于偉娜,馮繼紅,丁陳波,高紹瑩,徐 林,袁建波,羅軍敏
(1.遵義醫(yī)學(xué)院免疫學(xué)教研室暨貴州省免疫學(xué)研究生教育創(chuàng)新基地,貴州 遵義 563099;2.遵義醫(yī)學(xué)院附院附屬腫瘤醫(yī)院,貴州 遵義 563099;3.遵義醫(yī)學(xué)院 醫(yī)學(xué)檢驗(yàn)系2011級(jí)學(xué)生,貴州 遵義 563099)
臨床醫(yī)學(xué)研究
PRDX2基因沉默促進(jìn)結(jié)直腸癌細(xì)胞上皮-間質(zhì)轉(zhuǎn)化增強(qiáng)侵襲轉(zhuǎn)移能力
于偉娜1,馮繼紅2,丁陳波1,高紹瑩1,徐 林1,袁建波3,羅軍敏1
(1.遵義醫(yī)學(xué)院免疫學(xué)教研室暨貴州省免疫學(xué)研究生教育創(chuàng)新基地,貴州 遵義 563099;2.遵義醫(yī)學(xué)院附院附屬腫瘤醫(yī)院,貴州 遵義 563099;3.遵義醫(yī)學(xué)院 醫(yī)學(xué)檢驗(yàn)系2011級(jí)學(xué)生,貴州 遵義 563099)
目的 探討Peroxiredoxin 2(PRDX2)基因沉默在結(jié)直腸癌細(xì)胞上皮-間質(zhì)轉(zhuǎn)化(EMT)中的作用及其參與侵襲轉(zhuǎn)移的可能機(jī)制。方法 將帶有抗性的對(duì)照shRNA(shRNA-Control)和PRDX2 shRNA(shRNA-PRDX2)慢病毒顆粒分別轉(zhuǎn)染結(jié)直腸癌SW480細(xì)胞,轉(zhuǎn)染后細(xì)胞分為PRDX2沉默組(shRNA-PRDX2)和空載對(duì)照組(shRNA-Control)。采用免疫印跡法(Western blot)和實(shí)時(shí)熒光定量PCR(qRT-PCR)法分別檢測(cè)兩組細(xì)胞中PRDX2蛋白和mRNA表達(dá);以5 ng/mL的TGF-β1作用兩組細(xì)胞,在0、48、72 h觀察兩組細(xì)胞形態(tài)學(xué)變化;劃痕實(shí)驗(yàn)和Transwell小室分別檢測(cè)兩組細(xì)胞體外遷移和侵襲能力;Western blot和qRT-PCR法檢測(cè)E-cadherin、Vimentin、Snail、Twist1和MMP-9蛋白及mRNA表達(dá)。結(jié)果 與對(duì)照組相比,PRDX2 沉默組細(xì)胞PRDX2蛋白和mRNA的表達(dá)顯著降低(P<0.05),成功建立PRDX2基因沉默穩(wěn)定細(xì)胞株;經(jīng)TGF-β1作用后,PRDX2沉默組細(xì)胞呈紡錘體樣間質(zhì)細(xì)胞表型,且遷移和侵襲能力明顯增強(qiáng);E-cadherin蛋白及mRNA水平表達(dá)顯著下降,Vimentin、Snail、Twist1和MMP-9蛋白及mRNA表達(dá)水平均明顯增加(P<0.05)。結(jié)論 PRDX2基因沉默可促進(jìn)結(jié)直腸癌SW480細(xì)胞發(fā)生EMT,從而增強(qiáng)細(xì)胞的侵襲和轉(zhuǎn)移能力,可能與其轉(zhuǎn)錄調(diào)節(jié)Snail、Twist1表達(dá)有關(guān)。
Peroxiredoxin 2;基因沉默;結(jié)直腸癌;上皮-間質(zhì)轉(zhuǎn)化;侵襲轉(zhuǎn)移
結(jié)直腸癌(colorectal cancer, CRC)是世界上最常見的惡性腫瘤之一,是導(dǎo)致癌癥相關(guān)死亡的第4大主要原因,僅次于肺癌、胃癌和肝癌[1]。最新GLOBOCAN數(shù)據(jù)顯示全球每年大約有120萬新確診病例,并且有超過60萬名患者死于CRC[2]。CRC在歐洲、北美和大洋洲較為多發(fā),其發(fā)病率約在45.7/10萬[3]。我國(guó)也處在CRC的相對(duì)較高發(fā)地區(qū),據(jù)統(tǒng)計(jì)分析其發(fā)病率為29.44/10萬,死亡率為14.23/10萬,且5年生存率為60%左右[4]。盡管目前外科手術(shù)、放化療可有效提高患者的生存率,但大部分患者最終還是死于轉(zhuǎn)移和局部復(fù)發(fā)。