• 
    

    
    

      99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看 ?

      膠質(zhì)瘤中甲基化沉默腫瘤抑制因子的研究進(jìn)展

      2016-05-09 00:48:03涂艷陽(yáng)徐曉珊張永生成迎端
      關(guān)鍵詞:膠質(zhì)瘤研究進(jìn)展

      涂艷陽(yáng),徐曉珊,張永生,成迎端

      (第四軍醫(yī)大學(xué):1唐都醫(yī)院實(shí)驗(yàn)外科,2唐都醫(yī)院,陜西西安710038;3賽福地,美國(guó)新澤西08902)

      ?

      膠質(zhì)瘤中甲基化沉默腫瘤抑制因子的研究進(jìn)展

      涂艷陽(yáng)1,徐曉珊1,張永生2,成迎端3

      (第四軍醫(yī)大學(xué):1唐都醫(yī)院實(shí)驗(yàn)外科,2唐都醫(yī)院,陜西西安710038;3賽福地,美國(guó)新澤西08902)

      【摘 要】腫瘤抑制基因(tumor suppressor genes,TSGs)的表觀遺傳沉默對(duì)膠質(zhì)瘤的發(fā)生和發(fā)展至關(guān)重要.越來(lái)越多的與細(xì)胞周期、增殖、凋亡、遷移、浸潤(rùn)、DNA修復(fù)和信號(hào)通路密切相關(guān)的且被甲基化調(diào)控的TSGs被鑒定出來(lái).TSGs在膠質(zhì)瘤里特異性的甲基化表明其既可以作為分子診斷的標(biāo)記物,也可能成為治療膠質(zhì)瘤的一個(gè)靶標(biāo).本研究對(duì)人腦膠質(zhì)瘤中被甲基化沉默的TSGs進(jìn)行了總結(jié),這為理解膠質(zhì)瘤發(fā)展過(guò)程中表觀遺傳學(xué)修飾的紊亂提供了依據(jù).

      【關(guān)鍵詞】膠質(zhì)瘤;甲基化沉默腫瘤;抑制因子;研究進(jìn)展

      0 引言

      膠質(zhì)瘤是成人中樞神經(jīng)系統(tǒng)最常見(jiàn)的惡性腫瘤之一[1].按細(xì)胞的組織學(xué)特征不同將神經(jīng)膠質(zhì)瘤分為以下幾種:室管膜瘤(室管膜細(xì)胞),星形細(xì)胞瘤(星形細(xì)胞)和少突膠質(zhì)細(xì)胞瘤(少突膠質(zhì)細(xì)胞)[2].根據(jù)世界衛(wèi)生組織對(duì)腦腫瘤進(jìn)行分類(lèi),神經(jīng)膠質(zhì)瘤可進(jìn)一步被分成低級(jí)(Ⅰ級(jí)和Ⅱ級(jí))及高級(jí)(Ⅲ級(jí)和Ⅳ級(jí))腫瘤.Ⅳ級(jí)膠質(zhì)母細(xì)胞瘤(glioblastom,GBM)是最嚴(yán)重的惡性膠質(zhì)瘤.盡管各種治療手段如手術(shù)、放療、光動(dòng)力療法和化療等在不斷進(jìn)步,然而Ⅳ級(jí)膠質(zhì)母細(xì)胞瘤的中位生存期仍然只有14.6個(gè)月[3-4].大約60%的高級(jí)別膠質(zhì)瘤為GBM,其發(fā)病率約為0.003%[5].研究這種惡性腫瘤的分子機(jī)制和早期診斷對(duì)于GBM的成功治療至關(guān)重要.從一個(gè)正常細(xì)胞發(fā)展至一個(gè)腫瘤是一個(gè)復(fù)雜的多步驟過(guò)程.有很多原癌基因、抑癌基因和信號(hào)轉(zhuǎn)導(dǎo)通路都參與其中[6-7].癌癥的發(fā)生過(guò)程是由遺傳和表觀遺傳改變的累積所致,這直接導(dǎo)致了細(xì)胞增殖與凋亡之間的不平衡.異常的表觀遺傳修飾,如DNA甲基化,組蛋白修飾,核小體定位,非編碼RNA和microRNA在癌癥的發(fā)展過(guò)程中起重要作用[7].目前,在腫瘤發(fā)生過(guò)程中表觀遺傳學(xué)改變比遺傳學(xué)變化更重要,這一觀點(diǎn)已經(jīng)被廣泛接受[8].目前已經(jīng)鑒定出了很多在多種腫瘤中被甲基化沉默的腫瘤抑制因子,而且這些基因可以作為癌癥診斷的生物標(biāo)記物[9-14].在膠質(zhì)瘤中甲基化的腫瘤抑制因子無(wú)疑是非常好的生物標(biāo)記分子[15].在膠質(zhì)瘤中已經(jīng)鑒定出了非常多的抑癌基因,包括p16INK4a、p14ARF、MLH1、O6?甲基鳥(niǎo)嘌呤?DNA甲基轉(zhuǎn)移酶(methyl guanine?DNA methyltransferase,MGMT)、NDRG2、SLIT2等.本研究將總結(jié)在膠質(zhì)瘤中已經(jīng)研究過(guò)的被甲基化修飾的腫瘤抑制因子并根據(jù)他們的生物學(xué)功能進(jìn)行分類(lèi),如細(xì)胞周期、細(xì)胞凋亡、入侵、DNA修復(fù)和通路相關(guān)的基因(表1).一些有代表性的基因也在本研究中給出了詳細(xì)的描述,所有的這些基因都有可能在以后的研究中成為潛在的診斷標(biāo)記物和治療靶標(biāo).

