李若彤, 王 麗, 曹 亭, 陳圣文, 費(fèi)洪榮, 王鳳澤△
(1泰山醫(yī)學(xué)院基礎(chǔ)醫(yī)學(xué)院,山東 泰安 271000; 泰山醫(yī)學(xué)院 2生命科學(xué)學(xué)院, 3藥學(xué)院,山東 泰安 271016)
?
Perifosine通過(guò)抑制PI3K/Akt途徑調(diào)節(jié)人膠質(zhì)瘤U251細(xì)胞增殖、凋亡與自噬*
李若彤1,王麗2,曹亭2,陳圣文2,費(fèi)洪榮3,王鳳澤2△
(1泰山醫(yī)學(xué)院基礎(chǔ)醫(yī)學(xué)院,山東 泰安 271000; 泰山醫(yī)學(xué)院2生命科學(xué)學(xué)院,3藥學(xué)院,山東 泰安 271016)
目的: 探討Akt激酶抑制劑perifosine對(duì)人膠質(zhì)瘤U251細(xì)胞凋亡、細(xì)胞周期和自噬的影響,確定perifosine誘導(dǎo)的細(xì)胞自噬與其促進(jìn)膠質(zhì)瘤細(xì)胞凋亡的相關(guān)性。方法:Perifosine處理U251細(xì)胞后,采用MTT法檢測(cè)細(xì)胞活力;流式細(xì)胞術(shù)分析perifosine對(duì)U251細(xì)胞周期的影響;Annexin V-FITC/PI雙標(biāo)法檢測(cè)perifosine對(duì)膠質(zhì)瘤細(xì)胞凋亡的作用;免疫印跡法檢測(cè)細(xì)胞內(nèi)P21、P27和cyclin B1等細(xì)胞周期調(diào)控相關(guān)蛋白以及caspase-9、PARP等細(xì)胞凋亡相關(guān)蛋白的表達(dá)水平;通過(guò)觀察細(xì)胞內(nèi)自噬標(biāo)志分子LC3-II的分布與表達(dá)來(lái)確定perifosine對(duì)自噬的誘導(dǎo)作用。結(jié)果:Perifosine劑量依賴性地抑制U251細(xì)胞活力,能夠通過(guò)抑制cyclin B1的表達(dá)而阻滯膠質(zhì)瘤細(xì)胞周期于G2期。Perifosine促進(jìn)了U251細(xì)胞內(nèi)caspase-9和PARP的剪切,抑制survivin表達(dá),從而誘導(dǎo)膠質(zhì)瘤細(xì)胞發(fā)生凋亡。同時(shí),perifosine與自噬抑制劑氯喹聯(lián)用后,U251細(xì)胞凋亡數(shù)量明顯增加。結(jié)論:Perifosine能夠抑制U251細(xì)胞的增殖,同時(shí)誘導(dǎo)細(xì)胞發(fā)生凋亡與自噬,抑制自噬促進(jìn)了perifosine誘導(dǎo)的膠質(zhì)瘤細(xì)胞凋亡。
Perifosine; Akt; 膠質(zhì)瘤; 細(xì)胞凋亡; 自噬
磷脂酰肌醇3-激酶/蛋白激酶B(phosphatidylinositol 3-kinase/protein kinase B,PI3K/Akt)信號(hào)通路廣泛參與調(diào)控細(xì)胞的增殖、存活和分化等生理病理活動(dòng)。由于11該信號(hào)途徑上游抑制因子PTEN 的突變,或者PI3K基因的擴(kuò)增及Akt 的過(guò)度活化,導(dǎo)致PI3K/Akt 信號(hào)通路在人類腫瘤譜中廣泛失調(diào)[1-2]。因此,抑制該通路可能成為惡性腫瘤治療的有效手段。Perifosine是新型的烷基磷脂類化合物,主要通過(guò)調(diào)節(jié)PI3K/Akt 和MAPK 等信號(hào)通路發(fā)揮抗腫瘤作用[3-4]。我們?cè)谇捌谘芯堪l(fā)現(xiàn)perifosine能夠抑制胃癌細(xì)胞SGC-7901的增殖并誘導(dǎo)其凋亡,表現(xiàn)出良好的體外抗腫瘤活性[5]。本研究以人膠質(zhì)瘤細(xì)胞系U251為實(shí)驗(yàn)材料,探討perifosine抑制細(xì)胞增殖的分子機(jī)理,在觀察perifosine對(duì)U251細(xì)胞凋亡和自噬影響的同時(shí),分析perifosine誘導(dǎo)細(xì)胞自噬與其促進(jìn)U251細(xì)胞凋亡的相關(guān)性。