腫瘤轉(zhuǎn)移過程中上皮-間質(zhì)轉(zhuǎn)化(epithelial-mesenchymal transition,EMT)起到了關(guān)鍵性的作用,且腫瘤微環(huán)境中多種轉(zhuǎn)錄因子可參與調(diào)控EMT過程[5]。TGF-β1作為一個(gè)多功能的細(xì)胞因子,是目前已知的誘導(dǎo)腫瘤細(xì)胞發(fā)生EMT的關(guān)鍵細(xì)胞因子[6]。硫氧還蛋白過氧化物酶2(peroxiredoxin 2,PRDX2)是近年新發(fā)現(xiàn)的一類過氧化酶,廣泛存在于原核和真核生物中。研究發(fā)現(xiàn)PRDX2在多種腫瘤細(xì)胞及組織中高表達(dá),從而參與腫瘤的細(xì)胞增殖、凋亡以及侵襲轉(zhuǎn)移。我們前期研究發(fā)現(xiàn)PRDX2高表達(dá)于結(jié)直腸癌組織中,與淋巴結(jié)轉(zhuǎn)移密切相關(guān)[7];而且,過表達(dá)的PRDX2可抑制人結(jié)直腸癌細(xì)胞的遷移和EMT的發(fā)生[8],然而對(duì)于PRDX2基因沉默對(duì)人結(jié)直腸癌細(xì)胞SW480細(xì)胞EMT及轉(zhuǎn)移的影響卻鮮有報(bào)道。本研究前期利用TGF-β1成功誘導(dǎo)SW480細(xì)胞發(fā)生EMT[9],在此基礎(chǔ)上在SW480細(xì)胞內(nèi)利用PRDX2 shRNA慢病毒載體沉默PRDX2基因,觀察PRDX2基因沉默對(duì)細(xì)胞EMT過程和侵襲轉(zhuǎn)移影響及相關(guān)機(jī)制,為針對(duì)結(jié)直腸癌的靶向治療提供新的線索和契機(jī)。
1.1 材料 人結(jié)直腸癌Dukes B期細(xì)胞株SW480購自中科院上海細(xì)胞庫;PRDX2shRNA慢病毒載體及對(duì)照載體Control shRNA購自上海吉?jiǎng)P基因化學(xué)技術(shù)有限公司;胎牛血清(fetal bovine serum,F(xiàn)BS)購自杭州四季青公司;Leibovitz’s L-15(L-15)培養(yǎng)基購自Gibco公司;0.25%胰酶(含EDTA)購于Solarbio公司;單克隆兔抗人E-cadherin、Vimentin購自Abcam公司;兔抗人Twist1、Snail和MMP-9抗體購自Santa Cruz公司;嘌呤霉素(puromycin)購自Sigma公司;全蛋白提取試劑盒購自凱基生物科技有限公司;ECL試劑盒、鼠抗人β-actin、辣根過氧化物酶( HRP) 標(biāo)志的羊抗兔IgG二抗、DAPI染色液、SDS-PAGE凝膠配制試劑盒購自Beyotime公司;TGF-β1購自Peprotech公司;FastQuant cDNA第一鏈合成試劑盒購自天根生化科技有限公司;Trizol、SYBR?Premix Ex TaqTMII購自TaKaRa公司;PCR引物由上海生工生物工程有限公司合成。
1.2 方法
1.2.1 細(xì)胞培養(yǎng) SW480細(xì)胞在含10%FBS的L-15培養(yǎng)基中,37 ℃、5%CO2飽和濕度的培養(yǎng)箱培養(yǎng)。
1.2.2 細(xì)胞轉(zhuǎn)染與分組 取對(duì)數(shù)生長(zhǎng)期SW480細(xì)胞,以2×105/孔接種于6孔板中,次日根據(jù)SW480細(xì)胞MOI值計(jì)算所需PRDX2 shRNA及對(duì)照shRNA病毒顆粒數(shù),分別加入各孔中,培養(yǎng)28 h后更換嘌呤霉素篩選培養(yǎng)基(3 μg/mL),繼續(xù)抗性篩選培養(yǎng)3周建立穩(wěn)定轉(zhuǎn)染細(xì)胞株,通過Western blot和qRT-PCR對(duì)轉(zhuǎn)染效果進(jìn)行鑒定。實(shí)驗(yàn)設(shè)空載對(duì)照組(shRNA-Control)和PRDX2沉默組(shRNA-PRDX2)。
1.2.3 細(xì)胞形態(tài)學(xué)觀察 取兩組處于對(duì)數(shù)生長(zhǎng)期的細(xì)胞以2×105/孔密度接種于6孔板中,每組細(xì)胞設(shè)3復(fù)孔,待細(xì)胞貼壁后,更換含5ng/mL TGF-β1的L-15培養(yǎng)基,分別于0、48、72h在倒置相差顯微鏡下觀察細(xì)胞形態(tài)并拍照。
