程鑫宇,吳小榮,賈博深,沙建軍,陳勇輝,黃吉煒,張進(jìn),劉東明,黃翼然(上海交通大學(xué)醫(yī)學(xué)院附屬仁濟(jì)醫(yī)院,上海200001)
下調(diào)FoxM1基因表達(dá)對(duì)腎癌786-O細(xì)胞生物學(xué)行為的影響
程鑫宇,吳小榮,賈博深,沙建軍,陳勇輝,黃吉煒,張進(jìn),劉東明,黃翼然
(上海交通大學(xué)醫(yī)學(xué)院附屬仁濟(jì)醫(yī)院,上海200001)
摘要:目的觀察下調(diào)腎癌786-O細(xì)胞中叉頭框轉(zhuǎn)錄因子M1( FoxM1)基因的表達(dá)對(duì)腎癌細(xì)胞生物學(xué)行為的影響。方法采用FoxM1的干擾序列和短發(fā)夾RNA( shRNA)構(gòu)建shFoxM1質(zhì)粒。將786-O細(xì)胞分為shFoxM1組與對(duì)照組,分別轉(zhuǎn)染shFoxM1、scramble質(zhì)粒48 h。分別用Real-time PCR、Western blot法檢測(cè)細(xì)胞FoxM1 mRNA及蛋白,平板克隆形成實(shí)驗(yàn)觀察1周細(xì)胞克隆形成率,采用流式細(xì)胞儀檢測(cè)細(xì)胞周期及細(xì)胞凋亡率,Transwell實(shí)驗(yàn)觀察細(xì)胞遷移和侵襲能力,MTT法檢測(cè)細(xì)胞培養(yǎng)0、24、48、72 h時(shí)的細(xì)胞吸光度值。結(jié)果與對(duì)照組比較,shFoxM1組FoxM1 mRNA和蛋白表達(dá)減少,細(xì)胞克隆形成率、細(xì)胞吸光度值降底,G1期細(xì)胞比例增加而G2、S期減少,早期凋亡率增加,遷移、侵襲的穿膜細(xì)胞減少,P均<0.05。結(jié)論下調(diào)FoxM1基因表達(dá),可抑制腎癌786-O細(xì)胞的增殖、遷移、侵襲能力,使細(xì)胞阻滯于G1期,促進(jìn)細(xì)胞的早期凋亡。
關(guān)鍵詞:叉頭框轉(zhuǎn)錄因子M1;腎腫瘤;透明細(xì)胞癌;細(xì)胞增殖;細(xì)胞凋亡;核糖核酸干擾技術(shù)
Effect of down-regulating FoxM1 expression on biological behavior of renal cancer 786-O cells
CHENG Xin-yu,WU Xiao-rong,JIA Bo-shen,SHA Jian-jun,CHEN Yong-hui,HUANG Ji-wei,ZHANG Jin,LIU Dong-ming,HUANG Yi-ran
( Renji Hospital,Shanghai Jiaotong University School of Medicine,Shanghai 200001,China)
Abstract:Objective To observe the down-regulation of forkhead box protein M1 ( FoxM1) gene expression and to investigate its effect on the biological behavior of human renal cancer cell line 786-O.Methods The shFoxM1 plasmid was constructed by RNA interference sequence and shRNA.786-O cells were divided into the shFoxM1 group and the control group which were transfected with shFoxM1 plasmid and scramble plasmid for 48 h,respectively.The expression of FoxM1 mRNA and protein was tested by real-time PCR and Western blotting.Then we analyzed the efficiency of plate colony formation in one week by using colony formation assay,and we used flow cytometry to detect the cell cycle and apoptosis of 786-O cells.The migration and invasion abilities were analyzed by Transwell assay,and the OD value in 0,24,48 and 72 hours was tested using MTT assay to analyze the activity of proliferation.Results Compared with the control group,the FoxM1 mRNA and protein expression was decreased,the efficiency of plate colony formation and the cell proliferation were decreased,the cells were blocked in G1phase,cells of G2phase and S phase reduced,the early apoptosis rate rose and the migration and invasion cells were decreased in the shFoxM1 group ( all P<0.05).Conclusions Down-regulating FoxM1 gene expression suppressed the proliferation,migration and invasion abilities of 786-O cells,blocking cells in G1phase and promoting early apoptosis.