      1 細(xì)胞周期和增殖

      在正常細(xì)胞中,細(xì)胞周期和增殖是由一系列因子調(diào)控的.在這個(gè)調(diào)控過(guò)程中的錯(cuò)誤與腫瘤的發(fā)生有很大關(guān)系.p16INK4a和p14ARF是CDKN2A的選擇性剪接的兩個(gè)亞型,位于人類(lèi)染色體9p21上[16-17].作為細(xì)胞周期蛋白依賴(lài)性激酶4(CDK4)和6(CDK6)的抑制劑,p16維持腫瘤抑制基因Rb的非磷酸化狀態(tài)從而抑制細(xì)胞周期進(jìn)程.p16的啟動(dòng)子甲基化在腦膠質(zhì)瘤中是很常見(jiàn)的,它的甲基化導(dǎo)致細(xì)胞周期進(jìn)程失去控制,引起膠質(zhì)瘤細(xì)胞的大量增殖[15-18].p14可通過(guò)誘導(dǎo)MDM2蛋白的構(gòu)象變化抑制MDM2介導(dǎo)的p53蛋白降解.p14ARF的表觀遺傳失活增加了p53蛋白的降解,從而導(dǎo)致p53誘導(dǎo)的細(xì)胞凋亡的下調(diào)[19].p16INK4a和p14ARF在膠質(zhì)瘤里的甲基化已經(jīng)有很多文獻(xiàn)報(bào)道過(guò)[19-22].除了這些眾所周知的細(xì)胞周期的調(diào)控因子外,還有很多其它被甲基化抑制的基因也具有調(diào)節(jié)神經(jīng)膠質(zhì)瘤細(xì)胞周期和增殖的能力.NSD1基因編碼一個(gè)參與染色質(zhì)調(diào)控的組蛋白甲基轉(zhuǎn)移酶.它包含多個(gè)功能區(qū)域,一個(gè)SET結(jié)構(gòu)域,植物同源結(jié)構(gòu)域,和脯氨酸?色氨酸?色氨酸?脯氨酸結(jié)構(gòu)域[23].在人類(lèi)神經(jīng)母細(xì)胞瘤和神經(jīng)膠質(zhì)瘤細(xì)胞中NSD1基因會(huì)被啟動(dòng)子甲基化抑制.在轉(zhuǎn)化的細(xì)胞中,NSD1的表觀遺傳失活導(dǎo)致組蛋白賴(lài)氨酸殘基H4?K20和H3?K36的甲基化特異性減少.NSD1可以通過(guò)減少集落形成密度和抑制細(xì)胞生長(zhǎng)來(lái)達(dá)到抑制腫瘤的功能.在神經(jīng)母細(xì)胞瘤和腦膠質(zhì)瘤中NSD1的CpG島過(guò)甲基化經(jīng)常被檢測(cè)到.最重要的是,NSD1啟動(dòng)子過(guò)甲基化是在高風(fēng)險(xiǎn)神經(jīng)母細(xì)胞瘤不良預(yù)后的一個(gè)預(yù)測(cè)因素.這些結(jié)果表明,NSD1的失活導(dǎo)致了組蛋白甲基化紊亂,且它具有很好的應(yīng)用價(jià)值[23].

      受體蛋白酪氨酸磷酸酶促進(jìn)因子PTPRD是高度保守的受體家族PTP中的一個(gè)成員.PTPRD基因編碼帶有細(xì)胞質(zhì)酪氨酸磷酸酶結(jié)構(gòu)域的跨膜蛋白,因?yàn)閱?dòng)子CpG島的過(guò)甲基化此基因經(jīng)常被沉默.它的失活有50%以上發(fā)生在惡性膠質(zhì)瘤,同時(shí)這種失活也對(duì)應(yīng)了膠質(zhì)瘤患者不良的預(yù)后反應(yīng).功能研究表明野生型PTPRD通過(guò)癌蛋白STAT3去磷酸化從而抑制GBM和其他腫瘤的生長(zhǎng).這些結(jié)果表明PTPRD是一個(gè)在GBM的發(fā)生期間時(shí)被沉默的癌癥抑制因子[24].

      另一個(gè)細(xì)胞周期調(diào)節(jié)因子,Krüppel樣因子4 (KLF4),在神經(jīng)膠質(zhì)瘤中也被甲基化調(diào)控.KLF4是KLF鋅指結(jié)構(gòu)轉(zhuǎn)錄因子家族的一個(gè)成員.它包含三個(gè)高度保守C2H2類(lèi)型的的鋅指結(jié)構(gòu)與一個(gè)N端反式激活結(jié)構(gòu)域[25-26].KLF4在RNA和蛋白水平上的下調(diào)有40%以上是發(fā)生在原發(fā)性的髓母細(xì)胞瘤中.體內(nèi)和體外實(shí)驗(yàn)表明KLF4在D283髓母細(xì)胞系的會(huì)顯著抑制髓母細(xì)胞瘤的生長(zhǎng)[27].這些都說(shuō)明KLF4已經(jīng)被證實(shí)在髓母細(xì)胞瘤中是一個(gè)腫瘤抑制因子.

      NDRG2,N?Myc的下游調(diào)節(jié)基因2,位于14號(hào)染色體q11.2上.研究發(fā)現(xiàn)NDRG2在人腦膠質(zhì)瘤中的表達(dá)顯著低于癌旁正常組織,而這種下調(diào)也是由啟動(dòng)子甲基化引起的,且它的下調(diào)與膠質(zhì)瘤患者的癌癥等級(jí)和患者預(yù)后呈負(fù)相關(guān)性.體外實(shí)驗(yàn)表明,在人源膠質(zhì)瘤細(xì)胞U373和U138細(xì)胞中過(guò)表達(dá)NDGR2能夠抑制細(xì)胞增殖.深層次的研究表明,NDRG2主要通過(guò)調(diào)節(jié)組蛋白乙酰化的水平來(lái)控制神經(jīng)膠質(zhì)瘤細(xì)胞的生長(zhǎng).

      2 細(xì)胞凋亡

      細(xì)胞凋亡在許多生理過(guò)程中起關(guān)鍵作用,如胚胎發(fā)育和組織更新.當(dāng)細(xì)胞凋亡發(fā)生異常時(shí)會(huì)導(dǎo)致多種疾病的發(fā)生,其中也包括癌癥[28].研究發(fā)現(xiàn),一些與凋亡相關(guān)的基因在神經(jīng)膠質(zhì)瘤中也會(huì)被DNA甲基化沉默.RANK/TNFRSF11A基因編碼I型跨膜蛋白,其通過(guò)與其配體RANKL結(jié)合,磷酸化腫瘤壞死因子受體相關(guān)蛋白從而激活信號(hào)傳導(dǎo)途徑,如核轉(zhuǎn)錄因子NF?κB、JNK、ERK、p38和AKT/PKB[29].RANK/TN?FRSF11A的啟動(dòng)子甲基化在膠質(zhì)瘤組織和膠質(zhì)瘤細(xì)胞系中是很常見(jiàn)的.在膠質(zhì)瘤細(xì)胞系中恢復(fù)RANK/TNFRSF11A的表達(dá)會(huì)引起克隆形成顯著減少,細(xì)胞凋亡明顯增加.該基因通過(guò)調(diào)控NF?κB、環(huán)磷酸腺苷(cAMP)/蛋白激酶α(cAMP應(yīng)答元件[CRE])、缺氧誘導(dǎo)因子、Oct4和Wnt等通路調(diào)控凋亡[30].

      Neogenin在細(xì)胞凋亡,分化和軸突定向發(fā)育的調(diào)控中起作用,同時(shí)研究人員也發(fā)現(xiàn)其在結(jié)腸癌組織中表達(dá)被抑制.研究表明在膠質(zhì)瘤中Neogenin有很大幾率通過(guò)啟動(dòng)子甲基化而下調(diào),Neogenin的下調(diào)不僅與膠質(zhì)瘤惡性程度而且與神經(jīng)膠質(zhì)瘤復(fù)發(fā)呈負(fù)相關(guān).在SHG?44細(xì)胞中過(guò)表達(dá)Neogenin能夠促進(jìn)細(xì)胞凋亡.目前的研究表明,Neogenin的下調(diào)可以促進(jìn)腦膠質(zhì)瘤發(fā)生的[31].