1.2.4 劃痕實(shí)驗(yàn) 將處于對(duì)數(shù)生長(zhǎng)期的兩組細(xì)胞以5×105/孔密度接種于6孔板中,待細(xì)胞單層鋪滿后,用10 μL槍頭分別在各組細(xì)胞上垂直劃痕,經(jīng)PBS洗滌后加入終濃度為5 ng/mL TGF-β1的含1% FBS的L-15培養(yǎng)基,分別于24、48、72 h 100倍倒置相差顯微鏡下觀察劃痕中細(xì)胞遷移情況并拍照,同時(shí)測(cè)量細(xì)胞劃痕間距。
1.2.5 Transwell侵襲實(shí)驗(yàn) 取50 μLMatrigel膠包被小室基底膜上室,置于超凈工作臺(tái)過夜風(fēng)干。使用前以100 μL/孔加入無血清L-15培養(yǎng)基,37 ℃孵育30 min使Matrigel膠重新水化。收集兩組細(xì)胞,以含0.1% BSA的無血清L-15培養(yǎng)基重懸,調(diào)整細(xì)胞濃度為2.5×105個(gè)/mL,取各組細(xì)胞200 μL加入各小室,下室加入500 μL含5 ng/mLTGF-β1的L-15培養(yǎng)基,常規(guī)培養(yǎng)48 h后,4%多聚甲醛固定,0.1%結(jié)晶紫染色,倒置相差顯微鏡下隨機(jī)選取5個(gè)視野拍照并計(jì)數(shù)。
1.2.6 Western blot檢測(cè) 收集各組細(xì)胞,PBS洗滌后提取總蛋白并測(cè)定其濃度,上樣,10%SDS- PAGE 凝膠電泳分離蛋白,電轉(zhuǎn)移至PVDF膜,5%脫脂奶粉室溫封閉1 h。分別加入一定稀釋比例的一抗,4 ℃孵育過夜,PBST漂洗3次,分別加入辣根過氧化物標(biāo)記的羊抗兔/鼠二抗,室溫孵育1 h,PBST漂洗3次,ECL顯色,自動(dòng)凝膠數(shù)字成像系統(tǒng)采集圖像。
1.2.7 qRT-PCR檢測(cè) 收集經(jīng)和未經(jīng)TGF-β1(5 ng/mL)誘導(dǎo)的shRNA-Control組和shRNA-PRDX2組細(xì)胞,使用Trizol分別提取各組細(xì)胞總RNA,檢測(cè)RNA濃度并將其逆轉(zhuǎn)錄為cDNA,隨后進(jìn)行PCR循環(huán)擴(kuò)增。PCR引物由上海生工生物工程有限公司合成,引物序列如表1。反應(yīng)條件為:95 ℃,5 min預(yù)變性;95 ℃,20 s;60 ℃,1 min,擴(kuò)增40個(gè)循環(huán)。2-ΔΔCT法計(jì)算mRNA的相對(duì)表達(dá)量,以GAPDH為內(nèi)參。
表1 qRT-PCR引物序列
目的基因上游引物/下游引物PRDX2E-cadherinVimentinSnailTwist1MMP-9GAPDH5'-CACCTGGCTTGGATCAACACC-3'5'-CAGCACGCCGTAATCCTCAG-3'5'-GCTCGGCCTGAAGTGACTCG-3'5'-CCGCTTCCTTCATAGTCAAACAC-3'5'-CCAGGCAAAGCAGGAGTCCAC-3'5'-GCTTCCTGTAGGTGGCAATCTC-3'5'-AGCCTGGGTGCCCTCAAG-3'5'-GAGCACACGCCTGGCACTG-3'5'-GTCCATGTCCGCGTCCCACATG-3'5'-ACGGGCCTGTCTCGCTTTCTC-3'5'-TGGTCCTGGTGCTCCTGGTG-3'5'-GCTGCCTGTCGGTGAGATTGG-3'5'-GAGCCAAACGGGTCATCATCT-3'5'-GAGGGGCCATCCACAGTCTT-3'
1.2.8 統(tǒng)計(jì)學(xué)方法 采用SPSS16.0軟件進(jìn)行統(tǒng)計(jì)分析,兩組間均數(shù)比較采用t檢驗(yàn),不同時(shí)段蛋白表達(dá)比較采用單因素方差分析,有差異者Dunnett’s test行兩兩比較;經(jīng)和未經(jīng)TGF-β1誘導(dǎo)的兩組細(xì)胞mRNA表達(dá)比較用兩因素析因設(shè)計(jì)資料方差分析。以P<0.05為差異有統(tǒng)計(jì)學(xué)意義。