Key words:forkhead box protein M1; kidney neoplasms; clear cell carcinoma; cell proliferation; apoptosis; RNA interference technology
腎癌是常見(jiàn)的泌尿系統(tǒng)腫瘤,近年來(lái)發(fā)病率和病死率持續(xù)上升[1,2],腎透明細(xì)胞癌( ccRCC)是其主要病理類型[3]。叉頭框轉(zhuǎn)錄因子M1( FoxM1)是一種增殖性轉(zhuǎn)錄因子,與多種腫瘤的發(fā)生、發(fā)展密切相關(guān)[4,5];其可通過(guò)對(duì)多種靶蛋白的轉(zhuǎn)錄激活作用影響腫瘤細(xì)胞的生物學(xué)行為,如細(xì)胞的增殖、凋亡、遷移、侵襲等[6]。2015年1~4月,我們構(gòu)建了靶向FoxM1基因的短發(fā)夾RNA( shRNA),并將其轉(zhuǎn)染人腎癌786-O細(xì)胞,觀察下調(diào)FoxM1表達(dá)對(duì)腎癌細(xì)胞生物學(xué)行為的影響?,F(xiàn)報(bào)告如下。
1.1材料人ccRCC細(xì)胞株786-O為仁濟(jì)醫(yī)院泌尿外科自有;轉(zhuǎn)染試劑X-tremeGENE HP( Roche,Switzerland) ; Opti-MEM培養(yǎng)基、RPMI-1640培養(yǎng)基、胎牛血清( FBS)、胰酶、TRIzol ( Life technologies,USA) ;逆轉(zhuǎn)錄試劑盒、SYBR Green I kit ( TaKaRa,Japan) ;兔抗人FoxM1抗體( Santa Cruz,USA) ;二甲基亞砜( DMSO)、牛血清白蛋白( BSA) ;四甲基偶氮唑鹽( MTT)、碘化丙啶( PI)、鼠抗人GAPDH抗體( Sigma-Aldrich,USA) ;辣根過(guò)氧化物酶( HRP)標(biāo)記的抗兔、抗鼠二抗( DAKO,USA) ;總蛋白提取試劑盒(碧云天,中國(guó)) ; Matrigel膠、Transwell小室及Annexin V-FITC/PI凋亡試劑盒( BD,USA) ;限制性內(nèi)切酶BamHⅠ、HindⅢ、Taq DNA聚合酶、T4 DNA連接酶( NEB,USA) ; pRFP-C-RS質(zhì)粒、scramble質(zhì)粒( OriGene,USA)。
1.2 shFoxM1構(gòu)建根據(jù)Genbank設(shè)計(jì)針對(duì)FoxM1的干擾序列,由Invitrogen中國(guó)公司合成,經(jīng)Blast檢測(cè)不與其他人類基因互補(bǔ);頸環(huán)結(jié)構(gòu)為T(mén)CAAGAG,5'及3'端引入BamHⅠ、HindⅢ酶切位點(diǎn);正義鏈為5'-GATCGCTTGCAGCCAATCGTTCTCT GACAGAAGGTCAAGAGGAACGTCGGTTAGCAAGAGACTGTCTTCCTTTTTTGA-3',反義鏈為5'-AGCTTCAAAAAACTTGCAGCCAATCGTTCTCTGACAGAAGGCTCTTGAGAACGTCGGTTAGCAAGAGACTGTCTTCCC-3',經(jīng)上海鉑尚生物技術(shù)有限公司測(cè)序證實(shí)。
