汪小莉 李華 李東 徐成 林冰雪 陽泰 張濤
中圖分類號(hào) R285 文獻(xiàn)標(biāo)志碼 A 文章編號(hào) 1001-0408(2021)17-2164-07
DOI 10.6039/j.issn.1001-0408.2021.17.20
摘 要 目的:了解葫蘆素B的抗腫瘤作用機(jī)制,以期為該化合物的進(jìn)一步開發(fā)應(yīng)用提供理論參考。方法:以“葫蘆素B”“腫瘤”“癌癥”“cucurbitacin B”“CuB”“tumor”“cancer”等為關(guān)鍵詞,在中國知網(wǎng)、萬方數(shù)據(jù)、維普網(wǎng)、EBSCO、PubMed、Web of Science 等數(shù)據(jù)庫中組合查詢2000年1月-2021年6月發(fā)表的相關(guān)文獻(xiàn),對(duì)葫蘆素B抗腫瘤作用機(jī)制進(jìn)行總結(jié)歸納。結(jié)果與結(jié)論:葫蘆素B對(duì)多種惡性腫瘤均具有顯著的抗腫瘤作用,其作用機(jī)制主要是通過調(diào)控Janus激酶/信號(hào)轉(zhuǎn)導(dǎo)和轉(zhuǎn)錄激活因子3 (JAK/STAT3)、絲裂原活化蛋白激酶(MAPK)、磷脂酰肌醇激酶-3激酶/蛋白激酶B(PI3K/Akt)、Notch、Wnt、血管內(nèi)皮生長因子/黏附灶激酶/基質(zhì)金屬蛋白酶9(VEGF/FAK/MMP-9)等信號(hào)通路,進(jìn)而發(fā)揮抑制細(xì)胞增殖、阻滯細(xì)胞周期、促進(jìn)細(xì)胞凋亡、抑制細(xì)胞遷移和侵襲、抗血管生成、破壞細(xì)胞骨架、改變表觀遺傳、誘導(dǎo)自噬、誘導(dǎo)細(xì)胞衰老等作用。此外,葫蘆素B的抗腫瘤作用往往涉及多個(gè)腫瘤表型,是一種多靶點(diǎn)、多途徑的抗腫瘤藥物,其對(duì)信號(hào)通路的效應(yīng)具有腫瘤差異性。葫蘆素B還可以有效改善腫瘤細(xì)胞對(duì)阿霉素、吉非替尼、紫杉醇等藥物的耐藥現(xiàn)象,與臨床常用的抗腫瘤藥物如順鉑、多西他賽、吉西他濱等聯(lián)合使用也可極大地提高治療效果。
關(guān)鍵詞 葫蘆素B;腫瘤;作用機(jī)制;信號(hào)通路
癌變是一個(gè)多環(huán)節(jié)、多步驟的過程,每個(gè)階段都伴隨著不同的腫瘤表型變化,從而也預(yù)示著惡性腫瘤的治療具有多途徑、多靶點(diǎn)的特征。許多天然化學(xué)成分均被證明對(duì)多種腫瘤具有抑制細(xì)胞增殖、阻滯細(xì)胞周期、誘導(dǎo)細(xì)胞凋亡、抑制腫瘤血管生成、抑制細(xì)胞遷移和侵襲等作用,且在癌癥進(jìn)展過程中對(duì)多個(gè)靶點(diǎn)均具有顯著作用優(yōu)勢(shì)[1]。葫蘆素(cucurbitacin) 是一類從葫蘆科植物中分離提取的四環(huán)三萜類化合物,其母核結(jié)構(gòu)為四環(huán)三萜,具有高度氧化性[2]。據(jù)報(bào)道,葫蘆科家族(包括葫蘆科、苦瓜屬、黃瓜屬等)中至少有100種葫蘆素存在[3]。已知的葫蘆素及其衍生物主要有40種,按化學(xué)結(jié)構(gòu)分為葫蘆素A、B、C、D、E、I、H、Q、R和二氫葫蘆素B等12類[4]。其中,葫蘆素B是葫蘆素家族中含量最豐富的成員之一,具有抗炎、抗腫瘤、抗病毒、保肝、免疫調(diào)節(jié)等藥理作用[5]。且相關(guān)研究發(fā)現(xiàn),葫蘆素B對(duì)多種惡性腫瘤具有顯著的抗腫瘤作用,其作用機(jī)制涉及多個(gè)腫瘤表型和信號(hào)通路;另外,葫蘆素B還可改善腫瘤治療的耐藥現(xiàn)象,與多種抗腫瘤藥物聯(lián)合應(yīng)用可發(fā)揮協(xié)同增效的作用[6]?;诖?,筆者以“葫蘆素B”“腫瘤”“癌癥”“cucurbitacin B”“CuB”“tumor”“cancer”等為關(guān)鍵詞,在中國知網(wǎng)、萬方數(shù)據(jù)、維普網(wǎng)、EBSCO、PubMed、Web of Science 等數(shù)據(jù)庫中組合查詢2000年1月-2021年6月發(fā)表的相關(guān)文獻(xiàn),對(duì)葫蘆素B抗腫瘤作用機(jī)制的研究進(jìn)行綜述,以期為該化合物的進(jìn)一步開放應(yīng)用提供理論參考。
1 抑制細(xì)胞增殖
細(xì)胞增殖是生物體的重要生命特征,也是生物體生長、發(fā)育、繁殖和遺傳的基礎(chǔ)。惡性腫瘤是一種不受控制的增殖綜合征,因此,抑制腫瘤細(xì)胞增殖或生長是目前抗腫瘤藥物發(fā)揮作用的最常見機(jī)制之一。信號(hào)轉(zhuǎn)導(dǎo)和轉(zhuǎn)錄激活因子3(STAT3)是癌癥中異常激活的致癌基因,可介導(dǎo)Janus激酶(JAK)/STAT3信號(hào)通路[7]。Khan等[8]研究發(fā)現(xiàn),葫蘆素B對(duì)表皮生長因子受體(EGFR)野生型和突變型肺癌細(xì)胞均表現(xiàn)出抗增殖作用,其機(jī)制涉及抑制磷脂酰肌醇激酶-3激酶/哺乳動(dòng)物雷帕霉素靶蛋白(PI3K/mTOR)信號(hào)通路和STAT3信號(hào)通路。譚立君等[9]也研究發(fā)現(xiàn),葫蘆素B可通過抑制STAT3蛋白的表達(dá),引起細(xì)胞周期阻滯并誘導(dǎo)細(xì)胞凋亡,進(jìn)而抑制卵巢癌細(xì)胞的生長。黃琛[10]和張國建等[11]均研究發(fā)現(xiàn),葫蘆素B對(duì)結(jié)腸癌細(xì)胞的增殖抑制作用與STAT3活性有關(guān)。