      非甾體抗炎藥活化基因NAG?1是轉(zhuǎn)化生長(zhǎng)因子β家族成員.研究表明,NAG?1在膠質(zhì)瘤組織和膠質(zhì)瘤細(xì)胞系中經(jīng)常被甲基化.NAG?1的基礎(chǔ)表達(dá)水平與神經(jīng)膠質(zhì)瘤分級(jí)成反比.在膠質(zhì)瘤細(xì)胞它的表達(dá)可以通過(guò)去甲基化藥物恢復(fù)[32].NAG?1作為一個(gè)抑癌基因,在神經(jīng)膠質(zhì)瘤細(xì)胞中是通過(guò)誘導(dǎo)凋亡發(fā)揮作用的,而PI3K/Akt和Smad信號(hào)途徑在NAG?1誘導(dǎo)的GBM細(xì)胞凋亡中則發(fā)揮相反的作用[33].

      3 侵襲和遷移

      癌細(xì)胞的遷移和侵襲的能力使得它能夠從原發(fā)灶轉(zhuǎn)移到周?chē)慕M織[34].參與轉(zhuǎn)移的過(guò)程中,細(xì)胞?細(xì)胞和細(xì)胞?基質(zhì)間相互作用至關(guān)重要.細(xì)胞粘附的破壞導(dǎo)致接觸生長(zhǎng)抑制作用消失,這是腫瘤產(chǎn)生的早期步驟[15].WNK2,絲氨酸/蘇氨酸激酶,是WNK蛋白激酶亞家族的一員.WNK2是一個(gè)抑癌基因,在膠質(zhì)瘤中通過(guò)啟動(dòng)子甲基化表達(dá)下調(diào).沒(méi)有WNK2表達(dá)的患者預(yù)后都較差.研究發(fā)現(xiàn),WNK2下調(diào)后JNK失活,引起MMP2表達(dá)減少和活性降低,從而導(dǎo)致腦膠質(zhì)瘤細(xì)胞的侵襲增加[35-36].

      粘附連接相關(guān)蛋白1(adherens junctional associ?ated protein?1,AJAP1)是在細(xì)胞粘附連接處的一個(gè)跨膜蛋白,具有抑制神經(jīng)膠質(zhì)瘤細(xì)胞粘附和遷移的功能.AJAP1可以易位至細(xì)胞核,并通過(guò)其與β連環(huán)蛋白復(fù)合物的相互作用調(diào)節(jié)基因表達(dá).AJAP1的啟動(dòng)子甲基化在少突膠質(zhì)細(xì)胞中經(jīng)常發(fā)生,這直接導(dǎo)致了AJAP1的表達(dá)水平降低,而該基因的低表達(dá)與降低患者生存率下降直接相關(guān)[37-38].

      膠質(zhì)瘤侵襲也被分泌的蛋白酶(例如組織蛋白酶)和內(nèi)源性抑制劑(半胱氨酸蛋白酶抑制劑)之間的相互作用所調(diào)節(jié).半胱氨酸蛋白酶抑制劑E/M (CST6)是在神經(jīng)膠質(zhì)瘤中經(jīng)常過(guò)表達(dá)的組織蛋白酶B的強(qiáng)效抑制劑.研究表明,在膠質(zhì)瘤樣本中CST6被頻繁下調(diào),此種下調(diào)與CST6啟動(dòng)子甲基化有關(guān).在神經(jīng)膠質(zhì)瘤細(xì)胞系中過(guò)表達(dá)半胱氨酸蛋白酶抑制劑E/M會(huì)導(dǎo)致細(xì)胞運(yùn)動(dòng)和侵襲能力的下調(diào)[39-40].

      SLIT2是一個(gè)由細(xì)胞外基質(zhì)分泌的與膜相關(guān)的糖蛋白.SLIT2/ROBO1是一對(duì)保守的配體與受體.在神經(jīng)發(fā)育過(guò)程中,它們通過(guò)相互作用共同調(diào)節(jié)軸突和生長(zhǎng)錐之間的關(guān)系.在神經(jīng)膠質(zhì)瘤細(xì)胞系和腫瘤組織中SLIT2啟動(dòng)子經(jīng)常通過(guò)啟動(dòng)子甲基化下調(diào).用去甲基化試劑Aza處理SLIT2甲基化的細(xì)胞能夠恢復(fù)SLIT2基因的表達(dá).深層次的機(jī)制研究表明SLIT2主要通過(guò)SLIT2/ROBO1通路抑制CDC42?GTP,從而抑制膠質(zhì)瘤細(xì)胞的遷移和侵襲[41-42].

      4 DNA修復(fù)

      DNA修復(fù)基因可以修復(fù)在DNA復(fù)制過(guò)程中發(fā)生在微衛(wèi)星序列段的復(fù)制錯(cuò)誤[43].如果這些基因出現(xiàn)問(wèn)題,會(huì)使DNA修復(fù)出現(xiàn)問(wèn)題,導(dǎo)致遺傳的不穩(wěn)定性.目前已知一個(gè)在DNA損傷時(shí)被特異性激活錯(cuò)配修復(fù)酶MLH1,會(huì)在膠質(zhì)瘤中被甲基化沉默,而且研究表明MLH1啟動(dòng)子甲基化是膠質(zhì)瘤發(fā)生、發(fā)展以及克隆形成的早期事件[44].

      DNA修復(fù)酶MGMT可以拮抗烷化劑的毒性作用.MGMT啟動(dòng)子甲基化在膠質(zhì)瘤患者體內(nèi)十分常見(jiàn)且它是MGMT失活的一個(gè)關(guān)鍵機(jī)制.MGMT的表觀遺傳沉默對(duì)接受烷化劑化療的膠質(zhì)瘤患者是有利的[45-48].

      5 RAS途徑

      在膠質(zhì)瘤中一些信號(hào)通路也被DNA甲基化調(diào)控.在原發(fā)性腦腫瘤和神經(jīng)膠質(zhì)瘤細(xì)胞系中,Ras相關(guān)結(jié)構(gòu)域家族1A(RASSF1A)會(huì)被啟動(dòng)子甲基化失活.在所有的甲基化的細(xì)胞系中用去Aza處理后RASSF1A又可以被重新表達(dá).該基因主要通過(guò)MST1 或MOAP?1蛋白誘導(dǎo)細(xì)胞凋亡.研究表明,RASSF1A啟動(dòng)子CpG島甲基化可能在神經(jīng)膠質(zhì)瘤與髓母細(xì)胞瘤的發(fā)病機(jī)制中發(fā)揮重要作用[49-52].

