,FOXC1的异常表达与多种癌症的发生和发展有关,包括乳腺癌、肝细胞癌、胰腺癌和非小细胞肺癌[8-11]。然而,FOXC1的表达及其在宫颈癌中的潜在作用尚不清楚。本研究旨在探讨沉默FOXC1的宫颈癌细胞的侵袭及转移能力的影响情况,现将结果报告如下。

材料及方法

一、材料

正常宫颈细胞NC104与人宫颈癌 CaSki、ME-180、HeLa细胞购于中科院上海肿瘤研究所,细胞均用含10%胎牛血清、100 U/mL 青霉素、100 μg/mL链霉素的DMEM培养液,置于37℃、5% CO2的恒温培养箱中培养。引物设计后由上海生工合成,LipofectamineTM转染试剂购于汉恒生物,pLKO.1-shFOXC1(本实验室克隆)。β-catenin抗体、Vimentin抗体、E-cadherin抗体购于美国Affinty 公司,TRANSWELL小室及Matrigel基质胶购于corning公司。DMEM培养基和小牛血清购自Gibco公司。

二、四种细胞中FOXC1mRNA及蛋白水平表达

1.细胞总RNA用TRIzol试剂盒提取:RT-PCR 95℃ 30 s,95℃ 5 s 40个循环,60℃ 30 s。引物序列如下。

FOXC1:

5′-CTG CCC GAC TAC TCT CTG C-3′

5′-CAC CGA GTG GAA GTT CTG C-3′;

GAPDH:

5′-CGA GAT CCC TCC AAA ATC AA-3′

5′-TTC ACA CCC ATG ACG AAC AT-3′。

采用2-ΔΔCT法计算FOXC1 mRNA相对表达量。

2.Western Blot检测四种宫颈癌细胞中FOXC1基因的表达:FOXC1蛋白表达采用Western blot法。收集四组细胞,加入蛋白裂解液,冰上裂解充分约30 min,提取总蛋白,进行蛋白定量检测并制备蛋白样品。SDS-PAGE电泳,湿转法转膜,5% BSA摇床室温封闭1 h,1×TBST洗涤3次,每次15 min。加入FOXC1(1∶1 000)和内参GAPDH(1∶1000)一抗,4 ℃孵育过夜,1×TBST洗涤3次,每次15 min;加入二抗(1∶2 000)室温杂交1 h,1×TBST洗涤3次,每次15 min;ECL显影、曝光。实验均重复3次以上,采用Image J软件分析蛋白条带灰度值,以目的条带与内参条带灰度值的比值计算FOXC1蛋白相对表达量。根据RT-PCR及Western blot检测宫颈癌HeLa细胞中FOXC1表达量较高。

三、基因转染

取对数生长期的HeLa细胞,无血清培养基重悬,调整细胞密度为4×105/mL,6孔板中每孔2 mL细胞悬液接种,常规培养。待细胞状态良好且融合度达60%~80%时,更换为无血清培养基。实验分为3组。 (1)对照组:转染阴性的Hela细胞; (2)阴性对照组:用pc DNA3.1 (+) -neo质粒转染Hela细胞; (3) 转染特异性siRNA组:用pLKO.1-shFOXC1转染Hela细胞。转染方法参照Lipofectamine2000转染方法进行操作。转染48 h后进行培养, 3 d后嘌呤霉素筛选阳性细胞连代培养,通过倒置显微镜观察转染效率,RT-PCR及Western Blot检测转染后细胞中FOXC1mRNA及蛋白表达,达到稳定转染后进行后续实验。

四、Transwell小室迁移及侵袭实验

1.检测转染后细胞迁移能力:制备细胞悬液,调整细胞浓度为5×105/mL。取细胞悬液200 μL加入Transwell上室, 下室中加入500 μL的DMEM培养液,小心消除基质胶与培养液间的气泡。48 h后进行取样,吸净chamber上室内液体,无菌镊子取出chamber,移至800 μL甲醇溶液内,室温固定30 min。从甲醇溶液中取出chamber,吸干甲醇溶液,移至800 μL Giemsa染色液中,室温染色30 min,后用去离子水冲洗后吸干液体,湿棉棒擦去chamber上室未迁移细胞,PBS冲洗3遍。倒置显微镜下随机取9视野,并记数。