蛋白磷酸酶2A(CIP2A)是一種內(nèi)源性CIP2A(PP2A)抑制劑,能促進(jìn)多種實(shí)體腫瘤的生長和轉(zhuǎn)化,是腫瘤治療的潛在靶點(diǎn);肥大/干細(xì)胞生長因子受體(C-kit)屬于原癌基因C-kit編碼的Ⅲ型受體酪氨酸激酶家族成員,其可通過一系列反應(yīng)激活下游信號(hào)轉(zhuǎn)導(dǎo)通路,從而調(diào)節(jié)細(xì)胞的生長與增殖[12]。相關(guān)研究表明,葫蘆素B可通過下調(diào)CIP2A/PP2A/C-kit信號(hào)通路發(fā)揮抑制急性髓性白血病細(xì)胞生長和誘導(dǎo)細(xì)胞凋亡的作用[12]。整合素(integrins)是一種細(xì)胞黏附分子,具有聯(lián)系細(xì)胞外部與內(nèi)部結(jié)構(gòu)的作用,在復(fù)雜的多步驟惡性腫瘤進(jìn)展過程中扮演著重要角色[13]。Gupta等[2]研究發(fā)現(xiàn),葫蘆素B可通過integrin-人表皮生長因子受體2(HER2)信號(hào)通路抑制乳腺癌細(xì)胞生長。Shang等[14]研究發(fā)現(xiàn),葫蘆素B可通過上調(diào)磷酸酶-張力蛋白基因(PTEN),抑制Akt信號(hào)通路的激活,從而抑制神經(jīng)母細(xì)胞瘤細(xì)胞增殖。Aribi等[15]研究發(fā)現(xiàn),葫蘆素B聯(lián)合多西他賽或吉西他濱,可顯著抑制乳腺癌細(xì)胞增殖,降低乳腺癌異種移植瘤模型裸鼠的腫瘤體積。El-Senduny等[16]使用葫蘆素B對(duì)卵巢癌細(xì)胞進(jìn)行預(yù)處理,發(fā)現(xiàn)其可通過上調(diào)活性氧簇(ROS)水平,下調(diào)細(xì)胞外調(diào)節(jié)蛋白激酶1/2(pERK1/2)和磷酸化STAT3(p-STAT3)水平,顯著增加順鉑對(duì)卵巢癌細(xì)胞的毒性,從而抑制細(xì)胞的增殖。本課題組也研究發(fā)現(xiàn),葫蘆素B聯(lián)合索拉非尼可通過抑制STAT3磷酸化,進(jìn)而發(fā)揮協(xié)同抗肝癌細(xì)胞生長的效應(yīng)[17]。此外,本課題組還發(fā)現(xiàn),葫蘆素B可以抑制CD133+HepG2肝癌干細(xì)胞樣細(xì)胞的生長,其作用機(jī)制是通過下調(diào)STAT3磷酸化,進(jìn)而抑制細(xì)胞周期蛋白B1(Cyclin B1)、CDK1和CD133的表達(dá)[18]。
2 阻滯細(xì)胞周期
細(xì)胞周期是細(xì)胞生命活動(dòng)的基本過程,所有體細(xì)胞均通過細(xì)胞周期進(jìn)程決定的有絲分裂過程來進(jìn)行增殖[19]。靶向細(xì)胞周期,使細(xì)胞阻滯在某一時(shí)相從而阻斷細(xì)胞增殖,是目前腫瘤治療的研究熱點(diǎn)。研究表明,葫蘆素B可以通過抑制STAT3活性,阻滯多種腫瘤細(xì)胞(如胰腺癌Panc-1、Miapca-2細(xì)胞,白血病K562細(xì)胞,喉癌Hep-2細(xì)胞以及乳腺癌SKBR-3、MCF-7細(xì)胞)進(jìn)入G2/M期[3,20-21]。但也有部分研究指出,葫蘆素B阻滯細(xì)胞周期的作用機(jī)制不依賴于STAT3。例如Yasuda等[22]研究發(fā)現(xiàn),葫蘆素B對(duì)STAT3活性無影響,但可降低Cyclin B1、細(xì)胞分裂周期蛋白25C(CDC25C)的表達(dá),激活胱天蛋白酶(caspase)活性,進(jìn)而誘導(dǎo)結(jié)直腸癌細(xì)胞G2期阻滯和凋亡。Guo等[23]研究發(fā)現(xiàn),葫蘆素B不影響STAT3的激活,而是通過激活細(xì)胞周期檢測點(diǎn)激酶1-CDC25C-周期蛋白依賴性激酶1(Chk1-CDC25C-Cdk1)和p53-14-3- 3-σ信號(hào)通路,將肺癌細(xì)胞阻滯于G2/M期。據(jù)報(bào)道,其他信號(hào)通路也參與了葫蘆素B阻滯腫瘤細(xì)胞周期的作用。例如Dandawate等[24]研究發(fā)現(xiàn),葫蘆素B可通過抑制Notch信號(hào)通路,誘導(dǎo)G2/M期細(xì)胞周期阻滯。另有研究表明,葫蘆素B將細(xì)胞周期阻滯于不同時(shí)相的效應(yīng)還與腫瘤細(xì)胞的類型有關(guān)。例如Chan等[25]和Zhang等[26]研究發(fā)現(xiàn),葫蘆素B可分別誘導(dǎo)肝癌BEL-7402細(xì)胞和HepG2細(xì)胞發(fā)生S期阻滯;而Gao等[27]研究發(fā)現(xiàn),葫蘆素B可使前列腺癌細(xì)胞發(fā)生Sub-G0/G1期阻滯。這些研究為葫蘆素B的抗腫瘤作用機(jī)制提供了新的靶點(diǎn),同時(shí)也表明其阻滯細(xì)胞周期的作用和對(duì)信號(hào)通路的影響可因具體腫瘤或腫瘤細(xì)胞系的不同而有所差異。
3 誘導(dǎo)細(xì)胞凋亡