      另一個(gè)N端RAS關(guān)聯(lián)域家族的基因,RAAS F10,在星形膠質(zhì)細(xì)胞瘤中經(jīng)常被甲基化.據(jù)報(bào)道,RASSF10在Ⅱ級(jí)和Ⅲ級(jí)星形細(xì)胞瘤以及Ⅳ級(jí)原發(fā)性膠質(zhì)母細(xì)胞瘤中被甲基化的頻率高達(dá)67.5%,但在Ⅰ級(jí)星形細(xì)胞瘤和年齡匹配的正常對(duì)照腦組織樣本DNA中卻未被甲基化.在甲基化的神經(jīng)膠質(zhì)瘤細(xì)胞種可以通過(guò)Aza去甲基化恢復(fù)RASSF10表達(dá).在Ⅱ級(jí)神經(jīng)膠質(zhì)瘤中,RASSF10的甲基化與生存率,總生存期是獨(dú)立相關(guān)的,并在其發(fā)展的早期階段就會(huì)出現(xiàn)甲基化.功能研究發(fā)現(xiàn),在兩種RAS F10甲基化的膠質(zhì)瘤細(xì)胞系中過(guò)表達(dá)RASSF10會(huì)抑制克隆形成,RASSF10敲除則會(huì)增加U87膠質(zhì)瘤細(xì)胞增殖[53].

      6 STAT信號(hào)轉(zhuǎn)導(dǎo)途徑

      JAK/STAT途徑參與幾種類(lèi)型癌癥的發(fā)生過(guò)程[54-55].細(xì)胞因子信號(hào)抑制3(SOCS3)可以通過(guò)抑制JAK/STAT信號(hào)通路而抑制腫瘤.報(bào)告發(fā)現(xiàn),在原發(fā)性GBM中經(jīng)常檢測(cè)到SOCS3啟動(dòng)子甲基化,同時(shí)會(huì)伴隨著表皮生長(zhǎng)因子受體(epidermal growth factor receptor,EGFR)擴(kuò)增與高表達(dá).SOCS3缺失會(huì)大大增加腫瘤細(xì)胞的浸潤(rùn)能力,但對(duì)腫瘤細(xì)胞增殖無(wú)明顯影響.在膠質(zhì)瘤中,SOCS3被甲基化后,可以激活STAT3和FAK,從而可以促進(jìn)腦膠質(zhì)瘤細(xì)胞的侵潤(rùn)[56-57].在膠質(zhì)瘤中SOCS1的過(guò)甲基化也有報(bào)道[56,58].將SOCS1重新導(dǎo)入膠質(zhì)瘤細(xì)胞可增加其對(duì)輻射所致?lián)p傷的敏感性[58].

      7 WNT信號(hào)途徑

      Wnt信號(hào)途徑的異常激活參與各種癌癥的發(fā)病過(guò)程,包括腦膠質(zhì)瘤[59].有相當(dāng)多的Wnt通路抑制劑被甲基化SFRP1,SFRP2和NKD2的甲基化有40%以上發(fā)生在原發(fā)性GBM中,而DKK1的甲基化有50%是發(fā)生在Ⅱ級(jí)GBM中.用去甲基化藥物處理SFRP1?,SFRP5?,DKK1?,DKK3?,NKD1?和NKD2高甲基化的U87?MG GBM細(xì)胞以后,它們的表達(dá)會(huì)增加[60].在神經(jīng)膠質(zhì)瘤中沉默Wnt信號(hào)的抑制劑導(dǎo)致Wnt途徑的激活,進(jìn)而引起腫瘤再生長(zhǎng).

      LATS2是一個(gè)抑癌基因,它通過(guò)破壞β連環(huán)蛋白/BCL9的相互作用來(lái)抑制致癌的Wnt/β?catenin介導(dǎo)的轉(zhuǎn)錄[61].在星形細(xì)胞瘤中LATS2經(jīng)常被甲基化,但在正常的腦組織中則不會(huì).LATS2在甲基化的星形細(xì)胞瘤中的mRNA水平要顯著明顯低于那些沒(méi)有甲基化的星形細(xì)胞瘤.在U251和SHG?44細(xì)胞系中檢測(cè)到LATS2甲基化,用Aza處理后LATS2表達(dá)恢復(fù)[62].LATS2的甲基化狀態(tài)為星形細(xì)胞瘤的診斷分析提供了重要的線索.

      8 臨床應(yīng)用

      膠質(zhì)瘤是一種最常見(jiàn)的原發(fā)性腦腫瘤,每年全世界大約有20萬(wàn)患者被診斷患膠質(zhì)瘤[63].現(xiàn)在很多可用的臨床治療手段如手術(shù)、放療和化療都取得了很大的進(jìn)步,但GBM患者的中位生存期僅為14.6個(gè)月,能夠有5年存活期的概率<5%[3-4,64-65].了解膠質(zhì)瘤的分子機(jī)制對(duì)于尋找新的治療策略至關(guān)重要.識(shí)別有用的生物標(biāo)志物將大大有利于膠質(zhì)瘤患者的早期診斷及提高治療效率.在膠質(zhì)瘤中許多抑癌基因的甲基化都與患者的預(yù)后密切相關(guān)(表1).

      表1 在膠質(zhì)瘤里啟動(dòng)子甲基化后沉默的抑癌基因的總結(jié)

      DNA異常甲基化在癌癥診斷上主要有以下三種應(yīng)用方式:①作為檢測(cè)癌細(xì)胞或檢測(cè)癌細(xì)胞DNA;②預(yù)測(cè)預(yù)后;③預(yù)測(cè)治療效果[66].DNA樣本比RNA樣品更穩(wěn)定,檢測(cè)啟動(dòng)子甲基化的生物標(biāo)記物更有優(yōu)勢(shì).通常來(lái)說(shuō),用于臨床檢驗(yàn)的樣品,如血液、尿、痰、唾液和糞便中,含有的腫瘤細(xì)胞DNA樣品是很少量的.由于DNA甲基化檢測(cè)技術(shù)的發(fā)展,即使含量很少,這些表觀遺傳學(xué)的標(biāo)記物也很容易被檢測(cè)到[66].且臨床上已經(jīng)能夠很好的檢測(cè)膠質(zhì)瘤患者血清樣品中MGMT和p16INK4a基因的啟動(dòng)子甲基化,以用于治療指導(dǎo)[67-69].