1實(shí)驗(yàn)材料
Perifosine購(gòu)自Selleck Chemicals;胎牛血清購(gòu)自Gibco;MTT、氯喹(chloroquine,CQ)、碘化丙啶(propidium iodine,PI)、β-actin抗體和LC3-Ⅱ抗體購(gòu)自Sigma;Annexin V-FITC/PI凋亡檢測(cè)試劑盒購(gòu)自BD Biosciences;Akt抗體、caspase-9 抗體、PARP抗體、survivin抗體和Cdc2抗體購(gòu)自CST;CyclinB1抗體購(gòu)自Santa Cruz;P21抗體和P27抗體購(gòu)自BD Biosciences;p-Akt 抗體購(gòu)自Abcam;II 抗均購(gòu)自北京中杉金橋生物技術(shù)有限公司。ECL化學(xué)發(fā)光檢測(cè)試劑盒購(gòu)自Thermo。
2實(shí)驗(yàn)方法
2.1細(xì)胞培養(yǎng)人膠質(zhì)瘤細(xì)胞株U251購(gòu)自上海中科院細(xì)胞庫(kù),在37 ℃、5% CO2條件下,用含有10% 胎牛血清的DMEM 培養(yǎng)基培養(yǎng)。
2.2MTT實(shí)驗(yàn)檢測(cè)細(xì)胞活力胰酶消化U251細(xì)胞并接種于96孔板中,過(guò)夜培養(yǎng)后加入不同劑量的perifosine 處理24 h,對(duì)照組加入PBS。終止培養(yǎng)前4 h,加20 μL MTT(5 g/L)于各個(gè)孔中。吸去培養(yǎng)基,加入150 μL二甲基亞砜,室溫振蕩10 min,490 nm波長(zhǎng)下測(cè)定各孔吸光度值。實(shí)驗(yàn)重復(fù)3次。
2.3流式細(xì)胞術(shù)檢測(cè)細(xì)胞周期不同濃度的perifosine作用細(xì)胞24 h后,胰酶消化并制備單細(xì)胞懸液,然后加入預(yù)冷的75%無(wú)水乙醇于4 ℃固定18 h。接著用PBS洗滌細(xì)胞2次,加入終濃度為50 mg/L RNase A于37 ℃孵育30 min;加入終濃度為50 mg/L的PI于4 ℃閉光染色20 min,然后上機(jī)檢測(cè)分析各組細(xì)胞的周期分布。實(shí)驗(yàn)重復(fù)3次。
2.4細(xì)胞凋亡的檢測(cè)采用Annexin V-FITC/PI雙標(biāo)法檢測(cè)細(xì)胞凋亡,具體操作方法參照試劑盒說(shuō)明書進(jìn)行,根據(jù)Annexin V-FITC/PI的熒光強(qiáng)度計(jì)算細(xì)胞凋亡百分率。實(shí)驗(yàn)重復(fù)3次。
2.5免疫印跡實(shí)驗(yàn)細(xì)胞經(jīng)不同劑量perifosine處理24 h后,進(jìn)行免疫印跡分析。各組細(xì)胞經(jīng)PBS洗滌2次后,用預(yù)冷的RIPA蛋白裂解液冰上裂解30 min,然后4 ℃ 12 000 r/min離心20 min,收集上清并用BCA 法定量。經(jīng)SDS-PAGE分離蛋白,將凝膠上的蛋白質(zhì)電轉(zhuǎn)至硝酸纖維素膜上;再用5%脫脂奶粉封閉過(guò)夜,加入 I 抗,室溫孵育3h后加入 II 抗,最后ECL 發(fā)光液曝光顯影。實(shí)驗(yàn)重復(fù)3次。
2.6細(xì)胞內(nèi)GFP-LC3 融合蛋白的表達(dá)接種U251細(xì)胞于6 孔板中,待細(xì)胞融合至80% 左右用Lipofectamine 2000 轉(zhuǎn)染GFP-LC3 質(zhì)粒。轉(zhuǎn)染24 h 后,加入不同濃度的perifosine 繼續(xù)處理24 h,倒置熒光顯微鏡下觀察并拍照。無(wú)自噬時(shí),GFP-LC3 融合蛋白一般呈彌散狀態(tài)分布于胞漿中,當(dāng)細(xì)胞發(fā)生自噬時(shí),GFP-LC3 融合蛋白轉(zhuǎn)位至自噬體膜,在熒光顯微鏡下形成明亮的綠色斑點(diǎn)(相當(dāng)于自噬體)。
3統(tǒng)計(jì)學(xué)處理