細(xì)胞凋亡和細(xì)胞增殖是維持機(jī)體細(xì)胞數(shù)量動(dòng)態(tài)平衡的基本機(jī)制。細(xì)胞凋亡減少是惡性腫瘤的標(biāo)志之一,因此細(xì)胞凋亡也成為目前抗腫瘤治療的常見靶點(diǎn)。Zhang等[28]指出,葫蘆素B可通過抑制STAT3的磷酸化,誘導(dǎo)胰腺癌細(xì)胞凋亡。何臣等[29]研究發(fā)現(xiàn),葫蘆素B可抑制STAT3信號(hào)通路的活化,下調(diào)Bcl-2蛋白表達(dá),從而誘導(dǎo)肺癌H1975細(xì)胞凋亡。Yar Saglam等[30]將葫蘆素B與吉非替尼聯(lián)合作用于結(jié)直腸癌細(xì)胞,發(fā)現(xiàn)可顯著抑制細(xì)胞生長并誘導(dǎo)細(xì)胞凋亡,其作用機(jī)制與調(diào)節(jié)EGFR和JAK/STAT信號(hào)通路相關(guān)。Ding等[31]指出,葫蘆素B聯(lián)合三氧化二砷誘導(dǎo)淋巴瘤Ramos細(xì)胞凋亡,其作用與抑制該細(xì)胞中STAT3的磷酸化有關(guān)。MAPK信號(hào)通路是參與細(xì)胞增殖、分化和凋亡的重要通路,p38MAPK、ERK和c-Jun氨基末端激酶(JNK)是MAPK家族的重要成員[32]。相關(guān)研究表明,葫蘆素B可通過影響STAT3和MAPK信號(hào)通路,誘導(dǎo)神經(jīng)母細(xì)胞瘤SH-SY5Y細(xì)胞和白血病K562細(xì)胞凋亡[33-34]。還有研究發(fā)現(xiàn),葫蘆素B可通過影響PI3K和MAPK信號(hào)通路,誘導(dǎo)細(xì)胞凋亡,最終抑制非小細(xì)胞肺癌細(xì)胞的生長[35]。馮杰[36]研究發(fā)現(xiàn),葫蘆素B聯(lián)合奧沙利鉑可協(xié)同抑制結(jié)直腸癌細(xì)胞的增殖并誘導(dǎo)其凋亡,其作用機(jī)制與抑制Akt/mTOR信號(hào)通路,下調(diào)Bcl-2/Bcl-2相關(guān)X蛋白(Bax)比值以及上調(diào)凋亡相關(guān)蛋白表達(dá)有關(guān)。除此之外,Qin等[37]研究發(fā)現(xiàn),葫蘆素B可通過CIP2A/PP2A/Akt信號(hào)通路,誘導(dǎo)膠質(zhì)瘤細(xì)胞凋亡。Klungsaeng等[38]研究發(fā)現(xiàn),葫蘆素B可通過黏附灶激酶(FAK)及其下游PI3K/Akt信號(hào)通路,促進(jìn)膽管癌KKU-100 CCA細(xì)胞凋亡。Dakeng等[39]研究發(fā)現(xiàn),葫蘆素B可阻斷Wnt信號(hào)通路并增加聚腺苷二磷酸核糖聚合酶(PARP)的裂解,從而誘導(dǎo)乳腺癌細(xì)胞凋亡。Bakar[40]研究發(fā)現(xiàn),葫蘆素B聯(lián)合伊馬替尼,可通過抑制基質(zhì)金屬蛋白酶2(MMP-2)表達(dá),誘導(dǎo)乳腺癌MCF-7細(xì)胞和結(jié)直腸癌SW480細(xì)胞的凋亡。還有研究報(bào)道,葫蘆素B可顯著誘導(dǎo)胃癌、骨肉瘤、前列腺癌細(xì)胞的凋亡[41-43]。以上研究表明,葫蘆素B可誘導(dǎo)多種腫瘤細(xì)胞發(fā)生細(xì)胞凋亡,并且其調(diào)控細(xì)胞凋亡的分子機(jī)制較為復(fù)雜。
4 抑制細(xì)胞遷移和侵襲
腫瘤細(xì)胞向周圍組織和血管的遷移和侵襲是腫瘤轉(zhuǎn)移的第一步,而腫瘤轉(zhuǎn)移又是導(dǎo)致癌癥患者死亡的主要原因,因此尋求可以有效抑制腫瘤細(xì)胞遷移和侵襲的治療藥物具有重要意義。Chai等[44]和周彤等[45]均研究發(fā)現(xiàn),葫蘆素B可通過Hippo-YAP信號(hào)通路發(fā)揮抑制結(jié)直腸細(xì)胞增殖和侵襲的作用。Shukla等[46]觀察到葫蘆素B對(duì)轉(zhuǎn)移性非小細(xì)胞肺癌表現(xiàn)出很強(qiáng)的抗遷移、抗侵襲能力和顯著的抑制腫瘤血管生成的作用,其機(jī)制可能與Wnt/β-聯(lián)蛋白(Wnt/β-catenin)信號(hào)通路有關(guān)。Zhang等[42]研究發(fā)現(xiàn),葫蘆素B可通過調(diào)控MAPK、JAK2/STAT3信號(hào)通路進(jìn)而抑制MMP-2和MMP-9活性,最終抑制骨肉瘤細(xì)胞的遷移。喻婷婷等[47]研究發(fā)現(xiàn),葫蘆素B抑制骨肉瘤細(xì)胞侵襲的機(jī)制可能與抑制Akt信號(hào)通路的活性和上皮間質(zhì)轉(zhuǎn)化(EMT)的發(fā)生有關(guān)。Gupta等[48]研究發(fā)現(xiàn),葫蘆素B可通過抑制HER2介導(dǎo)的EMT,從而抑制乳腺癌腦轉(zhuǎn)移。Luo等[49]報(bào)道,葫蘆素B可通過FAK途徑抑制ROS依賴的乳腺癌細(xì)胞轉(zhuǎn)移。Promkan等[50]研究發(fā)現(xiàn),葫蘆素B可抑制乳腺癌1號(hào)基因(BRCA1)缺陷型乳腺癌細(xì)胞的增殖、遷移和侵襲。梁菁[51]研究發(fā)現(xiàn),葫蘆素B可有效抑制乳腺癌細(xì)胞黏附、遷移、侵襲,其機(jī)制可能與抑制波形蛋白/F-肌動(dòng)蛋白/FAK/黏著斑蛋白(Vimentin/F-actin/FAK/Vinculin)的表達(dá)和RAC1/CDC42/RhoA的信號(hào)轉(zhuǎn)導(dǎo)有關(guān)。
5 抗血管生成
腫瘤血管生成是一個(gè)極其復(fù)雜的過程,一般包括血管內(nèi)皮基質(zhì)降解、內(nèi)皮細(xì)胞的移行、增殖和管道化分支形成血管環(huán)和形成新的基底膜等步驟,而腫瘤細(xì)胞和內(nèi)皮細(xì)胞的相互作用自始至終貫穿于腫瘤血管生成的全過程[52]。腫瘤缺氧是許多癌癥的共同特征,其中缺氧誘導(dǎo)因子1 (HIF-1)可刺激血管內(nèi)皮生長因子(VEGF)等促血管生成基因的表達(dá),進(jìn)而刺激新生血管生成以增加向缺氧腫瘤組織的血供,所以缺氧和HIF對(duì)血管生成的調(diào)控至關(guān)重要[53]。由于腫瘤血管生成可進(jìn)一步引起腫瘤的生長和轉(zhuǎn)移,因此抗血管生成也是抗腫瘤的重要靶點(diǎn)。Piao等[54]在體外研究中發(fā)現(xiàn),葫蘆素B可顯著抑制人臍靜脈內(nèi)皮細(xì)胞(HUVEC)的增殖、遷移和小管生成;進(jìn)一步在雞胚絨毛膜尿囊膜實(shí)驗(yàn)中發(fā)現(xiàn),葫蘆素B可阻斷血管生成,且其抗血管生成機(jī)制與抑制VEGF受體的活性有關(guān)。