国产日韩欧美一区二区三区三州_亚洲少妇熟女av_久久久久亚洲av国产精品_波多野结衣网站一区二区_亚洲欧美色片在线91_国产亚洲精品精品国产优播av_日本一区二区三区波多野结衣 _久久国产av不卡

?

間充質(zhì)干細(xì)胞外泌體對(duì)食管癌ECA109細(xì)胞生物學(xué)行為的影響

2025-03-01 00:00:00馬莉莉李子沐王亮許彭李秀梅
天津醫(yī)藥 2025年2期
關(guān)鍵詞:細(xì)胞增殖細(xì)胞凋亡外泌體

摘要:目的 探討間充質(zhì)干細(xì)胞外泌體(MSC-Exos)對(duì)食管癌ECA109細(xì)胞增殖、凋亡及遷移能力的影響。方法 培養(yǎng)人臍帶間充質(zhì)干細(xì)胞,提取并分離外泌體,采用透射電鏡、納米粒徑測(cè)定及表征蛋白(TSG101、CD63)進(jìn)行鑒定。實(shí)驗(yàn)設(shè)MSC-Exo組(MSC-Exos與食管癌ECA109細(xì)胞共培養(yǎng))和空白對(duì)照組(僅培養(yǎng)食管癌ECA109細(xì)胞),分別培養(yǎng)0、24、48 h,采用CCK-8增殖、劃痕實(shí)驗(yàn)分別檢測(cè)各組食管癌EAC109細(xì)胞增殖、遷移能力;培養(yǎng)48 h后,采用流式細(xì)胞儀檢測(cè)細(xì)胞凋亡及細(xì)胞周期變化,Western blot檢測(cè)磷酸化磷酸肌醇3-激酶(p-PI3K)、兔源磷酸化蛋白激酶B(p-Akt)、β-catenin蛋白的表達(dá)。結(jié)果 經(jīng)鑒定,所得MSC-Exos符合要求標(biāo)準(zhǔn)。細(xì)胞培養(yǎng)0 h時(shí),2組細(xì)胞增殖能力和遷移愈合率差異均無(wú)統(tǒng)計(jì)學(xué)意義(P>0.05);培養(yǎng)24 h時(shí),MSC-Exo組細(xì)胞增殖能力低于空白對(duì)照組(P<0.05);培養(yǎng)48 h時(shí),MSC-Exo組細(xì)胞增殖能力和遷移愈合率均低于空白對(duì)照組(P<0.05)。MSC-Exo組細(xì)胞凋亡率高于空白對(duì)照組,G2+S期細(xì)胞比例低于空白對(duì)照組(P<0.05)。MSC-Exo組p-PI3K、p-Akt和β-catenin蛋白表達(dá)水平均低于空白對(duì)照組(P<0.05)。結(jié)論 MSC-Exos能夠抑制食管癌細(xì)胞增殖、遷移,促進(jìn)細(xì)胞凋亡,其對(duì)食管癌細(xì)胞的抑癌作用可能與抑制PI3K、Akt蛋白的活化、下調(diào)β-catenin蛋白表達(dá)有關(guān)。