1.3細(xì)胞分組與轉(zhuǎn)染將786-O細(xì)胞于含10% FBS的RPMI-1640培養(yǎng)基中培養(yǎng),分為shFoxM1組、對(duì)照組,每組約5×106個(gè)細(xì)胞。待各組細(xì)胞生長(zhǎng)融合至70%時(shí),將shFoxM1組、對(duì)照組按照X-tremeGENE HP說(shuō)明書(shū)[7]分別轉(zhuǎn)染shFoxM1、scramble質(zhì)粒(陰性對(duì)照),轉(zhuǎn)染后將細(xì)胞置于含10% FBS的RPMI-1640培養(yǎng)基,于37℃恒溫細(xì)胞培養(yǎng)箱培養(yǎng)48 h。以熒光顯微鏡下觀察到>15%的細(xì)胞被紅色熒光標(biāo)記,為轉(zhuǎn)染成功。
1.4相關(guān)指標(biāo)觀察
1.4.1 FoxM1 mRNA表達(dá)檢測(cè)采用Real-time PCR法。根據(jù)PrimerBank設(shè)計(jì)FoxM1基因PCR引物,由上海鉑尚生物技術(shù)公司合成。FoxM1上游引物: 5'-GGAGCAGCGACAGGTTAAGG-3',下游引物: 5'-GTTGATGGCGAATTGTATCATGG-3'; GAPDH:上游引物5'-GGAGCGAGATCCCTCCAAAAT-3',下游引物5'-GGCTGTTGTCATACTTCTCATGG-3'。反應(yīng)條件: 95℃10 min,95℃15 s,60℃退火1 min,共40個(gè)循環(huán)。采用相對(duì)定量法計(jì)算2-ΔΔCt值,以此表示FoxM1 mRNA相對(duì)表達(dá)量。
1.4.2 FoxM1蛋白表達(dá)檢測(cè)采用Western blot法,檢測(cè)各組FoxM1蛋白表達(dá)。所有操作均嚴(yán)格按照使用說(shuō)明書(shū)進(jìn)行,實(shí)驗(yàn)重復(fù)3次,以目的蛋白與β-actin電泳條帶吸光度比值表示FoxM1蛋白表達(dá)量。
1.4.3細(xì)胞增殖觀察采用MTT法。取各組細(xì)胞以2×103/孔接種于96孔板培養(yǎng),每組12個(gè)復(fù)孔。于培養(yǎng)0、24、48、72 h時(shí)各取3個(gè)復(fù)孔,棄去原培養(yǎng)液后每孔加入含0.5 mg/mL MTT的新鮮培養(yǎng)液100 μL,37℃孵育4 h ;吸去培養(yǎng)液,加入DMSO 100 μL/孔,搖床輕搖15 min;待紫色結(jié)晶充分溶解后,在酶標(biāo)儀上570 nm處測(cè)量并記錄每孔吸光度值,取每組平均值。
1.4.4細(xì)胞克隆形成觀察取各組細(xì)胞,胰酶消化后充分吹打至細(xì)胞分散;接種于6孔板中,5× 102/孔;每組3個(gè)復(fù)孔,置于37℃含5% CO2的恒溫細(xì)胞培養(yǎng)箱中培養(yǎng)1周。用70%乙醇固定,0.5%結(jié)晶紫染色10 min;將6孔板倒置并疊加一張帶網(wǎng)格的透明膠片,肉眼計(jì)數(shù)各孔克隆數(shù)目,取平均值??寺⌒纬陕?克隆數(shù)/接種細(xì)胞數(shù)×100%。
1.4.5細(xì)胞周期和凋亡觀察①細(xì)胞周期檢測(cè):取各組細(xì)胞,70%乙醇4℃固定30 min; PBS洗滌,用500 μL PBS重懸;加入2.5 mg/mL PI 10 μL及1 mg/mL RNA酶10 μL,37℃培養(yǎng)箱孵育30 min;上BD FACS calibur流式細(xì)胞儀,測(cè)算G1期、G2期和S期細(xì)胞比例。②細(xì)胞凋亡檢測(cè):取各組細(xì)胞,重懸后室溫避光條件下分別予Annexin V/FITC及PI染色15 min;上BD FACS calibur流式細(xì)胞儀,計(jì)算早期及晚期細(xì)胞凋亡率并取平均值。