Sinha等[55]研究發(fā)現(xiàn),葫蘆素B可通過下調(diào)VEGF/FAK/MMP-9信號(hào)通路抑制高轉(zhuǎn)移性乳腺癌細(xì)胞的遷移和血管形成。此外,Touihri-Barakati等[56]研究發(fā)現(xiàn),葫蘆素B不僅可抑制人膠質(zhì)母細(xì)胞瘤U87細(xì)胞增殖,還可有效抑制體外人微血管內(nèi)皮細(xì)胞(HMEC)的血管生成,其機(jī)制與抑制integrins有關(guān),揭示了其作為一種特異性抗integrins藥物的潛在價(jià)值。由此可見,葫蘆素B不僅可直接抑制腫瘤細(xì)胞的生長,還可通過抗血管生成進(jìn)而抑制腫瘤細(xì)胞遷移。
6 破壞細(xì)胞骨架
細(xì)胞骨架是真核細(xì)胞維持其基本形態(tài)的重要結(jié)構(gòu),主要包含微管、微絲和中間纖維等結(jié)構(gòu),破壞細(xì)胞骨架可擾亂細(xì)胞的多種生理活動(dòng)[57]。Liang等[58]研究發(fā)現(xiàn),葫蘆素B可以通過RAC1/CDC42/RhoA信號(hào)通路介導(dǎo)細(xì)胞骨架蛋白的重組和分布,改善細(xì)胞黏附和變形的機(jī)械性能,從而抑制乳腺癌細(xì)胞遷移和侵襲。Wakimoto等[59]研究發(fā)現(xiàn),葫蘆素B可引起乳腺癌細(xì)胞微管和肌動(dòng)蛋白破壞,并推測葫蘆素B可能是除紫杉類藥物和長春堿類藥物以外的第三類以微管為靶點(diǎn)的藥物。還有研究發(fā)現(xiàn),葫蘆素B可引起膠質(zhì)母細(xì)胞瘤細(xì)胞和肺癌細(xì)胞發(fā)生明顯的形態(tài)學(xué)變化,并破壞這兩種細(xì)胞的肌動(dòng)蛋白和微管,從而破壞細(xì)胞骨架[60-61]。另外,Yang等[62]研究發(fā)現(xiàn),葫蘆素B可誘導(dǎo)多發(fā)性骨髓瘤細(xì)胞的絲切蛋白(cofilin)去磷酸化,使肌動(dòng)蛋白的解聚活性增強(qiáng),從而破壞細(xì)胞骨架。由此可見,葫蘆素B可破壞多種腫瘤細(xì)胞的骨架,并且具有開發(fā)為第三類抗微管重聚合類抗腫瘤藥物的潛力。
7 改變表觀遺傳
表觀遺傳是指獨(dú)立于DNA序列的遺傳分子決定因素,其特征包括DNA甲基化、組蛋白修飾、非編碼RNA和染色質(zhì)結(jié)構(gòu)[63]。近年來研究發(fā)現(xiàn),包括啟動(dòng)子異常甲基化在內(nèi)的表觀遺傳變化在人類惡性腫瘤發(fā)生發(fā)展中具有重要作用[64]。Mao等[65]研究發(fā)現(xiàn),葫蘆素B可通過激活B細(xì)胞易位基因(BTG3),抑制結(jié)腸癌細(xì)胞增殖并誘導(dǎo)其凋亡。Shukla等[66]研究發(fā)現(xiàn),葫蘆素B可通過抑制DNA甲基轉(zhuǎn)移酶(DNMT)和組蛋白去乙?;福℉DAC)活性,激活腫瘤抑癌基因(TSG),并下調(diào)致癌基因c-Myc、K-RAS以及腫瘤啟動(dòng)子基因(TPG)。Dittharot等[67]也發(fā)現(xiàn),葫蘆素B可使c-Myc、Cyclin D1和生存素(survivin)的啟動(dòng)子發(fā)生甲基化,并下調(diào)其表達(dá),從而抑制乳腺癌細(xì)胞增殖。由此說明,葫蘆素B可通過誘導(dǎo)致癌基因啟動(dòng)子甲基化等一系列表觀遺傳修飾來改變腫瘤相關(guān)基因的表達(dá),最終抑制腫瘤細(xì)胞增殖或誘導(dǎo)腫瘤細(xì)胞凋亡。
8 誘導(dǎo)自噬
細(xì)胞自噬是指細(xì)胞在外界環(huán)境因素的影響下,利用溶酶體降解自身受損、變性或衰老的大分子物質(zhì)以及細(xì)胞器的自我消化過程。研究發(fā)現(xiàn),自噬在腫瘤中發(fā)揮著“雙刃劍”的作用:一方面自噬限制了氧化應(yīng)激、慢性組織損傷和致癌信號(hào)傳導(dǎo),進(jìn)而抑制腫瘤的發(fā)生;另一方面,自噬能在不利環(huán)境中提高腫瘤細(xì)胞對(duì)應(yīng)激的耐受能力,進(jìn)而維持腫瘤的生長[68]。Niu等[69]研究發(fā)現(xiàn),葫蘆素B不僅可誘導(dǎo)肝癌BEL-7402細(xì)胞DNA損傷和凋亡,還可通過調(diào)控Akt/mTOR信號(hào)通路誘導(dǎo)細(xì)胞發(fā)生保護(hù)性自噬;當(dāng)添加自噬抑制劑3-MA和CQ后,可增強(qiáng)葫蘆素B對(duì)細(xì)胞死亡的誘導(dǎo)作用,進(jìn)一步提示葫蘆素B誘導(dǎo)肝癌細(xì)胞發(fā)生的自噬是保護(hù)性自噬。Zhu等[70]也發(fā)現(xiàn),葫蘆素B不僅可以抑制白血病Jurkat細(xì)胞增殖,誘導(dǎo)細(xì)胞G2/M期阻滯和凋亡,還通過上調(diào)自噬標(biāo)志物輕鏈3-Ⅱ(LC3-Ⅱ)的表達(dá)進(jìn)而誘導(dǎo)細(xì)胞自噬;此外,該研究還發(fā)現(xiàn)抑制葫蘆素B誘導(dǎo)的自噬顯著增加了caspase-3的激活,提示葫蘆素B誘導(dǎo)的自噬可能是一種促生存反應(yīng)。還有研究報(bào)道,葫蘆素B可抑制CIP2A/PP2A/mTORC1信號(hào)通路,誘導(dǎo)順鉑耐藥的人胃癌SGC7901/DDP細(xì)胞發(fā)生自噬,進(jìn)而抑制該胃癌細(xì)胞的生長[71-72]。目前,在乳腺癌領(lǐng)域也有較多的關(guān)于葫蘆素B與自噬關(guān)系的研究。