2.1 PRDX2 shRNA慢病毒對(duì)SW480細(xì)胞PRDX2表達(dá)的影響 將慢病毒載體shRNA-Control和shRNA-PRDX2轉(zhuǎn)染SW480細(xì)胞后,Western blot和qRT-PCR法檢測(cè)細(xì)胞內(nèi)PRDX2蛋白和mRNA表達(dá)水平。結(jié)果顯示,與對(duì)照組相比,PRDX2沉默組細(xì)胞PRDX2蛋白和mRNA表達(dá)水平均顯著降低(P<0.05,見圖1~2)。
圖1 PRDX2基因沉默后SW480細(xì)胞中PRDX2蛋白表達(dá)
圖2 PRDX2沉默后SW480細(xì)胞中PRDX2 mRNA表達(dá)
2.2 細(xì)胞形態(tài)學(xué)觀察 與對(duì)照組相比,隨著時(shí)間的變化, shRNA-PRDX2組細(xì)胞形態(tài)由鵝卵石樣逐漸轉(zhuǎn)變?yōu)榧忓N體樣間質(zhì)細(xì)胞形態(tài),且細(xì)胞間連接也逐漸消失,細(xì)胞變的松散,突起亦隨之增多(見圖3)。
2.3 PRDX2沉默后在TGF-β1作用下對(duì)細(xì)胞遷移能力的影響 shRNA-Control組和shRNA-PRDX2組細(xì)胞在5ng/mLTGF-β1誘導(dǎo)下,隨著時(shí)間的推移,兩組細(xì)胞的劃痕間距均明顯變窄,但在TGF-β1作用72 h后shRNA-PRDX2組細(xì)胞劃痕間距明顯低于shRNA-Control組,差異有統(tǒng)計(jì)學(xué)意義(P<0.05,見圖4)。
圖3 PRDX2沉默后在TGF-β1誘導(dǎo)下對(duì)SW480細(xì)胞形態(tài)學(xué)影響(×200)
圖4 PRDX2沉默對(duì)TGF-β1作用后細(xì)胞遷移能力的影響(×100)
2.4 PRDX2沉默對(duì)TGF-β1誘導(dǎo)后SW480細(xì)胞侵襲能力的影響 侵襲實(shí)驗(yàn)結(jié)果顯示,在5ng/mLTGF-β1誘導(dǎo)下shRNA-PRDX2組穿過人工基底膠細(xì)胞數(shù)明顯多于shRNA-Control組(P<0.05,見圖5)。
A:shRNA-Control組;B:shRNA-ARDX2組。
2.5 Western blot檢測(cè)各組細(xì)胞中E-cadherin、Vimentin、Snail、Twist1和MMP-9蛋白的表達(dá) 在PRDX2基因沉默后,EMT標(biāo)志分子E-cadherin的表達(dá)明顯降低,而Vimentin、Snail、Twist1和MMP-9表達(dá)則明顯升高(P<0.05);隨著TGF-β1誘導(dǎo)時(shí)間的延長(zhǎng),shRNA-PRDX2組細(xì)胞內(nèi)E-cadherin蛋白表達(dá)水平明顯低于對(duì)照組,Vimentin、Snail、Twist1和MMP-9表達(dá)較對(duì)照組顯著增加(P<0.05,見圖6)。
圖6 Western blot檢測(cè)各組細(xì)胞E-cadherin、Vimentin、Snail、Twist1及MMP-9蛋白表達(dá)
2.6 qRT-PCR檢測(cè)各組細(xì)胞中E-cadherin、Vimentin、Snail、Twist1和MMP-9 mRNA表達(dá)情況 結(jié)果顯示在PRDX2沉默后,shRNA-PRDX2組E-cadherin mRNA表達(dá)量明顯降低,同時(shí)Vimentin、Snail、Twist1和MMP-9表達(dá)升高(P<0.05);且在5ng/mLTGF-β1誘導(dǎo)后,shRNA-PRDX2組細(xì)胞內(nèi)E-cadherin mRNA水平明顯低于shRNA-Control組,同時(shí)Vimentin、Snail、Twist1和MMP-9 mRNA表達(dá)顯著增加(P<0.05,見圖7)。
主效應(yīng)F值=11.82,交互效應(yīng)F值=27.74,P值均<0.05。
結(jié)直腸癌是一種上皮細(xì)胞來源的惡性腫瘤,早期較易發(fā)生轉(zhuǎn)移,Enquist等[10]通過建立小鼠模型研究發(fā)現(xiàn)結(jié)直腸癌腫瘤細(xì)胞能夠在周圍淋巴結(jié)蔓延之前直接轉(zhuǎn)移到肝臟。肝轉(zhuǎn)移是結(jié)直腸癌的主要轉(zhuǎn)移途徑,同時(shí)也是結(jié)直腸癌患者死亡的主要原因之一,因此探討結(jié)直腸癌侵襲轉(zhuǎn)移機(jī)制對(duì)其臨床治療與預(yù)后具有重要意義。