      腫瘤抑制因子啟動(dòng)子的甲基化也與膠質(zhì)瘤患者對(duì)藥物和放療的敏感性有關(guān)[70].例如,重新激活膠質(zhì)瘤中已經(jīng)沉默的SOCS1,會(huì)導(dǎo)致通過(guò)抑制MAPK來(lái)增加膠質(zhì)瘤對(duì)放療的敏感性[58].病毒介導(dǎo)的BEX1 或BEX2的再表達(dá)也可以增加腫瘤細(xì)胞對(duì)化療的敏感性,誘導(dǎo)細(xì)胞凋亡[71].這些基因甲基化的狀態(tài)還可以為患者特異性治療提供新的思路.眾所周知的例子是MGMT啟動(dòng)子甲基化和DNA烷化劑之間的關(guān)系.DNA烷化劑的機(jī)理是在DNA鄰近位置之間形成交聯(lián),從而抑制DNA的復(fù)制殺死細(xì)胞.MGMT能夠直接和特異性除去由這些烷基化劑在鳥(niǎo)嘌呤脫氧核糖核苷酸O6位置上形成的細(xì)胞毒性烷基加合物.激活的MGMT會(huì)抑制烷化劑的殺傷效率[72].因?yàn)樵趷盒阅[瘤中MGMT基因很少缺失或突變,所以MGMT的表觀遺傳學(xué)沉默可以做為臨床放療和烷化劑治療應(yīng)答的一個(gè)生物預(yù)測(cè)標(biāo)記.在若干臨床研究中,MGMT啟動(dòng)子甲基化的預(yù)測(cè)價(jià)值已經(jīng)被確認(rèn),并且在針對(duì)經(jīng)由放療和烷基化劑治療的膠質(zhì)瘤患者的研究中發(fā)現(xiàn),MGMT甲基化水平與顯著增長(zhǎng)的生存期密切相關(guān).

      總之,遺傳學(xué)和表觀遺傳學(xué)的改變對(duì)神經(jīng)膠質(zhì)瘤的發(fā)生都起到了重要作用.抑癌基因的沉默后將無(wú)法抑制膠質(zhì)瘤的發(fā)生和發(fā)展.目前已經(jīng)將抑癌基因啟動(dòng)子的甲基化作為生物記物,這將有助于膠質(zhì)瘤的診斷、預(yù)后與治療.在本研究中我們總結(jié)了最新鑒定的甲基化沉默的抑癌基因,還將進(jìn)一步研究它們的分子機(jī)制,以加深我們對(duì)這些生物標(biāo)記分子的認(rèn)識(shí)與了解并將它們更好地應(yīng)用在臨床上.

      【參考文獻(xiàn)】

      [1]Tu Y,Gao X,Li G,et al.MicroRNA?218 inhibits glioma invasion,migration,proliferation,and cancer stem?like cell self?renewal by targeting the polycomb group gene Bmi1[J].Cancer Res,2013,73(19):6046-6055.

      [2]Kaba SE,Kyritsis AP.Recognition and management of gliomas[J].Drugs,1997,53(2):235-244.

      [3]Louis DN.Molecular pathology of malignant gliomas[J].Annu Rev Pathol,2006,1:97-117.

      [4]Stupp R,Mason WP,van den Bent MJ,et al.Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma[J].N Engl J Med,2005,352(10):987-996.

      [5]Wang J,Su HK,Zhao HF,et al.Progress in the application of molecular biomarkers in gliomas[J].Biochem Biophys Res Com?mun,2015,465(1):1-4.

      [6]Cheng Y,Geng H,Cheng SH,et al.KRAB zinc finger protein ZNF382 is a proapoptotic tumor suppressor that represses multiple oncogenes and is commonly silenced in multiple carcinomas[J].Cancer Res,2010,70(16):6516-6526.

      [7]Coleman WB,Tsongalis GJ.Molecular mechanisms of human carci?nogenesis[J].EXS,2006(96):321-349.

      [8]Jones PA,Baylin SB.The fundamental role of epigenetic events in cancer[J].Nat Rev Genet,2002,3(6):415-428.

      [9]Shu XS,Li L,Ji M,et al.FEZF2,a novel 3p14 tumor suppressor gene,represses oncogene EZH2 and MDM2 expression and is frequently methylated in nasopharyngeal carcinoma[J].Carcinogene?sis,2013,34(9):1984-1993.

      [10]Cheng Y,Liang P,Geng H,et al.A novel 19q13 nucleolar zinc finger protein suppresses tumor cell growth through inhibiting ribo?some biogenesis and inducing apoptosis but is frequently silenced in multiple carcinomas[J].Mol Cancer Res,2012,10(7):925-936.

      [11]Wang S,Cheng Y,Du W,et al.Zinc?finger protein 545 is a novel tumour suppressor that acts by inhibiting ribosomal RNA transcription in gastric cancer[J].Gut,2013,62(6):833-841.

      [12]Fu L,Dong SS,Xie YW,et al.Down-regulation of tyrosine amin?otransferase at a frequently deleted region 16q22 contributes to the pathogenesis of hepatocellular carcinoma[J].Hepatology,2010,51(5):1624-1634.

      [13]Yu J,Liang QY,Wang J,et al.Zinc?finger protein 331,a novel putative tumor suppressor,suppresses growth and invasiveness of gastric cancer[J].Oncogene,2013,32(3):307-317.

      [14]He Z,Li B.Recent progress in genetic and epigenetic profile of diffuse gastric cancer[J].Cancer Transl Med,2015,1(3):80-93.

      [15]Qu Y,Dang S,Hou P.Gene methylation in gastric cancer[J].Clin Chim Acta,2013,424:53-65.

      [16]Watanabe T,Katayama Y,Yoshino A,et al.Aberrant hypermethyl?ation of p14ARF and O6?methylguanine?DNA methyltransferase genes in astrocytoma progression[J].Brain Pathol,2007,17(1):5-10.

      [17]Chin L,Pomerantz J,DePinho RA.The INK4a/ARF tumor suppres?sor:one gene?two products?two pathways[J].Trends Biochem Sci,1998,23(8):291-296.

      [18]Little M,Wainwright B.Methylation and p16:suppressing the sup?pressor[J].Nat Med,1995,1(7):633-634.

      [19]Arifin MT,Hama S,Kajiwara Y,et al.Cytoplasmic,but not nuclear,p16 expression may signal poor prognosis in high?grade astrocytomas[J].J Neurooncol,2006,77(3):273-277.

      [20]Fueyo J,Gomez?Manzano C,Bruner JM,et al.Hypermethylation of the CpG island of p16/CDKN2 correlates with gene inactivation in gliomas[J].Oncogene,1996,13(8):1615-1619.

      [21]Costello JF,Berger MS,Huang HS,et al.Silencing of p16/CDKN2 expression in human gliomas by methylation and chromatin condensa?tion[J].Cancer Res,1996,56(10):2405-2410.

      [22]Gonzalez?Gomez P,Bello MJ,Arjona D,et al.Promoter hypermeth?ylation of multiple genes in astrocytic gliomas[J].Int J Oncol,2003,22(3):601-608.