例如Ren等[73]研究發(fā)現(xiàn),葫蘆素B能引起乳腺癌MCF-7細(xì)胞自噬空泡的形成,并上調(diào)LC3-Ⅱ和自噬調(diào)控蛋白(Beclin-1)的表達(dá),從而表現(xiàn)出顯著的抗腫瘤作用。又如黃擎擎[74]研究發(fā)現(xiàn),葫蘆素B除抑制乳腺癌MCF-7細(xì)胞增殖和誘導(dǎo)其凋亡外,還可明顯升高LC3-Ⅱ/LC3-Ⅰ的比值,降低p62蛋白表達(dá)水平,提示葫蘆素B可能引起乳腺癌細(xì)胞發(fā)生自噬。此外,Kurman等[75]研究發(fā)現(xiàn),葫蘆素B與順鉑聯(lián)用可通過PI3K/Akt/mTOR信號(hào)通路誘導(dǎo)膀胱癌細(xì)胞的自噬,進(jìn)而抑制細(xì)胞增殖。由此可見,葫蘆素B誘導(dǎo)的自噬到底對(duì)腫瘤起保護(hù)性作用還是抑制性作用,與腫瘤的種類有關(guān)。
9 誘導(dǎo)細(xì)胞衰老
在癌癥發(fā)展過程中,衰老是阻止腫瘤發(fā)生的有力屏障,因此,誘導(dǎo)癌細(xì)胞衰老,或通過藥物干預(yù)衰老,已成為目前研究領(lǐng)域的熱點(diǎn)[76]。端粒是位于染色體末端的核蛋白結(jié)構(gòu),其功能是維持基因組的穩(wěn)定性,端粒的丟失也與體內(nèi)的衰老過程相關(guān)[77]。端粒酶是一種負(fù)責(zé)延長端粒的酶,抑制其活性可使端粒變短,從而導(dǎo)致細(xì)胞不再分裂,進(jìn)而使細(xì)胞及器官進(jìn)入衰老狀態(tài),因此,抑制端粒酶可作為癌癥的治療靶點(diǎn)。Duangmano等[78]研究表明,葫蘆素B可抑制3種乳腺癌細(xì)胞的生長,其作用機(jī)制可能與下調(diào)人類端粒酶逆轉(zhuǎn)錄酶(hTERT)和c-Myc的表達(dá)有關(guān)。另外,葫蘆素B聯(lián)合Withanone(一種環(huán)氧甾體內(nèi)酯)可通過上調(diào)核纖層蛋白A/C(Lamin A/C)、視網(wǎng)膜細(xì)胞瘤抑癌蛋白(RB)、分子伴侶(mortalin)和p53等蛋白的表達(dá),進(jìn)而誘導(dǎo)非小細(xì)胞肺癌細(xì)胞衰老[79]。
10 改善腫瘤耐藥
近年來,雖然抗腫瘤藥物極大提高了癌癥患者的生存時(shí)間,但腫瘤細(xì)胞對(duì)化療或靶向藥物的耐藥也已成為普遍現(xiàn)象,而耐藥又會(huì)導(dǎo)致腫瘤復(fù)發(fā)[80]。相關(guān)研究發(fā)現(xiàn),葫蘆素B能逆轉(zhuǎn)耐阿霉素乳腺癌MCF-7細(xì)胞的耐藥,其作用機(jī)制可能與抑制CIP2A從而增強(qiáng)PP2A活性有關(guān)[81-82]。還有研究報(bào)道,葫蘆素B可通過誘導(dǎo)EGFR溶酶體降解和下調(diào)CIP2A/PP2A/Akt信號(hào)通路來抑制吉非替尼耐藥肺癌細(xì)胞的生長和侵襲[83-84]。與此不同的是,曾超朋[85]研究發(fā)現(xiàn),葫蘆素B抑制吉非替尼耐藥肺癌細(xì)胞增殖的作用機(jī)制與抑制JAK2/STAT3信號(hào)轉(zhuǎn)導(dǎo)通路有關(guān)。另有研究發(fā)現(xiàn),葫蘆素B還可通過抑制CIP2A/PP2A/mTORC1信號(hào)通路,抑制順鉑耐藥胃癌細(xì)胞的增殖,并誘導(dǎo)caspase依賴性細(xì)胞凋亡和自噬[71-72]。Qu等[86]研究發(fā)現(xiàn),葫蘆素B可誘導(dǎo)紫杉醇耐藥卵巢癌細(xì)胞的周期阻滯,進(jìn)而促進(jìn)該細(xì)胞凋亡,其作用機(jī)制與增強(qiáng)p53和p21蛋白的表達(dá)以及下調(diào)p-糖蛋白(P-gp)的表達(dá)有關(guān)。
綜上所述,葫蘆素B對(duì)多種惡性腫瘤具有廣譜的抗腫瘤作用,其作用機(jī)制主要是通過調(diào)控JAK/STAT3、MARK、PI3K/Akt、Notch、Wnt、CIP2A/PP2A、integrin- HER2、Hippo-YAP、VEGF/FAK/MMP-9等信號(hào)通路,發(fā)揮抑制細(xì)胞增殖、阻滯細(xì)胞周期、促進(jìn)細(xì)胞凋亡、抑制細(xì)胞遷移和侵襲、抗血管生成、破壞細(xì)胞骨架、改變表觀遺傳、誘導(dǎo)自噬、誘導(dǎo)細(xì)胞衰老等作用。此外,葫蘆素B的抗腫瘤作用往往涉及多個(gè)腫瘤表型,是一種多靶點(diǎn)、多途徑的抗腫瘤藥物,其對(duì)信號(hào)通路的效應(yīng)具有腫瘤差異性。值得注意的是,葫蘆素B還可以有效改善腫瘤細(xì)胞對(duì)阿霉素、吉非替尼和紫杉醇等藥物的耐藥現(xiàn)象,與臨床常用的抗腫瘤藥物如順鉑、多西他賽、吉西他濱等聯(lián)合使用也可極大地提高治療效果。由此表明,葫蘆素B是一種具有極大潛力的抗腫瘤天然化合物,值得進(jìn)一步的開發(fā)和研究。
參考文獻(xiàn)
[ 1 ] RANJAN A,F(xiàn)OFARIA N M,KIM S H,et al. Modulation of signal transduction pathways by natural compounds in cancer[J]. Chin J Nat Med,2015,13(10):730-742.
[ 2 ] GUPTA P,SRIVASTAVA S K. Inhibition of integrin-HER2 signaling by cucurbitacin B leads to in vitro and in vivo breast tumor growth suppression[J]. Oncotarget,2014,5(7):1812-1828.