1.4.6細(xì)胞遷移及侵襲觀察取各組細(xì)胞,經(jīng)無(wú)血清RMPI-1640培養(yǎng)基饑餓處理2 h;用含0.1% BSA的無(wú)血清RMPI-1640培養(yǎng)基重懸,至細(xì)胞密度為1× 106/mL(遷移)或5×105/mL(侵襲)。將Transwell小室置于24孔板中,完成基底膠包被過(guò)程(遷移實(shí)驗(yàn)無(wú)
需此步驟) ;上室加200 μL細(xì)胞懸液,下室加500 μL 含10%FBS的新鮮培養(yǎng)基,放于培養(yǎng)箱中培養(yǎng)。6 h后取出小室,棉簽擦掉上室面細(xì)胞; PBS輕洗2遍,倒置風(fēng)干;70%乙醇固定,0.1%( w/v)結(jié)晶紫染下室面15 min,PBS輕洗1遍后風(fēng)干。顯微鏡下計(jì)數(shù)5個(gè)視野的細(xì)胞,以平均數(shù)表示遷移或侵襲能力。
1.5統(tǒng)計(jì)學(xué)方法采用SPSS18.0統(tǒng)計(jì)軟件。計(jì)量資料以珋x±s表示,組間比較采用單因素方差分析及t檢驗(yàn)。P<0.05為差異有統(tǒng)計(jì)學(xué)意義。
2.1兩組FoxM1 mRNA及蛋白表達(dá)比較見(jiàn)表1。
表1 兩組FoxM1 mRNA及蛋白表達(dá)比較(±s)
表1 兩組FoxM1 mRNA及蛋白表達(dá)比較(±s)
注:與對(duì)照組比較,*P<0.05。
組別FoxM1 mRNA 蛋白shFoxM1組 20.57±5.35* 25.33±5.60*對(duì)照組100.00±6.48 100.00±7.37
2.2兩組細(xì)胞增殖情況比較見(jiàn)表2。
表2 兩組細(xì)胞增殖情況比較(±s)
表2 兩組細(xì)胞增殖情況比較(±s)
注:與對(duì)照組同時(shí)點(diǎn)比較,*P<0.05。
組別吸光度值0 h 24 h 48 h 72 h shFoxM1組 0.20±0.01 0.23±0.04 0.28±0.06*0.45±0.04*對(duì)照組0.20±0.04 0.25±0.01 0.39±0.05 0.62±0.03
2.3兩組細(xì)胞克隆形成率比較接種后1周時(shí),shFoxM1組、對(duì)照組細(xì)胞克隆形成率分別為7%、18%,P<0.05。
2.4兩組細(xì)胞周期和凋亡率比較見(jiàn)表3。
2.5兩組細(xì)胞遷移及侵襲情況比較遷移實(shí)驗(yàn)中shFoxM1組、對(duì)照組穿膜細(xì)胞分別為( 240.67± 96.32)、( 989.33±100.47)個(gè),P<0.05;侵襲實(shí)驗(yàn)中shFoxM1組、對(duì)照組穿膜細(xì)胞分別為( 20.30± 7.41)、( 83.00±5.86)個(gè),P<0.05。
表3 兩組細(xì)胞周期和凋亡率比較( %,±s)
表3 兩組細(xì)胞周期和凋亡率比較( %,±s)
注:與對(duì)照組同時(shí)點(diǎn)比較,*P<0.05。
組別細(xì)胞周期G1期 G2期 S期凋亡率早期 晚期shFoxM1組 47.94±3.23* 2.76±1.43* 49.30±2.98* 12.33±1.50*9.29±2.00對(duì)照組 34.16±3.34 8.96±4.33 56.88±2.56 1.40±0.50 8.35±1.10
腎癌占全身惡性腫瘤的2%~3%[8],根治性手術(shù)為其主要治療方法;但早期局限性腎癌術(shù)后仍有30%可能發(fā)生遠(yuǎn)處轉(zhuǎn)移[9],預(yù)后較差。腎癌對(duì)放療、化療及免疫治療均不敏感,治療有效率<20%[10]。