      [23]Berdasco M,Ropero S,Setien F,et al.Epigenetic inactivation of the Sotos overgrowth syndrome gene histone methyltransferase NSD1 in human neuroblastoma and glioma[J].Proc Natl Acad Sci U S A,2009,106(51):21830-21835.

      [24]Veeriah S,Brennan C,Meng S,et al.The tyrosine phosphatase PT?PRD is a tumor suppressor that is frequently inactivated and mutated in glioblastoma and other human cancers[J].Proc Natl Acad Sci U S A,2009,106(23):9435-9440.

      [25]Wei D,Gong W,Kanai M,et al.Drastic down?regulation of Krüppel?like factor 4 expression is critical in human gastric cancer development and progression[J].Cancer Res,2005,65(7):2746-2754.

      [26]Li L,Qin X,Shi M,et al.Regulation of histone acetylation by NDRG2 in glioma cells[J].J Neurooncol,2012,106(3):485-492.

      [27]Nakahara Y,Northcott PA,Li M,et al.Genetic and epigenetic inactivation of Kruppel?like factor 4 in medulloblastoma[J].Neopla?sia,2010,12(1):20-27.

      [28]Vermeulen K,Van Bockstaele DR,Berneman ZN.Apoptosis:mech?anisms and relevance in cancer[J].Ann Hematol,2005,84(10):627-639.

      [29]Darnay BG,Besse A,Poblenz AT,et al.TRAFs in RANK signaling[J].Adv Exp Med Biol,2007,597:152-159.

      [30]von dem Knesebeck A,F(xiàn)elsberg J,Waha A,et al.RANK(TN?FRSF11A)is epigenetically inactivated and induces apoptosis in gliomas[J].Neoplasia,2012,14(6):526-534.

      [31]Wu X,Li Y,Wan X,et al.Down?regulation of neogenin accelerated glioma progression through promoter Methylation and its overexpres?sion in SHG?44 Induced Apoptosis[J].PLoS One,2012,7(5):e38074.

      [32]Kadowaki M,Yoshioka H,Kamitani H,et al.DNA methylation?me?diated silencing of nonsteroidal anti?inflammatory drug?activated gene (NAG?1/GDF15)in glioma cell lines[J].Int J Cancer,2012,130(2):267-277.

      [33]Zhang Z,Wu L,Wang J,et al.Opposing effects of PI3K/Akt and Smad?dependent signaling pathways in NAG?1?induced glioblastoma cell apoptosis[J].PLoS One,2014,9(4):e96283.

      [34]Friedl P,Wolf K.Tumour?cell invasion and migration:diversity and escape mechanisms[J].Nat Rev Cancer,2003,3(5):362-374.

      [35]Costa AM,Pinto F,Martinho O,et al.Silencing of the tumor sup?pressor gene WNK2 is associated with upregulation of MMP2 and JNK in gliomas[J].Oncotarget,2015,6(3):1422-1434.

      [36]Moniz S,Martinho O,Pinto F,et al.Loss of WNK2 expression by promoter gene methylation occurs in adult gliomas and triggers Rac1?mediated tumour cell invasiveness[J].Hum Mol Genet,2013,22(1):84-95.

      [37]Cogdell D,Chung W,Liu Y,et al.Tumor?associated methylation of the putative tumor suppressor AJAP1 gene and association between decreased AJAP1 expression and shorter survival in patients with glioma[J].Chin J Cancer,2011,30(4):247-253.

      [38]Zeng L,F(xiàn)ee BE,Rivas MV,et al.Adherens junctional associated protein?1:a novel 1p36 tumor suppressor candidate in gliomas (Review)[J].Int J Oncol,2014,45(1):13-17.

      [39]Qiu J,Ai L,Ramachandran C,et al.Invasion suppressor cystatin E/M(CST6):high?level cell type?specific expression in normal brain and epigenetic silencing in gliomas[J].Lab Invest,2008,88(9):910-925.

      [40]Kim TY,Zhong S,F(xiàn)ields CR,et al.Epigenomic profiling reveals novel and frequent targets of aberrant DNA methylation?mediated silencing in malignant glioma[J].Cancer Res,2006,66(15):7490-7501.

      [41]Dallol A,Krex D,Hesson L,et al.Frequent epigenetic inactivation of the SLIT2 gene in gliomas[J].Oncogene,2003,22(29):4611-4616.

      [42]Yiin JJ,Hu B,Jarzynka MJ,et al.Slit2 inhibits glioma cell invasion in the brain by suppression of Cdc42 activity[J].Neuro Oncol,2009,11(6):779-789.

      [43]Duval A,Hamelin R.Genetic instability in human mismatch repair deficient cancers[J].Ann Genet,2002,45(2):71-75.

      [44]G?m?ri E,Pál J,Mészáros I,et al.Epigenetic inactivation of the hMLH1 gene in progression of gliomas[J].Diagn Mol Pathol,2007,16(2):104-107.

      [45]Berghoff AS,Hainfellner JA,Marosi C,et al.Assessing MGMT methylation status and its current impact on treatment in glioblastoma[J].CNS Oncol,2015,4(1):47-52.

      [46]Weller M,Stupp R,Reifenberger G,et al.MGMT promoter methyl?ation in malignant gliomas:ready for personalized medicine[J].Nat Rev Neurol,2010,6(1):39-51.

      [47]Kanemoto M,Shirahata M,Nakauma A,et al.Prognostic prediction of glioblastoma by quantitative assessment of the methylation status of the entire MGMT promoter region[J].BMC Cancer,2014,14:641.

      [48]Weller M.Assessing the MGMT status in glioblastoma:one step forward,two steps back[J].Neuro Oncol,2013,15(3):253-254.

      [49]Horiguchi K,Tomizawa Y,Tosaka M,et al.Epigenetic inactivation of RASSF1A candidate tumor suppressor gene at 3p21.3 in brain tumors[J].Oncogene,2003,22(49):7862-7865.

      [50]Gao Y,Guan M,Su B,et al.Hypermethylation of the RASSF1A gene in gliomas[J].Clin Chim Acta,2004,349(1-2):173-179.

      [51]Hesson L,Bièche I,Krex D,et al.Frequent epigenetic inactivation of RASSF1A and BLU genes located within the critical 3p21.3 region in gliomas[J].Oncogene,2004,23(13):2408-2419.

      [52]Hesson LB,Cooper WN,Latif F.The role of RASSF1A methylation in cancer[J].Dis Markers,2007,23(1-2):73-87.

      [53]Hill VK,Underhill?Day N,Krex D,et al.Epigenetic inactivation of the RASSF10 candidate tumor suppressor gene is a frequent and an early event in gliomagenesis[J].Oncogene,2011,30(8):978-989.