PRDX2作為PRDXs家族的一個(gè)重要成員,是一種重要的細(xì)胞內(nèi)抗氧化劑。RRDX2可參與細(xì)胞的抗氧化防御系統(tǒng),能有效的對(duì)抗氧化應(yīng)激引起的細(xì)胞損傷,從而有利于腫瘤細(xì)胞的增殖和存活。近期研究發(fā)現(xiàn)PRDX2在腫瘤的侵襲轉(zhuǎn)移過程中也發(fā)揮著重要的作用,Agrawal-Singh等[11]證實(shí)急性髓細(xì)胞性白血病細(xì)胞中PRDX2蛋白表達(dá)與臨床不良預(yù)后以及轉(zhuǎn)移呈負(fù)相關(guān)關(guān)系。另外,PRDX2的過度表達(dá)可通過增加E-cadherin/β-catenin復(fù)合物的形成從而抑制黑色素瘤的進(jìn)展和轉(zhuǎn)移[12]。本研究在TGF-β1誘導(dǎo)結(jié)直腸癌細(xì)胞產(chǎn)生EMT表型改變的前期基礎(chǔ)之上,將shRNA慢病毒載體轉(zhuǎn)染結(jié)直腸癌細(xì)胞使PRDX2基因沉默,觀察PRDX2基因在結(jié)直腸癌細(xì)胞EMT及侵襲轉(zhuǎn)移過程中的作用。結(jié)果發(fā)現(xiàn),PRDX2基因沉默后SW480細(xì)胞發(fā)生了顯著的生物學(xué)行為變化,同時(shí)EMT標(biāo)志分子也有相應(yīng)改變。倒置相差顯微鏡下觀察細(xì)胞形態(tài)顯示,PRDX2沉默后細(xì)胞逐漸由上皮形態(tài)向間質(zhì)形態(tài)轉(zhuǎn)變,細(xì)胞遷移與侵襲能力也明顯增加;且經(jīng)TGF-β1誘導(dǎo)后,PRDX2基因沉默組細(xì)胞呈典型間質(zhì)細(xì)胞形態(tài),細(xì)胞散在分布、突起增多,且細(xì)胞遷移、侵襲能力均顯著增強(qiáng),表明PRDX2基因沉默促進(jìn)了結(jié)直腸癌細(xì)胞EMT過程并增強(qiáng)細(xì)胞的侵襲轉(zhuǎn)移。
在腫瘤侵襲轉(zhuǎn)移過程中,EMT起著關(guān)鍵性的作用。EMT可使上皮細(xì)胞轉(zhuǎn)化為間質(zhì)細(xì)胞狀態(tài),并發(fā)生一系列表型和分子的改變,促使細(xì)胞外基質(zhì)成分產(chǎn)生降解,從而增強(qiáng)腫瘤細(xì)胞的遷移、侵襲能力。經(jīng)歷EMT的腫瘤細(xì)胞在到達(dá)合適的生長(zhǎng)部位后又可經(jīng)過間質(zhì)-上皮轉(zhuǎn)化(MET)重新獲得上皮細(xì)胞特性進(jìn)而增殖形成新的轉(zhuǎn)移病灶。上皮細(xì)胞標(biāo)記分子E-鈣粘蛋白(E-cadherin)介導(dǎo)粘附的丟失常被認(rèn)為是腫瘤細(xì)胞侵襲和轉(zhuǎn)移的一個(gè)先決條件,且已有研究證實(shí)E-cadherin表達(dá)下調(diào)或缺失與EMT的發(fā)生密切相關(guān),并能夠促進(jìn)腫瘤的侵襲與轉(zhuǎn)移[13]。在腫瘤發(fā)生發(fā)展過程中腫瘤微環(huán)境中多種轉(zhuǎn)錄因子可參與調(diào)控EMT過程,如Snail、Twist1等。這些轉(zhuǎn)錄因子作為細(xì)胞間粘附分子E-cadherin的負(fù)性調(diào)控因子,可以競(jìng)爭(zhēng)性的結(jié)合E-cadherin的啟動(dòng)子區(qū)域,在發(fā)揮對(duì)EMT的誘導(dǎo)效應(yīng)中起到關(guān)鍵性作用[14]。Snail是一類鋅指轉(zhuǎn)錄因子,研究表明Snail可抑制E-cadherin表達(dá)從而誘導(dǎo)EMT發(fā)生,與乳腺癌的侵襲轉(zhuǎn)移密切相關(guān)[15]。堿性螺旋-環(huán)-螺旋(basic helix-loop-helix,bHLH)轉(zhuǎn)錄因子Twist1在胚胎發(fā)育和腫瘤進(jìn)展中均發(fā)揮重要調(diào)控作用,且其介導(dǎo)的EMT可能參與卵巢癌的浸潤(rùn)[16-17]。有研究證實(shí)肺癌中異常表達(dá)的Snail、Twist1可以直接抑制E-cadherin的轉(zhuǎn)錄和翻譯,從而誘使腫瘤細(xì)胞發(fā)生EMT[18]。