FoxM1是一類增殖性轉(zhuǎn)錄因子,廣泛表達(dá)于哺乳動(dòng)物組織中,與細(xì)胞增殖、更新、分化、遷移、有絲分裂及DNA損傷修復(fù)等[11]多種細(xì)胞生物學(xué)功能密切相關(guān)。其在肝癌、肺癌、前列腺癌等多種腫瘤中高表達(dá)。全基因組分析顯示,其可能是惡性腫瘤中表達(dá)上調(diào)頻率最高的基因之一[12],且腫瘤發(fā)生早期即出現(xiàn)高表達(dá)[13],提示其可能是促使腫瘤發(fā)生及進(jìn)展的關(guān)鍵因子之一。我們前期研究顯示,與正常腎組織相比,腎癌組織中FoxM1蛋白表達(dá)明顯增高,且與患者預(yù)后有關(guān),提示其可能與腎癌發(fā)生、發(fā)展有密切關(guān)聯(lián)[14]。
Janus等[15]研究發(fā)現(xiàn),對(duì)有絲分裂過(guò)程中紡錘體正確裝配起重要作用的中心體蛋白CEP55可能是FoxM1下游靶基因,可能是FoxM1促進(jìn)腫瘤細(xì)胞增殖的機(jī)制之一。本研究發(fā)現(xiàn),使用shRNA下調(diào)FoxM1表達(dá)可明顯抑制786-O細(xì)胞的增殖能力及克隆形成能力。說(shuō)明FoxM1是促進(jìn)腎癌細(xì)胞增殖的重要因子之一,其機(jī)制可能與FoxM1促進(jìn)增殖相關(guān)細(xì)胞因子的轉(zhuǎn)錄有關(guān)。本研究同時(shí)發(fā)現(xiàn),shFoxM1組處于G1期細(xì)胞比例明顯增加,S期及G2期細(xì)胞比例減少,說(shuō)明抑制FoxM1的表達(dá)可使786-O細(xì)胞發(fā)生G1期阻滯。FoxM1作為重要的轉(zhuǎn)錄因子,可調(diào)控Cyclin D1、Cyclin B1等周期相關(guān)因子的轉(zhuǎn)錄;抑制FoxM1表達(dá)后下調(diào)相關(guān)因子水平,從而阻斷正常細(xì)胞周期。同時(shí),流式細(xì)胞術(shù)檢測(cè)表明抑制FoxM1表達(dá)可明顯促進(jìn)凋亡特別是早期凋亡,提示FoxM1在腎癌細(xì)胞中具有抗凋亡作用。有研究表明,干擾FoxM1表達(dá)后,Caspase-3活化增多[16]; Kalin等[17]發(fā)現(xiàn),干擾前列腺癌細(xì)胞中FoxM1表達(dá),可下調(diào)Bcl-2表達(dá)。因此,F(xiàn)oxM1可能通過(guò)參與對(duì)細(xì)胞Bcl-2、Bax、Caspase-3等多種凋亡相關(guān)分子的轉(zhuǎn)錄調(diào)控發(fā)揮其抗凋亡作用。Wang等[18]研究表明,F(xiàn)oxM1可通過(guò)調(diào)控c-Jun氨基末端激酶1的轉(zhuǎn)錄促進(jìn)細(xì)胞侵襲,從而促進(jìn)腫瘤進(jìn)展。說(shuō)明FoxM1不僅在細(xì)胞增殖中具有重要作用,同時(shí)也參與了腫瘤侵襲轉(zhuǎn)移過(guò)程的調(diào)控?;|(zhì)金屬蛋白酶家族( MMPs)可通過(guò)降解細(xì)胞外基質(zhì)成分,在腫瘤侵襲及轉(zhuǎn)移過(guò)程中扮演關(guān)鍵角色,且在多種腫瘤組織中高表達(dá)[19]。FoxM1可激活MMPs成員如MMP2、MMP9表達(dá),Wang等[20]研究表明,F(xiàn)oxM1B轉(zhuǎn)基因小鼠中,MMP9呈過(guò)表達(dá)。Kong等[21]研究表明,在肺癌細(xì)胞中過(guò)表達(dá)