采用SPSS 16.0統(tǒng)計(jì)軟件分析,數(shù)據(jù)用均數(shù)±標(biāo)準(zhǔn)差(mean±SD)表示,多組間比較應(yīng)用單因素方差分析,組間兩兩比較應(yīng)用SNK-q檢驗(yàn),以P<0.05 為差異有統(tǒng)計(jì)學(xué)意義。
1Perifosine抑制U251細(xì)胞內(nèi)Akt的磷酸化
Akt 第473 位點(diǎn)絲氨酸磷酸化(p-AktS473)增強(qiáng)通常被作為Akt激活的指標(biāo)。由于perifosine是Akt 的抑制劑,所以本研究首先檢測(cè)了perifosine對(duì)U251細(xì)胞內(nèi)Akt 磷酸化(p-AktS473)的抑制作用。免疫印跡結(jié)果顯示經(jīng)perifosine處理24 h后,細(xì)胞內(nèi)Akt的磷酸化水平顯著受到抑制,且抑制作用呈劑量依賴性。細(xì)胞內(nèi)總Akt 的表達(dá)量無(wú)明顯變化,見(jiàn)圖1。
2Perifosine阻滯U251細(xì)胞于G2期
用MTT法檢測(cè)了perifosine作用24 h 后對(duì)U251細(xì)胞活力的影響。結(jié)果顯示perifosine可顯著抑制U251細(xì)胞活力,且抑制作用呈劑量依賴性,見(jiàn)圖2。用流式細(xì)胞術(shù)分析了perifosine對(duì)U251細(xì)胞周期的影響,結(jié)果如圖3和表1所示,perifosine處理24 h后,處于G2期的細(xì)胞數(shù)量明顯增多,表明perifosine能夠誘導(dǎo)細(xì)胞發(fā)生G2期阻滯。
Figure 1.The protein levels of p-Akt and total Akt in the U251 cells treated with perifosine for 24 h determined by Western blot. Mean±SD.n=3.*P<0.05vs0 μmol/L.
圖1Perifosine 抑制U251細(xì)胞內(nèi)Akt的磷酸化
Figure 2.The effects of perifosine treatment for 24 h at different concentrations on the viability of the U251 cells determined by MTT assay. Mean±SD.n=3.*P<0.05vs0 μmol/L.
圖2Perifosine抑制U251細(xì)胞活力
免疫印跡實(shí)驗(yàn)結(jié)果表明,perifosine抑制膠質(zhì)瘤細(xì)胞中P27和cyclin B1的表達(dá),而P21的蛋白水平則隨著perifosine的濃度增加而上調(diào),但Cdc2的表達(dá)水平無(wú)明顯變化,見(jiàn)圖4。
3Perifosine 誘導(dǎo)U251細(xì)胞發(fā)生凋亡
誘導(dǎo)腫瘤細(xì)胞發(fā)生凋亡如同抑制細(xì)胞增殖一樣,也是小分子藥物抗腫瘤作用的重要理論依據(jù)。因此,我們采用Annexin V-FITC/PI雙標(biāo)法檢測(cè)perifosine對(duì)膠質(zhì)瘤細(xì)胞凋亡的影響。如同對(duì)SGC-7901胃癌細(xì)胞凋亡的誘導(dǎo)作用,較高濃度的perifosine同樣促進(jìn)了膠質(zhì)瘤細(xì)胞U251發(fā)生凋亡,見(jiàn)表2。
免疫印跡法檢測(cè)了perifosine對(duì)U251細(xì)胞內(nèi)凋亡相關(guān)蛋白活性及表達(dá)水平的影響。發(fā)現(xiàn)較高濃度的perifosine促進(jìn)了胱天蛋白酶家族成員caspase-9和PARP的切割,而凋亡抑制蛋白survivin的表達(dá)則逐漸降低,見(jiàn)圖5。
Figure 3.Upon exposure to perifosine for 24 h, U251 cells exhiibted G2phase arrest.