[ 3 ] CAI Y,F(xiàn)ANG X,HE C,et al. Cucurbitacins:a systematic review of the phytochemistry and anticancer activity[J]. Am J Chin Med,2015,43(7):1331-1350.
[ 4 ] ALGHASHAM A A. Cucurbitacins:a promising target for cancer therapy[J]. Int J Health Sci,2013,7(1):77-89.
[ 5 ] LEE D H,IWANSKI G B,THOENNISSEN N H. Cucurbitacin:ancient compound shedding new light on cancer treatment[J]. Scientific World Journal,2010,10:413-418.
[ 6 ] KALIMUTHU S,GANGADARAN P,OH J M,et al. A new tyrosine kinase inhibitor K905-0266 inhibits prolifera- tion and sphere formation of glioblastoma cancer cells[J]. J Drug Target,2020,28(9):933-938.
[ 7 ] GUANIZO A C,F(xiàn)ERNANDO C D,GARAMA D J,et al. STAT3:a multifaceted oncoprotein[J]. Growth Factors,2018,36(1/2):1-14.
[ 8 ] KHAN N,JAJEH F,KHAN M I,et al. Sestrin-3 modulation is essential for therapeutic efficacy of cucurbitacin B in lung cancer cells[J]. Carcinogenesis,2017,38(2):184- 195.
[ 9 ] 譚立君,張大昕,閆志鈞,等.葫蘆素B對(duì)卵巢癌細(xì)胞SKOV3的生長抑制作用[J].腫瘤學(xué)雜志,2017,23(7):598-601.
[10] 黃琛.葫蘆素B對(duì)于結(jié)腸癌細(xì)胞生長與增殖抑制作用的研究[D].石家莊:河北醫(yī)科大學(xué),2015.
[11] 張國建,閻慶輝,黃琛,等.葫蘆素B對(duì)人結(jié)腸癌細(xì)胞株LS174-T的生長抑制作用[J].中華實(shí)驗(yàn)外科雜志,2014,31(12):2751-2753.
[12] MA W,XIANG Y,YANG R,et al. Cucurbitacin B induces inhibitory effects via the CIP2A/PP2A/C-KIT signaling axis in t(8;21) acute myeloid leukemia[J]. J Pharmacol Sci,2019,139(4):304-310.
[13] YOUSEFI H,VATANMAKANIAN M,MAHDIANNASSER M,et al. Understanding the role of integrins in breast cancer invasion,metastasis,angiogenesis,and drug resistance[J]. Oncogene,2021,40(6):1043-1063.
[14] SHANG Y,GUO X X,LI W W,et al. Cucurbitacin B inhibits neuroblastoma cell proliferation through up-regulation of PTEN[J]. Eur Rev Med Pharmacol Sci,2014,18(21):3297-3303.
[15] ARIBI A,GERY S,LEE D H,et al. The triterpenoid cucurbitacin B augments the antiproliferative activity of chemotherapy in human breast cancer[J]. Int J Cancer,2013,132(12):2730-2737.
[16] EL-SENDUNY F F,BADRIA F A,EL-WASEEF A M,et al. Approach for chemosensitization of cisplatin-resistant ovarian cancer by cucurbitacin B[J]. Tumour Biol,2016,37(1):685-698.
[17] WANG X,LI H,LI D,et al. Sorafenib and CuB exert? ?synergistic antitumor effects against hepatocellular carcinoma cells via inhibition of STAT3 phosphorylation[J]. FEBS Open Bio,2021,11(1):133-145.
[18] WANG X,BAI Y,YAN X,et al. Cucurbitacin B exhibits antitumor effects on CD133+HepG2 liver cancer stem cells by inhibiting JAK2/STAT3 signaling pathway[J]. Anticancer Drugs,2021,32(5):548-557.
[19] ALENZI F Q. Links between apoptosis,proliferation and the cell cycle[J]. Br J Biomed Sci,2004,61(2):99-102.
[20] XU J,CHEN Y,YANG R,et al. Cucurbitacin B inhibits gastric cancer progression by suppressing STAT3 activity[J]. Arch Biochem Biophys,2020,684:108314.
[21] GARG S,KAUL S C,WADHWA R. Cucurbitacin B and cancer intervention:chemistry,biology and mechanisms:review[J]. Int J Oncol,2018,52(1):19-37.
[22] YASUDA S,YOGOSAWA S,IZUTANI Y,et al. Cucurbi- tacin B induces G2 arrest and apoptosis via a reactive oxygen species-dependent mechanism in human colon adenocarcinoma SW480 cells[J]. Mol Nutr Food Res,2010,54(4):559-565.
[23] GUO J,WU G,BAO J,et al. Cucurbitacin B induced ATM-mediated DNA damage causes G2/M cell cycle arrest in a ROS-dependent manner[J]. PloS One,2014,9(2):e88140.
[24] DANDAWATE P,SUBRAMANIAM D,PANOVICH P,et al. Cucurbitacin B and I inhibits colon cancer growth by targeting the Notch signaling pathway[J]. Sci Rep,2020,10(1):1290.
[25] CHAN K T,MENG F Y,LI Q,et al. Cucurbitacin B induces apoptosis and S phase cell cycle arrest in BEL-7402 human hepatocellular carcinoma cells and is effective via oral administration[J]. Cancer Lett,2010,294(1):118- 124.
[26] ZHANG M,ZHANG H,SUN C,et al. Targeted constitutive activation of signal transducer and activator of transcription 3 in human hepatocellular carcinoma cells by cucurbitacin B[J]. Cancer Chemother Pharmacol,2009,63(4):635-642.
[27] GAO Y,ISLAM M S,TIAN J,et al. Inactivation of ATP citrate lyase by cucurbitacin B:a bioactive compound from cucumber,inhibits prostate cancer growth[J]. Cancer Lett,2014,349(1):15-25.
[28] ZHANG M,SUN C,SHAN X,et al. Inhibition of pan- creatic cancer cell growth by cucurbitacin B through? ?modulation of signal transducer and activator of transcription 3 signaling[J]. Pancreas,2010,39(6):923-929.
[29] 何臣,王菊輝,丘韶校,等.葫蘆素B抑制STAT3/Bcl-2表達(dá)誘導(dǎo)非小細(xì)胞肺癌H1975細(xì)胞凋亡[J].中國實(shí)用醫(yī)藥,2020,15(13):196-198.
[30] YAR SAGLAM A S,ALP E,ELMAZOGLU Z,et al. Treatment with cucurbitacin B alone and in combination with gefitinib induces cell cycle inhibition and apoptosis via EGFR and JAK/STAT pathway in human colorectal cancer cell lines[J]. Hum Exp Toxicol,2016,35(5):526- 543.
[31] DING X,CHI J,YANG X,et al. Cucurbitacin B synergistically enhances the apoptosis-inducing effect of arsenic trioxide by inhibiting STAT3 phosphorylation in lymphoma Ramos cells[J]. Leuk Lymphoma,2017,58(10):2439-2451.