      [54]Bromberg JF,Wrzeszczynska MH,Devgan G,et al.Stat3 as an on?cogene[J].Cell,1999,98(3):295-303.

      [55]Wen Z,Zhong Z,Darnell JE Jr.Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation[J].Cell,1995,82(2):241-250.

      [56]Martini M,Pallini R,Luongo G,et al.Prognostic relevance of SOCS3 hypermethylation in patients with glioblastoma multiforme[J].Int J Cancer,2008,123(12):2955-2560.

      [57]Lindemann C,Hackmann O,Delic S,et al.SOCS3 promoter methy?lation is mutually exclusive to EGFR amplification in gliomas and promotes glioma cell invasion through STAT3 and FAK activation[J].Acta Neuropathol,2011,122(2):241-51.

      [58]Zhou H,Miki R,Eeva M,et al.Reciprocal regulation of SOCS 1 and SOCS3 enhances resistance to ionizing radiation in glioblastoma multiforme[J].Clin Cancer Res,2007,13(8):2344-2353.

      [59]Zhang K,Zhang J,Han L,et al.Wnt/beta?catenin signaling in glioma[J].J Neuroimmune Pharmacol,2012,7(4):740-749.

      [60]G?tze S,Wolter M,Reifenberger G,et al.Frequent promoter hyper?methylation of Wnt pathway inhibitor genes in malignant astrocytic gliomas[J].Int J Cancer,2010,126(11):2584-2593.

      [61]Li J,Chen X,Ding X,et al.LATS2 suppresses oncogenic Wnt sig?naling by disrupting β?catenin/BCL9 interaction[J].Cell Rep,2013,5(6):1650-1663.

      [62]Jiang Z,Li X,Hu J,et al.Promoter hypermethylation?mediated down?regulation of LATS1 and LATS2 in human astrocytoma[J].Neurosci Res,2006,56(4):450-458.

      [63]Parkin DM,Bray F,F(xiàn)erlay J,et al.Global cancer statistics,2002[J].CA Cancer J Clin,2005,55(2):74-108.

      [64]Kondo Y,Katsushima K,Ohka F,et al.Epigenetic dysregulation in glioma[J].Cancer Sci,2014,105(4):363-369.

      [65]Wen PY,Kesari S.Malignant gliomas in adults[J].N Engl J Med,2008,359(5):492-507.

      [66]Hu XT,He C.Recent progress in the study of methylated tumor suppressor genes in gastric cancer[J].Chin J Cancer,2013,32(1):31-41.

      [67]Balańa C,Carrato C,Ramírez JL,et al.Tumour and serum MGMT promoter methylation and protein expression in glioblastoma patients[J].Clin Transl Oncol,2011,13(9):677-685.

      [68]Liu BL,Cheng JX,Zhang W,et al.Quantitative detection of multi?ple gene promoter hypermethylation in tumor tissue,serum,and cerebrospinal fluid predicts prognosis of malignant gliomas[J].Neu?ro Oncol,2010,12(6):540-548.

      [69]Lavon I,Refael M,Zelikovitch B,et al.Serum DNA can define tumor?specific genetic and epigenetic markers in gliomas of various grades[J].Neuro Oncol,2010,12(2):173-180.

      [70]Natsume A,Kondo Y,Ito M,et al.Epigenetic aberrations and ther?apeutic implications in gliomas[J].Cancer Sci,2010,101(6):1331-1336.

      [71]Foltz G,Ryu GY,Yoon JG,et al.Genome?wide analysis of epige?netic silencing identifies BEX1 and BEX2 as candidate tumor sup?pressor genes in malignant glioma[J].Cancer Res,2006,66(13):6665-6674.

      [72]Day RS 3rd,Ziolkowski CH,Scudiero DA,et al.Defective repair of alkylated DNA by human tumour and SV40?transformed human cell strains[J].Nature,1980,288(5792):724-727.

      [73]Sibin MK,Bhat DI,Narasingarao KV,et al.CDKN2A(p16)mR?NA decreased expression is a marker of poor prognosis in malignant high?grade glioma[J].Tumour Biol,2015,36(10):7607-7614.

      [74]Zhou B,Tang Z,Deng Y,et al.Tumor suppressor candidate gene,NDRG2 is frequently inactivated in human glioblastoma multiforme[J].Mol Med Rep,2014,10(2):891-896.

      [75]Deng Y,Yao L,Chau L,et al.N?Myc downstream?regulated gene 2 (NDRG2)inhibits glioblastoma cell proliferation[J].Int J Cancer,2003,106(3):342-347.

      [76]Waha A,F(xiàn)elsberg J,Hartmann W,et al.Epigenetic downregulation of mitogen?activated protein kinase phosphatase MKP?2 relieves its growth suppressive activity in glioma cells[J].Cancer Res,2010,70(4):1689-1699.

      [77]Yang Y,Huang JQ,Zhang X,et al.MiR?129?2 functions as a tumor suppressor in glioma cells by targeting HMGB1 and is down?regulated by DNA methylation[J].Mol Cell Biochem,2015,404(1-2):229-239.

      [78]Dong X,Deng Q,Nie X,et al.Downregulation of HTATIP2 expres?sion is associated with promoter methylation and poor prognosis in glioma[J].Exp Mol Pathol,2015,98(2):192-199.

      [79]Chu SH,F(xiàn)eng DF,Ma YB,et al.Promoter methylation and down?regulation of SLC22A18 are associated with the development and pro?gression of human glioma[J].J Transl Med,2011,9:156.

      [80]Chu SH,Ma YB,F(xiàn)eng DF,et al.Effect of 5?Aza?2’?deoxycytidine on SLC22A18 in glioma U251 cells[J].Mol Med Rep,2012,5(1):138-141.

      [81]Alaminos M,Dávalos V,Ropero S,et al.EMP3,a myelin?related gene located in the critical 19q13.3 region,is epigenetically silenced and exhibits features of a candidate tumor suppressor in glioma and neuroblastoma[J].Cancer Res,2005,65(7):2565-2571.

      [82]Kunitz A,Wolter M,van den Boom J,et al.DNA hypermethylation and aberrant expression of the EMP3 gene at 19q13.3 in Human Gliomas[J].Brain Pathol,2007,17(4):363-370.

      [83]Watanabe T,Huang H,Nakamura M,et al.Methylation of the p73 gene in gliomas[J].Acta Neuropathol,2002,104(4):357-362.

      [84]Jancalek R.The role of the TP73 gene and its transcripts in neuro?oncology[J].Br J Neurosurg,2014,28(5):598-605.

      [85]Chilukamarri L,Hancock AL,Malik S,et al.Hypomethylation and aberrant expression of the glioma pathogenesis?related 1 gene in Wilms tumors[J].Neoplasia,2007,9(11):970-978.