圖3Perifosine誘導(dǎo)U251細(xì)胞阻滯于G2期
表1Perifosine誘導(dǎo)U251細(xì)胞周期阻滯于G2期
Table 1.Upon exposure to perifosine for 24 h, U251 cells exhi-bited G2phase arrest (%. Mean±SD.n=3)
Perifosine(μmol/L)G0/G1SG2/M066.86±3.7518.53±1.5114.27±1.38559.26±3.77*17.89±1.6323.68±1.82*1041.31±2.93*18.03±1.8739.20±2.49*
*P<0.05vs0 μmol/L.
4Perifosine抑制U251細(xì)胞發(fā)生自噬
轉(zhuǎn)染GFP-LC3質(zhì)粒的膠質(zhì)瘤細(xì)胞經(jīng)perifosine處理24 h 后,GFP-LC3 細(xì)胞內(nèi)的點(diǎn)狀分布增加,證實(shí)perifosine能夠促進(jìn)膠質(zhì)瘤細(xì)胞自噬的發(fā)生,見(jiàn)圖6。同時(shí),免疫印跡法檢測(cè)perifosine對(duì)人膠質(zhì)瘤細(xì)胞內(nèi)LC3-Ⅱ蛋白表達(dá)的影響,發(fā)現(xiàn)不同濃度的perifosine處理細(xì)胞后,LC3-Ⅱ蛋白表達(dá)水平明顯增多,進(jìn)一步證明perifosine能夠誘導(dǎo)U251細(xì)胞發(fā)生自噬,見(jiàn)圖7。
Figure 4.The effects of perifosine treatment at different concentrations for 24 h on the expression of cell cycle-associated proteins in the U251 cells. Cell lysates were analyzed by Western blot. Mean±SD.n=3.*P<0.05vs0 μmol/L.
圖4Perifosine對(duì)細(xì)胞周期相關(guān)蛋白表達(dá)水平的影響
表2 Perifosine 促進(jìn)U251細(xì)胞發(fā)生凋亡
*P<0.05vs0 μmol/L.
5抑制自噬促進(jìn)perifosine誘導(dǎo)的U251細(xì)胞凋亡
CQ是特異性自噬抑制劑,主要抑制細(xì)胞內(nèi)自噬體和溶酶體的融合[6]。U251細(xì)胞經(jīng)CQ(10 μmol/L)與perifosine(20 μmol/L)聯(lián)合作用24 h后,Annexin V-FITC/PI雙標(biāo)法檢測(cè)了細(xì)胞凋亡的變化。結(jié)果表明抑制自噬促進(jìn)了perifosine誘導(dǎo)的U251細(xì)胞凋亡,見(jiàn)表3。同時(shí)免疫印跡結(jié)果顯示,自噬抑制劑CQ與perifosine 聯(lián)用促進(jìn)了PARP的切割,見(jiàn)圖8。
Figure 5.The effects of perifosine treatment for 24 h at different concentrations on the levels of apoptosis-related proteins in the U251 cells determined by Western blot. Mean±SD.n=3.*P<0.05vs0 μmol/L.
圖5免疫印跡檢測(cè)perifosine對(duì)U251細(xì)胞內(nèi)凋亡相關(guān)蛋白的影響
Figure 6.Representative images of GFP-LC3 in the U251 cells transfected with GFP-LC3 plasmid. After transfection for 24 h, the cells were treated with perifosine for another 24 h.