[32] KIM E K,CHOI E J. Compromised MAPK signaling in human diseases:an update[J]. Arch Toxicol,2015,89(6):867-882.
[33] 鄭倩.葫蘆素B對(duì)神經(jīng)母細(xì)胞瘤SH-SY5Y細(xì)胞的抑制作用機(jī)制研究[D].武漢:華中科技大學(xué),2014.
[34] CHAN K T,LI K,LIU S L,et al. Cucurbitacin B inhibits STAT3 and the Raf/MEK/ERK pathway in leukemia cell line K562[J]. Cancer Lett,2010,289(1):46-52.
[35] SILVA I T,GELLER F C,PERSICH L,et al. Cytotoxic effects of natural and semisynthetic cucurbitacins on lung cancer cell line A549[J]. Invest? New? Drugs,2016,34(2):139-148.
[36] 馮杰.葫蘆素B聯(lián)合奧沙利鉑調(diào)控AKT/mTOR通路誘導(dǎo)結(jié)腸癌細(xì)胞SW480凋亡的影響[D].錦州:錦州醫(yī)科大學(xué),2019.
[37] QIN S,LI J,SI Y,et al. Cucurbitacin B induces inhibitory effects via CIP2A/PP2A/Akt pathway in glioblastoma multiforme[J]. Mol Carcinog,2018,57(6):687-699.
[38] KLUNGSAENG S,KUKONGVIRIYAPAN V,PRAWAN A,et al. Cucurbitacin B induces mitochondrial-mediated apoptosis pathway in cholangiocarcinoma cells via suppressing focal adhesion kinase signaling[J]. Naunyn Schmiedebergs Arch Pharmacol,2019,392(3):271-278.
[39] DAKENG S,DUANGMANO S,JIRATCHARIYAKUL W,et al. Inhibition of Wnt signaling by cucurbitacin B in breast cancer cells:reduction of Wnt-associated proteins and reduced translocation of galectin-3-mediated β-catenin to the nucleus[J]. J Cell Biochem,2012,113(1):49-60.
[40] BAKAR F. Cucurbitacin B enhances the anticancer effect of imatinib mesylate through inhibition of MMP-2 expres- sion in MCF-7 and SW480 tumor cell lines[J]. Anticancer Agents Med Chem,2016,16(6):747-754.
[41] 張萌,邊志剛,何平.葫蘆素B對(duì)人胃癌BGC-823細(xì)胞增殖及凋亡的影響[J].實(shí)用藥物與臨床,2016,19(5):548- 552.
[42] ZHANG Z R,GAO M X,YANG K. Cucurbitacin B inhi- bits cell proliferation and induces apoptosis in human osteosarcoma cells via modulation of the JAK2/STAT3 and MAPK pathways[J]. Exp Ther Med,2017,14(1):805- 812.
[43] 張萌,邊志剛,張毅,等.葫蘆素B誘導(dǎo)G2/M期阻滯抑制人前列腺癌DU145細(xì)胞增殖并誘導(dǎo)細(xì)胞凋亡研究[J].中國醫(yī)刊,2016,51(2):63-67.
[44] CHAI Y,XIANG K,WU Y,et al. Cucurbitacin B inhibits the Hippo-YAP signaling pathway and exerts anticancer activity in colorectal cancer cells[J]. Med Sci Monit,2018,24:9251-9258.
[45] 周彤,楊書勝,羅明洲,等.葫蘆素B通過抑制Hippo-YAP通路抑制結(jié)腸癌細(xì)胞轉(zhuǎn)移的機(jī)制研究[J].湖北醫(yī)藥學(xué)院學(xué)報(bào),2020,39(1):16-21,105.
[46] SHUKLA S,SINHA S,KHAN S,et al. Cucurbitacin B inhibits the stemness and metastatic abilities of NSCLC via downregulation of canonical Wnt/β-catenin signaling? ? ?axis[J]. Sci Rep,2016,6:21860.
[47] 喻婷婷,徐麗,董超群,等.葫蘆素B抑制骨肉瘤143B細(xì)胞的增殖和侵襲并促進(jìn)其凋亡[J].腫瘤,2019,39(4):249-258.
[48] GUPTA P,SRIVASTAVA S K. HER2 mediated de novo production of TGFβ leads to SNAIL driven epithelial-to- mesenchymal transition and metastasis of breast cancer[J]. Mol Oncol,2014,8(8):1532-1547.
[49] LUO W W,ZHAO W W,LU J J,et al. Cucurbitacin B suppresses metastasis mediated by reactive oxygen species (ROS) via focal adhesion kinase (FAK) in breast cancer MDA-MB-231 cells[J]. Chin J Nat Med,2018,16(1):10-19.
[50] PROMKAN M,DAKENG S,CHAKRABARTY S,et al. The effectiveness of cucurbitacin B in BRCA1 defective breast cancer cells[J]. PloS One,2013,8(2):e55732.
[51] 梁菁.葫蘆素B通過改變細(xì)胞生物力學(xué)性質(zhì)抑制乳腺癌細(xì)胞遷移和侵襲[D].天津:天津醫(yī)科大學(xué),2019.
[52] ZANOTELLI M R,REINHART-KING C A. Mechanical forces in tumor angiogenesis[J]. Adv Exp Med Bio,2018,1092:91-112.
[53] MACKLIN P S,MCAULIFFE J,PUGH C W,et al. Hypoxia and HIF pathway in cancer and the placenta[J]. Placenta,2017,56:8-13.
[54] PIAO X M,GAO F,ZHU J X,et al. Cucurbitacin B inhi- bits tumor angiogenesis by triggering the mitochondrial signaling pathway in endothelial cells[J]. Int J Mol Med,2018,42(2):1018-1025.
[55] SINHA S,KHAN S,SHUKLA S,et al. Cucurbitacin B inhibits breast cancer metastasis and angiogenesis through VEGF-mediated suppression of FAK/MMP-9 signaling axis[J]. Int J Biochem Cell Biol,2016,77(Pt A):41-56.
[56] TOUIHRI-BARAKATI I,KALLECH-ZIRI O,AYADI W,et al. Cucurbitacin B purified from Ecballium elaterium (L.) A. Rich from Tunisia inhibits α5β1 integrin-mediated adhesion,migration,proliferation of human glioblastoma cell line and angiogenesis[J]. Eur J Pharmacol,2017,797:153-161.
[57] HALL A. The cytoskeleton and cancer[J]. Cancer Metastasis Rev,2009,28(1/2):5-14.
[58] LIANG J,ZHANG X L,YUAN J W,et al. Cucurbitacin B inhibits the migration and invasion of breast cancer cells by altering the biomechanical properties of cells[J]. Phytother Res,2019,33(3):618-630.