FoxM1可增強(qiáng)TGF-b1誘導(dǎo)的上皮間質(zhì)轉(zhuǎn)化能力,而EMT可促進(jìn)腫瘤細(xì)胞浸潤(rùn)及腫瘤轉(zhuǎn)移,敲除FoxM1則可逆轉(zhuǎn)其EMT作用。本研究結(jié)果顯示,抑制FoxM1表達(dá)后786-O細(xì)胞遷移能力與侵襲能力均明顯降低,可能與FoxM1對(duì)MMPs的激活作用及促使細(xì)胞表達(dá)間質(zhì)細(xì)胞標(biāo)志物從而增強(qiáng)EMT作用有關(guān),具體機(jī)制有待于進(jìn)一步研究。綜上所述,F(xiàn)oxM1的表達(dá)與腎癌細(xì)胞的生物學(xué)行為密切相關(guān),有望成為腎癌治療的新靶點(diǎn)。
參考文獻(xiàn):
[1]Siegel RL,Miller KD,Jemal A.Cancer statistics,2015[J].CA Cancer J Clin,2015,65( 1) : 5-29.
[2]Torre LA,Bray F,Siegel RL,et al.Global cancer statistics,2012 [J].CA Cancer J Clin,2015,65( 2) : 87-108.
[3]Kovacs G,Akhtar M,Beckwith BJ,et al.The Heidelberg classification of renal cell tumours[J].J Pathol,1997( 183) : 131-133.
[4]Zona S,Bella L,Burton MJ,et al.FOXM1: An emerging master regulator of DNA damage response and genotoxic agent resistance [J].Biochem Biophys Acta,2014,1839( 11) : 1316-1322.
[5]Chen T,Xiong J,Yang C,et al.Silencing of FOXM1 transcription factor expression by adenovirus-mediated RNA interference inhibits human hepatocellular carcinoma growth[J].Cancer Gene Ther,2014,21( 3) : 133-138.
[6]Yang C,Chen H,Tan G,et al.FOXM1 promotes the epithelial to mesenchymal transition by stimulating the transcription of Slug in human breast cancer[J].Cancer Lett,2013,340( 1) : 104-112.
[7]Han Y,Zhang Z,Shao C,et al.The expression of chicken selenoprotein W,selenocysteine-synthase ( SecS),and selenophosphate synthetase-1 ( SPS-1) in CHO-K1 cells[J].Biol Trace Elem Res,2012,148( 1) : 61-68.
[8]Rini BI,Campbell SC,Escudier B.Renal cell carcinoma[J].Lancet,2009,373( 9669) : 1119-1132.
[9]Jiang Z,Chu PG,Woda BA,et al.Combination of quantitative IMP3 and tumor stage: a new system to predict metastasis for patients with localized renal cell carcinomas[J].Clin Cancer Res,2008,14( 17) : 5579-5584.