圖6Perifosine對(duì)U251細(xì)胞GFP-LC3融合蛋白表達(dá)的影響
膠質(zhì)瘤是成人常見(jiàn)的惡性腦腫瘤,其侵襲性強(qiáng),術(shù)后復(fù)發(fā)率高。盡管治療手段不斷進(jìn)步,但中位生存期仍無(wú)明顯改進(jìn)。目前膠質(zhì)瘤的治療以手術(shù)治療為主,化療則是常規(guī)的輔助治療方法,但化療藥物的獲得性耐藥使得化療效果較差,因此迫切需要尋找新的藥物來(lái)輔助膠質(zhì)瘤的手術(shù)治療。PI3K/Akt信號(hào)通路廣泛存在于細(xì)胞中,通過(guò)調(diào)節(jié)細(xì)胞周期和細(xì)胞能量代謝等多種途徑發(fā)揮重要的生理功能。研究發(fā)現(xiàn),PI3K/Akt信號(hào)途徑在膠質(zhì)瘤中過(guò)度活化[7-8],提示該通路與膠質(zhì)瘤的發(fā)生發(fā)展關(guān)系密切,而PI3K和Akt激酶或許成為膠質(zhì)瘤臨床治療的有效靶點(diǎn)。
Perifosine作為新型的Akt抑制劑,能夠通過(guò)抑制Akt和細(xì)胞外信號(hào)調(diào)節(jié)激酶1/2(extracellular signal-regulated kinase 1/2,ERK1/2)的磷酸化而抑制小鼠膠質(zhì)祖細(xì)胞的增殖[9]。本研究以U251膠質(zhì)瘤細(xì)胞為實(shí)驗(yàn)材料,主要探討Akt抑制劑perifosine對(duì)U251細(xì)胞增殖、凋亡和自噬的影響。我們發(fā)現(xiàn),perifosine能夠阻滯細(xì)胞于G2期而抑制增殖;通過(guò)促進(jìn)caspase-9和PARP的切割,抑制survivin的表達(dá)而誘導(dǎo)細(xì)胞凋亡。細(xì)胞增殖失控是惡性腫瘤最基本的生物學(xué)特征,而細(xì)胞周期調(diào)控紊亂是增殖失控的根本原因。因此,調(diào)控細(xì)胞周期進(jìn)程與誘導(dǎo)細(xì)胞凋亡或壞死一樣已成為腫瘤治療的重要途徑之一。細(xì)胞周期蛋白是真核細(xì)胞周期進(jìn)程的主要調(diào)節(jié)蛋白,通過(guò)與細(xì)胞周期蛋白依賴性激酶(cyclin-dependent kinases,CDKs)結(jié)合形成cyclin-CDKs 復(fù)合物共同調(diào)控著細(xì)胞周期[10]。當(dāng)腫瘤細(xì)胞經(jīng)藥物或輻射處理后,通常出現(xiàn)G1/S期或G2/M期阻滯,以便修復(fù)受損的DNA。Cdc2/cyclin B1作為G2期檢驗(yàn)點(diǎn)的重要開(kāi)關(guān),與Cdc25C相互作用于細(xì)胞周期,開(kāi)啟細(xì)胞有絲分裂進(jìn)程[11]。本研究用perifosine處理U251細(xì)胞后,cyclin B1的蛋白水平下調(diào),使Cdc2/cyclin B1 復(fù)合物的活性降低,從而發(fā)生G2期阻滯。
Figure 7.The effect of perifosine treatment for 24 h at different concentrations on the expression of LC3-II in the U251 cells detected by Western blot. Mean±SD.n=3.*P<0.05vscontrol.
圖7Perifosine對(duì)U251細(xì)胞中LC3-II表達(dá)的影響
表3抑制自噬增強(qiáng)perifosine誘導(dǎo)的U251細(xì)胞凋亡
Table 3.The effects of autophagic inhibitor chloroquine (CQ) on perifosine-induced cell apoptosis (Mean±SD.n=3)
TreatmentEarlyapoptoticrate(%)Lateapoptoticornecroticrate(%)Control1.36±0.644.25±1.33Perifosine5.03±1.95*5.80±2.17CQ2.53±0.866.38±1.75Perifosine+CQ12.07±3.72*#11.40±3.69*#
*P<0.05vscontrol;#P<0.05vsperifosine group.
Figure 8.U251 cells were treated with 20 μmol/L perifosine and/or with 10 μmol/L CQ for 24 h, and the protein level of cleaved PARP was determined by Western blot. Mean±SD.n=3.*P<0.05vscontrol;#P<0.05vsperifosine group.