2.检测转染后细胞侵袭能力:用Matrigel基质胶包被Transwell小室基底膜,Matrigel胶4℃过夜融化备用,用4℃预冷的无血清DMEM培养基1∶3稀释Matrigel至终浓度为1 mg/mL,冰上操作;灭菌枪头包被小室的底部,37℃静置1 h,使其干成胶状。其余方法同迁移实验,并记数。

五、检测细胞转染后β-catenin、Vimentin及E-cadherin蛋白表达

收集三组转染48 h的细胞,实验分组包括空白对照组(HeLa)、空载组(GFP- HeLa)、HeLa-FOXC1(-)组,分别提取细胞总蛋白,Western blot检测 β-catenin、E-cadherin及Vimentin蛋白的表达差异,参照二-2中采用Western blot法检测β-catenin、Vimentin及E-cadherin蛋白相对表达量。

六、统计学处理:采用SPPS 17.0软件进行数据分析。计数资料比较采用χ2检验,计量资料用表示,采用单因素方差分析及两两比较的q检验。P<0.05为差异有统计学意义。

结 果

一、四种细胞中FOXC1 mRNA及蛋白表达

应用PCR及Western Blot方法检测正常宫颈细胞NC104及宫颈癌 CaSki、ME-180、HeLa细胞株中FOXC1表达。PCR及Western Blot结果提示宫颈癌HeLa细胞中FOXC1高表达。

二、慢病毒转染及筛选稳定转染FOXC1细胞

1.以MOI=30 慢病毒感染HeLa细胞系,48 h后荧光显微镜观察GFP荧光蛋白的表达情况见图2。并以嘌呤霉素筛选出稳定表达FOXC1 shRNA的细胞,流式细胞仪分选计数,其平均荧光细胞比率为(93.71±0.26)%。

图1 RT-PCR检测四组细胞中FOXC1 mRNA相对表达量及Western Blot检测四组细胞中FOXC1表达情况
Figure 1 The relative expression of FOXC1 mRNA detected by RT-PCR
and the expression of FOXC1 protein detected by Western blot

2.转染48 h后,PCR检测FOXC1mRNA的表达情况:各种的相对表达量见表1,其中转染FOXC1特异性shRNA组与阴性对照组、对照组相比较,FOXC1的mRNA相对表达量显著降低,差异具有统计学意义。

表1 FOXC1基因在各组中的表达情况
Table 1 The expression of FOXC1 gene
in three groups of

GroupRelative content of foxc1 mRNASh FOXC1 group0.1302±0.0398∗▲Negative control group1.0034±0.1450Control group 1.0106±0.0230

compared with the negative control group,*P<0.01;compared with the control group,P<0.01

A: Observation results of ordinary light microscope; B: Observation results of green fluorescence microscope;C: Fusion diagram of A and B
图2 荧光显微镜下观察HeLa-FOXC1(-)细胞系(放大倍数,×10倍)的荧光显示转染情况
Figure 2 The transfection of -FOXC1 (-) cell line
was observed under fluorescence microscope (magnification, ×10)

3.转染48 h后Western Blot检测转染后三组细胞中FOXC1蛋白水平表达:转染48 h后分别提取感染阴性组细胞HeLa、感染空载体组细胞HeLa-GFP以及感染FOXC1 shRNA组细胞HeLaFOXC1(-)的总蛋白,通过Western blot方法检测三种细胞中FOXC1蛋白的表达差异,GAPDH作为内参,结果可见HeLa-FOXC1(-)组细胞内FOXC1蛋白的表达明显降低(P<0.01)。见图3。

图3 三组细胞中FOXC1蛋白相对表达情况
Figure 3 The relative expression of FOXC1 protein in three groups