      [86]Li L,Abdel Fattah E,Cao G,et al.Glioma pathogenesis?related protein 1 exerts tumor suppressor activities through proapoptotic reac?tive oxygen species?c?Jun?NH2 kinase signaling[J].Cancer Res,2008,68(2):434-443.

      [87]Mueller W,Nutt CL,Ehrich M,et al.Downregulation of RUNX3 and TES by hypermethylation in glioblastoma[J].Oncogene,2007,26(4):583-593.

      [88]Bai Y,Zhang QG,Wang XH.Downregulation of TES by hypermeth?ylation in glioblastoma reduces cell apoptosis and predicts poor clini?cal outcome[J].Eur J Med Res,2014,19:66.

      [89]Karakoula K,Jacques TS,Phipps KP,et al.Epigenetic genome?wide analysis identifies BEX1 as a candidate tumour suppressor gene in paediatric intracranial ependymoma[J].Cancer Lett,2014,346(1):34-44.

      [90]Zhou X,Meng Q,Xu X,et al.Bex2 regulates cell proliferation and apoptosis in malignant glioma cells via the c?Jun NH2?terminal kinase pathway[J].Biochem Biophys Res Commun,2012,427(3):574-580.

      [91]Zhou X,Xu X,Meng Q,et al.Bex2 is critical for migration and invasion in malignant glioma cells[J].J Mol Neurosci,2013,50(1):78-87.

      [92]Xu Y,Li WL,F(xiàn)u L,et al.Slit2/Robo1 signaling in glioma migra?tion and invasion.Neurosci Bull,2010,26(6):474-478.

      [93]Tivnan A,Zhao J,Johns TG,et al.The tumor suppressor microR?NA,miR?124a,is regulated by epigenetic silencing and by the tran?scriptional factor,REST in glioblastoma[J].Tumour Biol,2014,35(2):1459-1465.

      [94]Fowler A,Thomson D,Giles K,et al.miR?124a is frequently down?regulated in glioblastoma and is involved in migration and invasion[J].Eur J Cancer,2011,47(6):953-963.

      [95]Lu SH,Jiang XJ,Xiao GL,et al.miR?124a restoration inhibits glioma cell proliferation and invasion by suppressing IQGAP1 andβ?catenin[J].Oncol Rep,2014,32(5):2104-2110.

      [97]Gessler F,Voss V,Seifert V,et al.Knockdown of TFPI?2 promotes migration and invasion of glioma cells[J].Neurosci Lett,2011,497(1):49-54.

      [98]George J,Gondi CS,Dinh DH,et al.Restoration of tissue factor pathway inhibitor?2 in a human glioblastoma cell line triggers caspase?mediated pathway and apoptosis[J].Clin Cancer Res,2007,13(12):3507-3517.

      [99]Bertrand KC,Mack SC,Northcott PA,et al.PCDH10 is a candi?date tumour suppressor gene in medulloblastoma[J].Childs Nerv Syst,2011,27(8):1243-1249.

      [100]Echizen K,Nakada M,Hayashi T,et al.PCDH10 is required for the tumorigenicity of glioblastoma cells[J].Biochem Biophys Res Commun,2014,444(1):13-18.

      [101]Mei PJ,Bai J,Liu H,et al.RUNX3 expression is lost in glioma and its restoration causes drastic suppression of tumor invasion and migration[J].J Cancer Res Clin Oncol,2011,137(12):1823-1830.

      [102]Levesley J,Lusher ME,Lindsey JC,et al.RASSF1A and the BH3?only mimetic ABT?737 promote apoptosis in pediatric medulloblas?toma cell lines[J].Neuro Oncol,2011,13(12):1265-1276.

      [103]Kongkham PN,Northcott PA,Croul SE,et al.The SFRP family of WNT inhibitors function as novel tumor suppressor genes epigeneti?cally silenced in medulloblastoma[J].Oncogene,2010,29(20):3017-3024.

      [104]Schiefer L,Visweswaran M,Perumal V,et al.Epigenetic regula?tion of the secreted frizzled?related protein family in human glioblas?toma multiforme[J].Cancer Gene Ther,2014,21(7):297-303.

      [105]Vibhakar R,F(xiàn)oltz G,Yoon JG,et al.Dickkopf?1 is an epigenetically silenced candidate tumor suppressor gene in medulloblastoma[J].Neuro Oncol,2007,9(2):135-144.

      [106]Mizobuchi Y,Matsuzaki K,Kuwayama K,et al.REIC/Dkk?3 in?duces cell death in human malignant glioma[J].Neuro Oncol,2008,10(3):244-253.

      ·基礎(chǔ)與轉(zhuǎn)化醫(yī)學(xué)·

      通訊作者:張永生.教授,主任醫(yī)師,院長(zhǎng).E?mail:zhangys@fmmu.edu.cn;成迎端.博士.E?mail:cyd116@hotmail.com

      作者簡(jiǎn)介:涂艷陽(yáng).E?mail:ayonst@qq.com

      收稿日期:2016-01-05;接受日期:2016-01-22

      文章編號(hào):2095?6894(2016)02?01?07

      【中圖分類(lèi)號(hào)】R730.264

      【文獻(xiàn)標(biāo)識(shí)碼】A

      猜你喜歡
      膠質(zhì)瘤研究進(jìn)展
      MiRNA-145在消化系統(tǒng)惡性腫瘤中的研究進(jìn)展
      離子束拋光研究進(jìn)展
      獨(dú)腳金的研究進(jìn)展
      中成藥(2017年9期)2017-12-19 13:34:44
      TGIF2調(diào)控膠質(zhì)瘤細(xì)胞的增殖和遷移
      EVA的阻燃研究進(jìn)展
      3D-ASL在膠質(zhì)瘤術(shù)前分級(jí)的應(yīng)用價(jià)值
      DCE-MRI在高、低級(jí)別腦膠質(zhì)瘤及腦膜瘤中的鑒別診斷
      磁共振成像(2015年8期)2015-12-23 08:53:14
      P21和survivin蛋白在腦膠質(zhì)瘤組織中的表達(dá)及其臨床意義
      Sox2和Oct4在人腦膠質(zhì)瘤組織中的表達(dá)及意義
      肝衰竭的研究進(jìn)展
      襄垣县| 上林县| 乐陵市| 肥东县| 卓资县| 白水县| 盐边县| 叶城县| 萍乡市| 鄱阳县| 偏关县| 泰安市| 眉山市| 辉南县| 富宁县| 孝昌县| 南通市| 石河子市| 蕲春县| 贵港市| 鄂托克旗| 文登市| 于田县| 祥云县| 贵阳市| 河曲县| 固原市| 稻城县| 博乐市| 宜昌市| 忻城县| 金山区| 凤凰县| 闵行区| 龙州县| 潜山县| 廉江市| 英山县| 牙克石市| 延边| 普陀区|