本研究發(fā)現(xiàn)在PRDX2基因沉默后,EMT標(biāo)志分子E-cadherin的表達(dá)顯著降低,Vimentin、Snail、Twist1表達(dá)則明顯升高;且在TGF-β1作用下,較對(duì)照空載組E-cadherin表達(dá)明顯降低,同時(shí)Vimentin、Snail、Twist1表達(dá)顯著增加,表明下調(diào)PRDX2的表達(dá)可促進(jìn)結(jié)直腸癌SW480細(xì)胞EMT的發(fā)生,并且可能是通過轉(zhuǎn)錄調(diào)節(jié)Snail和Twist1來實(shí)現(xiàn)的。侵襲和轉(zhuǎn)移是惡性腫瘤細(xì)胞的一個(gè)重要特征,細(xì)胞外基質(zhì)(ECM)是腫瘤細(xì)胞轉(zhuǎn)移的天然屏障。基質(zhì)金屬蛋白酶(matrix metalloproteinases,MMPs)是一類重要的蛋白水解酶,可由腫瘤細(xì)胞、內(nèi)皮細(xì)胞等合成和分泌,能有效的降解ECM及基膜,在腫瘤的侵襲轉(zhuǎn)移過程中占有重要角色[19]。MMP-9是MMPs中降解ECM的重要蛋白水解酶之一,其表達(dá)增加與腫瘤的浸潤(rùn)轉(zhuǎn)移極為相關(guān)。MMP-9在多種腫瘤細(xì)胞、腫瘤組織中表達(dá)增強(qiáng),且腫瘤細(xì)胞的高侵襲能力均與MMP-9的異常高表達(dá)有關(guān)[20-22]。同樣,本研究也發(fā)現(xiàn)在PRDX2基因沉默后,無論TGF-β1誘導(dǎo)與否SW480細(xì)胞中MMP-9表達(dá)量均明顯升高,且細(xì)胞的遷移、侵襲能力也顯著增加,進(jìn)一步說明PRDX2參與了結(jié)直腸癌細(xì)胞的侵襲和轉(zhuǎn)移過程。目前研究發(fā)現(xiàn)腫瘤細(xì)胞中過表達(dá)的PRDX2可增強(qiáng)細(xì)胞的DNA修復(fù)能力,進(jìn)而增加腫瘤細(xì)胞的化學(xué)抗性,促進(jìn)腫瘤的進(jìn)展[23-24]。對(duì)于這些不同的結(jié)果目前尚未有合理的解釋,猜測(cè)PRDX2對(duì)于不同病理類型的腫瘤細(xì)胞可能存在著不同的作用,同樣即使在同一腫瘤中其作用也可能截然不同。Shiota等[25]報(bào)道PRDX2在前列腺癌細(xì)胞的胞質(zhì)和胞核中存在不同的作用,細(xì)胞質(zhì)中過度表達(dá)的PRDX2可促進(jìn)雄激素受體的激活,而在細(xì)胞核中的作用則反之。前期研究發(fā)現(xiàn)PRDX2高表達(dá)于CRC LoVo細(xì)胞株,且高表達(dá)的PRDX2能有效的抵抗H2O2誘導(dǎo)的細(xì)胞凋亡;且在SW480細(xì)胞的研究中初步發(fā)現(xiàn)PRDX2可抑制結(jié)直腸癌細(xì)胞的EMT進(jìn)程和轉(zhuǎn)移過程,并提示可促進(jìn)MET過程的發(fā)生[8,26]。由此可見,PRDX2對(duì)腫瘤的發(fā)生發(fā)展具有相當(dāng)復(fù)雜而嚴(yán)格的調(diào)控機(jī)制,可能在同一腫瘤中同時(shí)發(fā)揮致癌基因和抑癌基因的雙重角色。因此,PRDX2調(diào)控結(jié)直腸癌細(xì)胞EMT和轉(zhuǎn)移過程的作用機(jī)制仍需進(jìn)一步研究探討。
綜上所述,PRDX2基因沉默促進(jìn)TGF-β1誘導(dǎo)的結(jié)直腸癌SW480細(xì)胞發(fā)生EMT,從而增強(qiáng)了細(xì)胞的侵襲和轉(zhuǎn)移,同時(shí)EMT標(biāo)志分子E-cadherin、Vimentin以及相關(guān)轉(zhuǎn)錄因子Snail、Twist1也發(fā)生相應(yīng)改變。由此我們可以看出PRDX2基因與結(jié)直腸癌細(xì)胞EMT的發(fā)生以及侵襲遷移均有著密切的聯(lián)系,這一研究結(jié)果可為臨床靶向基因治療以及后期侵襲轉(zhuǎn)移的機(jī)制探討提供一定的實(shí)驗(yàn)數(shù)據(jù)。
[1] Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012 [J]. Eur J Cancer, 2013,49 (6): 1374-1403.