[59] WAKIMOTO N,YIN D,OKELLY J,et al. Cucurbitacin B has a potent antiproliferative effect on breast cancer cells in vitro and in vivo[J]. Cancer Sci,2008,99(9):1793-1797.
[60] YIN D,WAKIMOTO N,XING H,et al. Cucurbitacin B markedly inhibits growth and rapidly affects the cytoskeleton in glioblastoma multiforme[J]. Int J Cancer,2008,123(6):1364-1375.
[61] KAUSAR H,MUNAGALA R,BANSAL S S,et al. Cucurbitacin B potently suppresses non-small-cell lung cancer growth:identification of intracellular thiols as critical targets[J]. Cancer Lett,2013,332(1):35-45.
[62] YANG T,LIU J,YANG M,et al. Cucurbitacin B exerts anti-cancer activities in human multiple myeloma cells in vitro and in vivo by modulating multiple cellular pathways[J]. Oncotarget,2017,8(4):5800-5813.
[63] KELLY A D,ISSA J J. The promise of epigenetic therapy:reprogramming the cancer epigenome[J]. Curr Opin Genet Dev,2017,42:68-77.
[64] GONZALO V,LOZANO J J,MU?OZ J,et al. Aberrant gene promoter methylation associated with sporadic multiple colorectal cancer[J]. PloS One,2010,5(1):e8777.
[65] MAO D,LIU A H,WANG Z P,et al. Cucurbitacin B inhibits cell proliferation and induces cell apoptosis in colorectal cancer by modulating methylation status of BTG3[J]. Neoplasma,2019,66(4):593-602.
[66] SHUKLA S,KHAN S,KUMAR S,et al. Cucurbitacin B alters the expression of tumor-related genes by epigenetic modifications in NSCLC and inhibits NNK-induced lung tumorigenesis[J]. Cancer Prev Res (Phila),2015,8(6):552-562.
[67] DITTHAROT K,DAKENG S,SUEBSAKWONG P,et al. Cucurbitacin B induces hypermethylation of oncogenes in breast cancer cells[J]. Planta Medica,2019,85(5):370- 378.
[68] SINGH S S,VATS S,CHIA A Y,et al. Dual role of autophagy in hallmarks of cancer[J]. Oncogene,2018,37(9):1142-1158.
[69] NIU Y,SUN W,LU J J,et al. PTEN activation by DNA damage induces protective autophagy in response to cucurbitacin B in hepatocellular carcinoma cells[J]. Oxid Med Cell Longev,2016,2016:4313204.
[70] ZHU J S,OUYANG D Y,SHI Z J,et al. Cucurbitacin B induces cell cycle arrest,apoptosis and autophagy asso-? ciated with G actin reduction and persistent activation of cofilin in Jurkat cells[J]. Pharmacology,2012,89(5/6):348-356.
[71] LIU X,DUAN C,JI J,et al. Cucurbitacin B induces autophagy and apoptosis by suppressing CIP2A/PP2A/mTORC1 signaling axis in human cisplatin resistant gastric cancer cells[J]. Oncol Rep,2017,38(1):271-278.
[72] 季娟麗,張?jiān)骑w,司淵,等.葫蘆素B抑制人順鉑耐藥胃癌細(xì)胞的作用及機(jī)制研究[J].湖北醫(yī)藥學(xué)院學(xué)報(bào),2017,36(5):386-390,403.
[73] REN G,SHA T,GUO J,et al. Cucurbitacin B induces DNA damage and autophagy mediated by reactive oxygen species (ROS) in MCF-7 breast cancer cells[J]. J Nat Med,2015,69(4):522-530.
[74] 黃擎擎.葫蘆素抑制乳腺癌增殖并誘導(dǎo)凋亡、自噬[D].蘇州:蘇州大學(xué),2016.
[75] KURMAN Y,KILICCIOGLU I,DIKMEN A U,et al. Cucurbitacin B and cisplatin induce the cell death pathways in MB49 mouse bladder cancer model[J]. Exp Biol Med (Maywood),2020,245(9):805-814.
[76] CALCINOTTO A,KOHLI J,ZAGATO E,et al. Cellular senescence:aging,cancer,and injury[J]. Physiol Rev,2019,99(2):1047-1078.
[77] TURNER K J,VASU V,GRIFFIN D K. Telomere biology and human phenotype[J]. Cells,2019,8(1):73.
[78] DUANGMANO S,DAKENG S,JIRATCHARIYAKUL W,et al. Antiproliferative effects of cucurbitacin B in breast cancer cells:down-regulation of the c-Myc/hTERT/telo- merase pathway and obstruction of the cell cycle[J]. Int J Mol Sci,2010,11(12):5323-5338.
[79] GARG S,HUIFU H,KUMARI A,et al. Induction of senescence in cancer cells by a novel combination of cucurbitacin B and withanone:molecular mechanism and therapeutic potential[J]. J Gerontol A Biol Sci Med Sci,2020,75(6):1031-1041.
[80] NIKOLAOU M,PAVLOPOULOU A,GEORGAKILAS A G,et al. The challenge of drug resistance in cancer treatment:a current overview[J]. Clin Exp Metastasis,2018,35(4):309-318.
[81] CAI F,ZHANG L,XIAO X,et al. Cucurbitacin B rever- ses multidrug resistance by targeting CIP2A to reactivate protein phosphatase 2A in MCF-7/adriamycin cells[J].? Oncol Rep,2016,36(2):1180-1186.
[82] 蔡芬,段超,黃秋月,等.葫蘆素B逆轉(zhuǎn)人乳腺癌細(xì)胞MCF-7/Dox多柔比星耐藥的作用及機(jī)制研究[J].湖北醫(yī)藥學(xué)院學(xué)報(bào),2017,36(1):28-35.
[83] LIU P,XIANG Y,LIU X,et al. Cucurbitacin B induces the lysosomal degradation of EGFR and suppresses the CIP2A/PP2A/Akt signaling axis in gefitinib-resistant non-small cell lung cancer[J]. Molecules,2019,24(3):647.
[84] 楊銳,武明薇,向雨晨,等.葫蘆素B誘導(dǎo)EGFR溶酶體降解并通過CIP2A/PP2A/Akt信號(hào)軸抑制吉非替尼耐藥的非小細(xì)胞肺癌的作用機(jī)制[J].湖北醫(yī)藥學(xué)院學(xué)報(bào),2020,39(1):7-15.
[85] 曾超朋.葫蘆素B抑制吉非替尼耐藥非小細(xì)胞肺癌的作用[D].廣州:南方醫(yī)科大學(xué),2018.
[86] QU Y,CONG P,LIN C,et al. Inhibition of paclitaxel resistance and apoptosis induction by cucurbitacin B in ova- rian carcinoma cells[J]. Oncol Lett,2017,14(1):145-152.
(收稿日期:2020-11-07 修回日期:2021-07-07)
(編輯:唐曉蓮)