圖8CQ抑制自噬增強(qiáng)perifosine誘導(dǎo)的PARP切割
細(xì)胞自噬是真核細(xì)胞所特有的一種胞內(nèi)代謝途徑,細(xì)胞通過(guò)自我消化折疊錯(cuò)誤的蛋白質(zhì)或受損的細(xì)胞器,為機(jī)體提供物質(zhì)能量從而保證大分子的合成[12]。自噬與腫瘤的發(fā)生、發(fā)展密切相關(guān)。有研究證明自噬能夠誘導(dǎo)腫瘤細(xì)胞發(fā)生非凋亡途徑的細(xì)胞死亡;但越來(lái)越多實(shí)驗(yàn)發(fā)現(xiàn)自噬不但能夠?yàn)槟[瘤細(xì)胞提供能量,促進(jìn)腫瘤細(xì)胞存活,而且與腫瘤細(xì)胞的化療耐藥性相關(guān)[13-15]。當(dāng)用化療藥物誘導(dǎo)腫瘤細(xì)胞凋亡時(shí),細(xì)胞自噬活性增強(qiáng),產(chǎn)生耐藥性,而與自噬抑制劑聯(lián)用能夠減弱這種耐藥性[16]。本研究中,我們觀察了perifosine對(duì)U251細(xì)胞自噬的影響,發(fā)現(xiàn)perifosine促進(jìn)了U251細(xì)胞發(fā)生自噬,抑制自噬增強(qiáng)了perifosine誘導(dǎo)的U251細(xì)胞凋亡,表明perifosine誘導(dǎo)的U251細(xì)胞自噬對(duì)細(xì)胞是保護(hù)性的。
總之,perifosine能夠誘導(dǎo)細(xì)胞周期阻滯、細(xì)胞凋亡和自噬。抑制自噬增強(qiáng)了perifosine誘導(dǎo)的U251細(xì)胞凋亡,本研究為膠質(zhì)瘤的臨床輔助治療提供了新的策略。
[1]Chia S, Gandhi S, Joy AA, et al. Novel agents and associated toxicities of inhibitors of the PI3K/Akt/mTOR pathway for the treatment of breast cancer [J]. Curr Oncol, 2015, 22(1):33-48.
[2]Fumarola C, Bonelli MA, Petronini PG, et al. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer[J]. Biochem Pharmacol, 2014, 90(3):197-207.
[3]Fei HR, Chen G, Wang JM, et al. Perifosine induces cell cycle arrest and apoptosis in human hepatocellular carcinoma cell lines by blockade of Akt phosphorylation [J]. Cytotechnology, 2010, 62(5):449-460.
[4]Chiarini F, Del Sole M, Mongiorgi S, et al. The novel Akt inhibitor, perifosine, induces caspase-dependent apoptosis and downregulates P-glycoprotein expression in multidrug-resistant human T-acute leukemia cells by a JNK-dependent mechanism[J]. Leukemia, 2008, 22(6):1106-1116.
[5]費(fèi)洪榮,陳洪蕾,辛?xí)悦?,? Akt抑制劑perifosine抑制胃癌細(xì)胞增殖并誘導(dǎo)凋亡的實(shí)驗(yàn)研究[J]. 中國(guó)病理生理雜志, 2011, 27(6):1084-1089.
[6]Zou Y, Ling YH, Sironi J, et al. The autophagy inhibitor chloroquine overcomes the innate resistance of wild-type EGFR non-small-cell lung cancer cells to erlotinib [J]. J Thorac Oncol, 2013, 8(6):693-702.
[7]Chao Y, Wang Y, Liu X, et al. Mst1 regulates glioma cell proliferation via the AKT/mTOR signaling pathway [J]. J Neurooncol, 2015, 121(2):279-288.
[8]Nicoletti NF, Erig TC, Zanin RF, et al. Mechanisms involved in kinin-induced glioma cells proliferation: the role of ERK1/2 and PI3K/Akt pathways[J]. J Neurooncol, 2014, 120(2):235-244.