[2] Brenner H, Kloor M, Pox C P. Colorectal cancer [J]. Lancet, 2014, 383 (9927):1490-1502.
[3] Center M M, Jemal A, Smith R A, et al. Worldwide variations in colorectal cancer [J]. CA Cancer J Clin, 2009, 59 (6):366-378.
[4] 王寧,孫婷婷,鄭榮壽,等.中國(guó)2009年結(jié)直腸癌發(fā)病和死亡資料分析[J].中國(guó)腫瘤,2013,22(7):515-520.
[5] Kang H, Lee M, Jang S W. Celastrol inhibits TGF-β1-induced epithelial-mesenchymal transition by inhibiting Snail and regulating E-cadherin expression[J]. Biochem Biophys Res Commun, 2013, 437 (4):550-556.
[6] Xu J, Lamouille S, Derynck R. TGF-β-induced epithelial to mesenchymal transition [J].Cell Res,2009,9(2):156-172.
[7] Wu X Y, Fu Z X, Wang X H. Peroxiredoxins in colorectal neoplasms [J]. Histol Histopathol, 2010, 25(10):1297-1303.
[8] Feng J, Fu Z, Guo J, et al. Overexpression of peroxiredoxin 2 inhibits TGF-β1-induced epithelial-mesenchymal transition and cell migration in colorectal cancer [J]. Mol Med Rep,2014,10(2): 867-873.
[9] 于偉娜,羅軍敏,徐林,等.TGF-β1誘導(dǎo)結(jié)腸癌細(xì)胞SW480發(fā)生上皮-間質(zhì)轉(zhuǎn)化及對(duì)Twist1表達(dá)的影響[J].免疫學(xué)雜志,2014,30(10):850-854.
[10] Enquist I B, Goog Z, Jubb A M, et al. Lymph node-independent liver metastasis in a model of metastatic colorectal cancer [J]. Nat Commun, 2014,Mar26,5:3530.doi:10.1038/ncomms4530.
[11] Agrawal-Singh S, Isken F, Agelopoulos K, et al. Genome-wide analysis of histone H3 acetylation patterns in AML identifies PRDX2 as an epigenetically silenced tumor suppressor gene[J].Blood, 2012,119(10):2346-2357.
[12] Lee D J, Kang D H, Choi M, et al. Peroxiredoxin2 represses melanoma metastasis by increasing Ecadherin/βcatenin complexes in adherens junctions [J].Cancer Res,2013,73(15): 4744-4757.
[13] Zhu Q C, Gao R Y, Wu W, et al. Epithelial-mesenchymal transition and its role in the pathogenesis of colorectal cancer[J]. Asian Pac J Cancer Prev, 2013, 14 (5): 2689-2698.
[14] Techasen A, Loilome W, Namwat N, et al. Cytokines released from activated human macrophages induce epithelial mesenchymal transition markers of cholangiocarcinoma cells[J]. Asian Pac J Cancer Prev, 2012,13( Suppl):115-118.
[15] Mani S A, Guo W, Liao M J, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells [J]. Cell, 2008, 133(4): 704-715.
[16] Cheng G Z, Chan J, Wang Q, et al. Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel [J]. Cancer Res, 2007, 67(5):1979-1987.
[17] 王文雙, 楊興升, 馬先瑩. Twist介導(dǎo)的上皮間質(zhì)轉(zhuǎn)化在卵巢癌組織中的作用[J]. 中國(guó)醫(yī)科大學(xué)學(xué)報(bào),2012,41(2):175-177.
[18] Argast G M, Krueger J S, Thomson S, et al. Inducible expression of TGFβ,Snail and Zeb1 recapitulates EMT in vitro and in vivo in a NSCLC model[J]. Clin Exp Metastasis, 2011,28(7):593-614.
[19] Watanabe H. Extracellular matrix-regulation of cancer invasion and metastasis [J]. Gan To Kagaku Ryoho, 2010, 37(11):2058-2061.
[20] Fan H X, Wang S, Zhao H, et al. Sonic hedgehog signaling may promote invasion and metastasis of oral squamous cell carcinoma by activating MMP-9 and E-cadherin expression[J].Med Oncol,2014,31(7):41.