三、3组细胞Transwell迁移及侵袭实验结果

1.使用Transwell小室24 h后检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株的细胞迁移情况:迁移结果显示,敲减FOXC1基因后三组细胞迁移能力比较, HeLa-FOXC1(-)组细胞迁移能力明显下降(P<0.05),HeLa组与 HeLa-GFP组无明显差异。见图4。

图4 Transwell小室24 h后三组细胞迁移情况(×40)
Figure 4 Cell migration of crystal violet staining of shFOXC1 and control group cells that
crossed the polycarbonate membrane of the Transwell chamber (×40)

2.用Transwell小室24 h时分别检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株的细胞侵袭情况:Transwell侵袭实验结果显示,敲减FOXC1基因后三组细胞侵袭能力比较, HeLa-FOXC1(-)组细胞侵袭能力明显下降(P<0.05),HeLa组与 HeLa-GFP组无明显差异。见图5。

图5 Transwell小室24 h后三组细胞侵袭情况(×40)
Figure 5 Cell invasion of crystal violet staining of the shFOXC1 and control group
cells that crossed the Matrigel-coated polycarbonate membrane of the Transwell chamber to (×40)

四、Western blot检测感染前后各组细胞 β-catenin、E-cadherin及Vimentin蛋白的表达情况

由图6所示,A:使用WB法检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株中E-cadherin,β-catenin和vimentin的蛋白表达情况;B:使用WB法检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株中E-cadherin的蛋白表达情况统计结果(P<0.05);C:使用WB法检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株中β-catenin的蛋白表达情况统计结果(P<0.05);D:使用WB法检测HeLa细胞系,GFP稳定株和FOXC1的干扰稳定株中vimentin的蛋白表达情况统计结果(P<0.05)。结果可见,HeLa-FOXC1(-)组细胞内β-catenin及Vimentin蛋白的表达明显降低,E-cadherin蛋白的表达有增高。

图6 Western blot检测感染前后各组细胞中 β-catenin、E-cadherin
及Vimentin蛋白的表达情况及各组细胞中蛋白的相对表达量
Figure 6 The expression of protein and the relative expression
quantity ofβ-catenin, E-Cadhenrin and Vimentin detected by Western blot

讨 论

侵袭和转移是肿瘤临床治疗的难题及肿瘤患者死亡的主要原因。恶性肿瘤中上皮-间质转变现象的发生是引起肿瘤远处转移的关键步骤。EMT出现的分子标志为N-cadherin、Fibronectin与Vimentin这3类间质标志物表达的提升,β-Catenin与E-cadherin这2类上皮标志物表达的缺失或降低[12-14]。本课题组前期研究中发现,FOXC1基因在宫颈癌患者的临床病理中高表达,且随着期别进展,FOXC1基因的表达有明显增高,差异有统计学意义。本研究主要通过慢病毒RNA干扰技术,敲减宫颈癌HeLa细胞中FOXC1基因的表达,并通过倒置荧光显微镜及PCR、Western Blot方法验证在HeLa-FOXC1(-)细胞中,FOXC1基因的mRNA及蛋白水平明显降低,通过细胞培养技术获得稳定沉默FOXC1的细胞系,通过Transwell迁移及侵袭实验验证,感染FOXC1-shRNA组细胞的迁移及侵袭能力明显降低,进一步通过Western blot检测三组细胞中β-catenin、E-cadherin及Vimentin的表达,发现FOXC1敲除组细胞系内β-catenin及Vimentin蛋白的表达明显降低,E-cadherin蛋白的表达有增高,进一步验证敲除FOXC1基因的宫颈癌细胞后,下调上皮-间转化(EMT)过程中与转移有关蛋白(β-catenin、Vimentin)的表达,并增加钙黏蛋白(E-cadherin)的表达,使其侵袭及转移能力随之下降。由此可见,FOXC1和EMT在宫颈癌发展过程中有一定作用,其异常表达可能作为宫颈癌检查新的标志物。