[9]Momota H, Nerio E, Holland EC. Perifosine inhibits multiple signaling pathways in glial progenitors and cooperates with temozolomide to arrest cell proliferation in gliomasinvivo[J]. Cancer Res, 2005, 65(16):7429-7435.
[10]Goldenson B, Crispino JD. The aurora kinases in cell cycle and leukemia[J]. Oncogene, 2015, 34(5):537-545
[11]Dillon MT, Good JS, Harrington KJ. Selective targeting of the G2/M cell cycle checkpoint to improve the therapeutic index of radiotherapy [J]. Clin Oncol (R Coll Radiol), 2014, 26(5):257-265.
[12]Galluzzi L, Pietrocola F, Levine B, et al. Metabolic control of autophagy[J]. Cell, 2014, 159(6):1263-1276.
[13]Liu Y, Levine B. Autosis and autophagic cell death: the dark side of autophagy[J]. Cell Death Differ, 2015, 22(3):367-376.
[14]張翠,姜文艷,吳玉梅,等. 抑制自噬促進(jìn)MK-2206誘導(dǎo)SGC-7901胃癌細(xì)胞發(fā)生DNA損傷[J]. 中國(guó)病理生理雜志, 2015, 31(9):1545-1549.
[15]周凱亮,吳凱,張小磊,等. 自噬抑制增強(qiáng)白藜蘆醇對(duì)人軟骨肉瘤細(xì)胞凋亡的誘導(dǎo)[J]. 中國(guó)病理生理雜志, 2015, 31(8):1401-1406.
[16]He J, Yu JJ, Xu Q, et al. Downregulation of ATG14 by EGR1-MIR152 sensitizes ovarian cancer cells to cisplatin-induced apoptosis by inhibiting cyto-protective autophagy[J]. Autophagy, 2015, 11(2):373-384.
(責(zé)任編輯: 林白霜, 羅森)
Perifosine regulates human brain glioma U251 cell proliferation, apoptosis and autophagy through suppression of PI3K/Akt pathway
LI Ruo-tong1, WANG Li2, CAO Ting2, CHEN Sheng-wen2, FEI Hong-rong3, WANG Feng-ze2
(1SchoolofBasicMedicalSciences,TaishanMedicalUniversity,Tai’an271000,China;2SchoolofLifeSciences,3SchoolofPharmaceuticalSciences,TaishanMedicalUniversity,Tai’an271016,China.E-mail:wfz221@sina.com)
AIM: To investigate the effect of perifosine, an inhibitor of protein kinase B (PKB/Akt), on the cell cycle, apoptosis and autophagy in human brain glioma U251 cells, and to determine the relationship between perifosine-induced autophagy and apoptosis of glioma. METHODS: The cell growth inhibition was determined by MTT assay. The cell cycle distribution of U251 cells was examined by flow cytometry. The cell apoptosis was analyzed by Annexin V-FITC apoptosis detection kit. The protein expression of P21, P27, cyclin B1, caspase-9 and PARP was examined by Wes-tern blot analysis. The distribution and expression of LC3-II, an autophagy marker, was observed to determine the effect of perifosine-induced autophagy. RESULTS: Perifosine inhibited the cell viability in a dose-dependent manner. In perifosine-treated U251 cells, the cell cycle was arrested in G2phase and the expression of cyclin B1 was inhibited. Perifosine induced apoptosis of U251 cells through activation of caspase-9 cleavage, PARP cleavage and survivin inhibition. In addition, suppression of autophagy by chloroquin, an inhibitor of autophagy, increased the number of apoptotic cells. CONCLUSION: Perifosine inhibits cell proliferation and triggers apoptosis and autophagy in human U251 cells. Blocking autophagy magnifies perifosine-induced glioma cell apoptosis.
Perifosine; Akt; Glioma; Apoptosis; Autophagy
1000- 4718(2016)04- 0644- 07
2015- 10- 27
2015- 12- 31
國(guó)家自然科學(xué)基金資助項(xiàng)目(No.81272683);山東省高等學(xué)??萍加?jì)劃項(xiàng)目(No.J15LM09)
Tel: 0538-6236991; E-mail: wfz221@sina.com
R730.23
A
10.3969/j.issn.1000- 4718.2016.04.011
雜志網(wǎng)址: http://www.cjpp.net