[21] Zhang M, Zhu G Y, Gao H Y, et al. Expression of tissue levels matrix metalloproteinases and tissue inhibits of metalloproteinases in gastric adenocarcinoma [J]. J Surg Oncol, 2011,103(3):243-247.
[22] 茍小麗,邢時(shí)云,楊衛(wèi)兵. 聯(lián)合檢測(cè)外周血CEA、CA125、VEGF、MT及MMP-9在晚期非小細(xì)胞肺癌表達(dá)的相關(guān)性[J].遵義醫(yī)學(xué)院學(xué)報(bào),2014,37(2):204-207.
[23] Lee K W, Lee D J, Lee J Y, et al. Peroxiredoxin II restrains DNA damageinduced death in cancer cells by positively regulating JNK dependent DNA repair[J].J Biol Chem,2011,286(10): 8394-8404.
[24] Kikuta K, Tochigi N, Saito S, et al. Peroxiredoxin 2 as a chemotherapy responsiveness biomarker candidate in osteosarcoma revealed by Proteomics[J]. Proteomics Clin, 2010, 4(5):560- 576.
[25] Shiota M, Yokomizo A, Kashiwagi E, et al. Peroxiredoxin 2 in the nucleus and cytoplasm distinctly regulates androgen receptor activity in prostate cancer cells [J]. Free Radic Biol Med, 2011,51 (1):78-87.
[26] Lu W, Fu Z, Wang H, et al. Peroxiredoxin 2 is upregulated in colorectal cancer and contributes to colorectal cancer cells’ survival by protecting cells from oxidative stress [J]. Mol Cell Biochem,2014,387(1-2):261-270.
[收稿2014-12-10;修回2015-01-05]
(編輯:王福軍)
Silencing of PRDX2 promotes epithelial-mesenchymal transition of colorectal cancer cells and enhances the abilities of invasion and metastasis
YuWeina1,FengJihong2,DingChenbo1,GaoShaoying1,XuLin1,YuanJianbo3,LuoJunmin1
(1.Department of Immunology , Immunology Innovation Base of Postgraduate Education of Guizhou Province , Zunyi Medical University, Zunyi Guizhou 563099,China;2.Cancer Hospital of The Affiliated Hospital of Zunyi Medical University, Zunyi Guizhou 563099,China;3.Department of Medical Laboratory,Zunyi Medical University,Students of Grade 2011,Zunyi Guizhou 563099,China)
Objective To explore the effects of PRDX2 silencing on epithelial-mesenchymal transition of colorectal cancer cells, and its underlying mechanism in invasion and metastasis.Methods Lentiviruses control shRNA (shRNA-Control) and PRDX2 shRNA (shRNA-PRDX2) with resistance were transfected respectively into colorectal cancer SW480 cells. The transfected cells were divided into PRDX2 silencing group (shRNA-PRDX2) and empty vector control group (shRNA-Control). The expressions of PRDX2 protein and mRNA in SW480 cells were examined by Immunoblot (Western blot) and quantitative real-time PCR (qRT- PCR). Under the treatment with 5 ng/ml TGF-β1, cell morphological alterations were observed at 0, 48, and 72 h. The cell migration and invasion capabilities were evaluated with wound healing assay and transwell chamber, respectively. The protein and mRNA expressions of E-cadherin, Vimentin, Snail, Twist1 and MMP-9 were detected by Western blot and qRT-PCR.Results Compared with control group, the expressions of PRDX2 protein and mRNA were significantly decreased in PRDX2 silencing group (P<0.05). PRDX2 silencing cell line was successfully constructed. The cells with PRDX2 silencing showed a spindle-like mesenchymal phenotype and the migration and invasion were significantly increased after treatment with TGF-β1. The expression of E-cadherin was significantly decreased, while the expressions of Vimentin, Snail, Twist1 and MMP-9 were significantly increased (P<0.05).Conclusion Silencing of PRDX2 promotes epithelial-mesenchymal transition possibly by regulating the expressions of Snail and Twist1 in colorectal cancer SW480 cells,and subsequently promotes cell invasion and metastasis.
Peroxiredoxin 2 (PRDX2);gene silencing;colorectal cancer;epithelial-mesenchymal transition;invasion and metastasis
貴州省科學(xué)技術(shù)基金資助項(xiàng)目(NO:黔科合J字[2013]2312);貴州省優(yōu)秀科技教育人才省長(zhǎng)基金資助項(xiàng)目(NO:黔科教辦[2010]04)。
羅軍敏,女,教授,碩士生導(dǎo)師,研究方向:腫瘤免疫,E-mail:luojm128@163.com。
R735.3
A
1000-2715(2015)01-0074-06