宫颈癌是女性非常常见的妇科癌症之一,现全世界范围内每年仍有超过30万人因此病死亡[2],当前形势下其具体病因及发病机制仍不明确。临床上,宫颈癌的进展是从局部浸润、侵犯周围器官及淋巴结转移与远处转移,其特点是早期可发生淋巴结及远处转移。据报道,无淋巴转移的宫颈癌患者5年生存率为82.3%,有淋巴转移的5年生存率为50.8%[15]。因此,控制肿瘤的侵袭转移是提高宫颈癌患者生存率的主要方法之一[16-17]。肿瘤的基因靶向治疗是未来癌症治疗的主要手段,在不久的将来,相信FOXC1有望成为宫颈癌治疗的新靶点。

参考文献

1 Small W,Bacon MA,Bajaj A,et al.Cervicalcancer:A global health crisis.Cancer,2017,123:2404-2412.

2 Duenas-Gonzalez A,Campbell S.Global strategies for the treatment of early-stage and advancedcervical cancer.Curr Opin Obstet Gynecol,2016,28:11-17.

3 Thompson JC,Hwang WT,Davis C,et al.Gene signatures of tumor inflammation and epithelial-to-mesenchymal transition (EMT) predict responses to immune checkpoint blockade in lung cancer with high accuracy.Lung cancer,2020,1:131-139.

4 Li Yl,Wang T,Sun Y,et al.p53-Mediated PI3K/AKT/mTOR Pathway Played a Role in PtoxDpt-Induced EMT Inhibition in Liver Cancer Cell Lines.Oxid Med Cell Longev,2019,11:2531-2538.

5 Ou-Yang L,Xiao SJ,Liu P,et al.Forkhead box C1 induces epithelialmesenchymal transition and is a potential therapeutic target in nasopharyngeal carcinoma.Mol Med Rep,2015,12:8003-8009.

6 Han B,Qu Y,Jin Y,et al.FOXC1 activates smoothened-independent hedgehog signaling in basal-like breast cancer.Cell Rep,2015,13:1046-1058.

7 Xu ZY,Ding SM,Zhou L,et al.FOXC1 contributes to microvascular invasion in primary hepatocellular carcinoma via regulating epithelial-mesenchymal transition.Int J Biol Sci,2012,8:1130-1141.

8 Xia L,Huang W,Tian D,et al.Overexpression of forkhead box C1 promotes tumor metastasis and indicates poor prognosis in hepa-tocellular carcinoma.Hepatology,2013,57:610-624.

9 Wang Z,Qu L,Deng B,et al.STYK1 promotes epithelial-mesenchymal transition and tumor metastasis in human hepatocellular carcinoma through MEK/ERK and PI3K/AKT signaling.Sci Rep,2016,6:33205.

10 Song Y,Washington MK,Crawford HC.Loss of FOXA1/2 is essential for the epithelial-to-mesenchymal transition in pancre-atic cancer.Cancer Res,2010,70:2115-2125.

11 Ray PS,Wang J,Qu Y,et al.FOXC1 is a potential prog-nostic biomarker with functional significance in basal-like breast cancer.Cancer Res,2010,70:3870-3876.

12 Schmalhofer O,Brabletz S,Brabletz T.E-cadherin,beta-catenin,and ZEB1 in malignant progression of cancer.Cancer Metastasis Rev,2009,28:151-166.

13 Kowalski PJ,Rubin MA,Kleer CG.E-cadherin expression in primary carcinomas of the breast and its distant metastases.Breast Cancer Res,2003,5:217-222.

14 Lamouille S,Xu J,Derynck R.Molecular mechanisms of epithelial-mesenchymal transition.Nat Rev Mol Cell Biol,2014,15:178-196.

15 Li D,Cai J,Kuang Y,et al.Surgical-pathologic risk factors of pelvic lymph node metastasis in stage Ib1-IIb cervical cancer.Acta Obstet Gynecol Scand,2012,91:802-809.

16 KrillL S,Tewari KS.Exploring the Therapeutic Rationale for Angiogenesis Blockade in Cervical Cancer.Clin Ther,2015,37:9-19.

17 Tewari KS,Monk BJ.New strategies in advanced cervicalcancer:from angiogenesis blockade to immunotherapy.Clin Cancer Res,2014,20:5349-5358.

(收稿日期:2020-03-23)