[10]Su D,Stamatakis L,Singer EA,et al.Renal cell carcinoma: molecular biology and targeted therapy[J].Curr Opin Oncol,2014,26( 3) : 321-327.
[11]Wierstra I.FOXM1 ( Forkhead box M1) in tumorigenesis: overexpression in human cancer,implication in tumorigenesis,oncogenic functions,tumor-suppressive properties,and target of anticancer therapy[J].Adv Cancer Res,2012( 119) : 191-419.
[12]Pilarsky C,Wenzig M,Specht T,et al.Identification and validation of commonly overexpressed genes in solid tumors by comparison of microarray data[J].Neoplasia,2004,6( 6) : 744-750.
[13]Gemenetzidis E,Bose A,Riaz AM,et al.FOXM1 upregulation is an early event in human squamous cell carcinoma and it is enhanced by nicotine during malignant transformation[J].PLoS One,2009,4( 3) : 4849.
[14]Wu XR,Chen YH,Liu DM,et al.Increased expression of forkhead box M1 protein is associated with poor prognosis in clear cell renal cell carcinoma[J].Med Oncol,2013,30( 1) : 346.
[15]Janus JR,Laborde RR,Greenberg AJ,et al.Linking expression of FOXM1,CEP55 and HELLS to tumorigenesis in oropharyngeal squamous cell carcinoma[J].Laryngoscope,2011,121 ( 12) : 2598-2603.
[16]Wei P,Zhang N,Wang Y,et al.FOXM1 Promotes Lung Adenocarcinoma Invasion and Metastasis by Upregulating SNAIL[J].Int J Biol Sci,2015,11( 2) : 186.
[17]Kalin TV,Wang IC,Ackerson TJ,et al.Increased levels of the FoxM1 transcription factor accelerate development and progression of prostate carcinomas in both TRAMP and LADY transgenic mice [J].Cancer Res,2006,66( 3) : 1712-1720
[18]Wang IC,Chen YJ,Hughes DE,et al.FoxM1 regulates transcription of JNK1 to promote the G1/S transition and tumor cell invasiveness[J].J Biol Chem,2008,283( 30) : 20770-20778.
[19]Khokha R,Denhardt DT.Matrix metalloproteinases and tissue inhibitor of metalloproteinases: a review of their role in tumorigenesis and tissue invasion[J].Inv Meta,1988,9( 6) : 391-405.
[20]Wang X,Bhattacharyya D,Dennewitz MB,et al.Rapid hepatocyte nuclear translocation of the Forkhead Box M1B ( FoxM1B) transcription factor caused a transient increase in size of regenerating transgenic hepatocytes[J].Gene Expr,2003,11( 3-4) : 149-162.
[21]Kong FF,Zhu YL,Yuan HH,et al.FOXM1 Regulated by ERK Pathway MediatesTGF-β1-Induced EMT in NSCLC[J].Oncol Res,2014,22( 1) : 29-37.
收稿日期:( 2015-04-12)
通信作者簡(jiǎn)介:劉東明( 1961-),男,碩士,主任醫(yī)師,教授,碩士生導(dǎo)師,研究方向?yàn)槟I癌的基礎(chǔ)與臨床。E-mail: liudm541@126.com
作者簡(jiǎn)介:第一程鑫宇( 1990-),男,碩士,研究方向?yàn)槊谀蛳到y(tǒng)腫瘤的基礎(chǔ)與臨床。E-mail: jobberknoll90@163.com
基金項(xiàng)目:國(guó)家自然科學(xué)基金資助項(xiàng)目( 81402084) ;上海市衛(wèi)生局課題資助項(xiàng)目( 2012206)。
文章編號(hào):1002-266X( 2015) 30-0001-04
文獻(xiàn)標(biāo)志碼:A
中圖分類號(hào):R737.1
doi:10.3969/j.issn.1002-266X.2015.30.001