關(guān)鍵詞:間質(zhì)干細(xì)胞;外泌體;食管腫瘤;細(xì)胞增殖;細(xì)胞運(yùn)動(dòng);細(xì)胞凋亡

中圖分類號(hào):R735.1 文獻(xiàn)標(biāo)志碼:A DOI:10.11958/20241497

Abstract: Objective To explore the effects of exosomes of mesenchymal stem cells (MSC-Exos) on the proliferation, apoptosis, migration and invasion of esophageal cancer ECA109 cells. Methods Human umbilical cord mesenchymal stem cells were cultured, exosomes were extracted and isolated, and identified by transmission electron microscopy. The nanoparticle size determination and protein characterization (TSG101, CD63) were measured by transmission electron microscope. There were the MSC-Exo group (MSC-Exos co-cultured with esophageal cancer ECA109 cells) and the blank control group (only esophageal cancer ECA109 cells), and cells were cultured for 0, 24 and 48 h, respectively. CCK-8 proliferation test and scratch test were used to detect the proliferation and migration ability of esophageal cancer EAC109 cells in each group, respectively. After 48 h of culture, cell apoptosis and cell cycle changes were detected by flow cytometry. The protein expression levels of phosphoinositol 3-kinase phosphorylation (p-PI3K), rabbit phosphorylated protein kinase B phosphorylation (p-Akt) and β-catenin were detected by Western blot assay. Results After identification, the obtained MSC-Exos meeted the required standard. Transmission electron microscopy, particle size measurement and marker protein results confirmed that the extracted exosomes of mesenchymal stem cells meeted the identification criteria. At 0 h of cell culture, there were no significant differences in cell proliferation and migration healing rate between the two groups (P>0.05). After 24 h culture, the cell proliferation ability was lower in the MSC-Exo group than that of the blank control group (P<0.05). After 48 h culture, the cell proliferation and migration healing rate were lower in the MSC-Exo group than those of the blank control group (P<0.05). The apoptosis rate of the MSC-Exo group was higher than that of the blank control group, and the proportion of G2+S phase cells was lower than that of blank control group (P<0.05). The expression levels of" " " " " "p-PI3K, p-Akt and β-catenin protein were significantly lower in the MSC-Exo group than those in the blank control group (P<0.05). Conclusion MSC-Exos can inhibit the proliferation and migration of esophageal cancer cells and promote cell apoptosis. The inhibitory effect of MSC-Exos on esophageal cancer cells may be related to inhibiting the activation of PI3K and Akt protein and the down-regulating expression of β-catenin protein.

Key words: mesenchymal stem cells; exosomes; esophageal neoplasms; cell proliferation; cell movement; apoptosis

食管癌是一種起源于食管黏膜上皮或腺體的惡性腫瘤,會(huì)直接影響患者的進(jìn)食功能。據(jù)統(tǒng)計(jì),食管癌在全球范圍內(nèi)是發(fā)病率和死亡率均居前十位的惡性腫瘤[1]。晚期食管癌發(fā)生遠(yuǎn)處轉(zhuǎn)移會(huì)嚴(yán)重影響患者預(yù)后,并增加病死率。因此,發(fā)現(xiàn)能夠抑制食管癌細(xì)胞增殖和侵襲的新治療靶點(diǎn)至關(guān)重要[2]。間充質(zhì)干細(xì)胞(mesenchymal stem cells,MSCs)是一類具有多向分化潛力及自我更新能力的多功能干細(xì)胞[3]。間充質(zhì)干細(xì)胞外泌體(extracellular vesicles of mesenchymal stem cells,MSC-Exos)包含多種能影響生物體功能和生理變化的物質(zhì),如mRNA、miRNA、lncRNA及表面標(biāo)記分子CD44、CD90等,MSC-Exos在癌癥治療中展現(xiàn)出了潛在的臨床應(yīng)用價(jià)值[4]。本研究旨在探討MSC-Exos對(duì)食管癌細(xì)胞增殖、凋亡和遷移能力的影響,評(píng)估其能否作為抑制腫瘤細(xì)胞轉(zhuǎn)移的有效治療靶點(diǎn)。

1 材料與方法

1.1 實(shí)驗(yàn)材料

1.1.1 細(xì)胞系 MSCs、食管癌ECA109細(xì)胞系由新疆組織細(xì)胞工程重點(diǎn)實(shí)驗(yàn)室提供。

1.1.2 主要儀器與試劑 人臍帶間充質(zhì)干細(xì)胞專用培養(yǎng)基(Clin-SFM-Human MSC)購(gòu)自北京科霖恩生物科技公司;RPMI1640培養(yǎng)基、胎牛血清購(gòu)自美國(guó)Gibco公司;PBS、BCA試劑盒、胰蛋白酶、青/鏈霉素購(gòu)自北京索萊寶生物科技有限公司;外泌體鑒定抗體TSG101、CD63、兔抗人β-catenin、磷酸肌醇3-激酶(PI3K)、兔源蛋白激酶B(Akt)抗體、羊抗兔IgG抗體購(gòu)自江蘇親科生物研究中心有限公司;細(xì)胞周期、凋亡試劑盒購(gòu)自美國(guó)BD公司;CCK-8檢測(cè)試劑盒購(gòu)自武漢博士德公司。透射電鏡HitachiS-4800購(gòu)自日本日立公司;ZetaView納米顆粒跟蹤分析儀購(gòu)自德國(guó)PMX公司;酶標(biāo)儀購(gòu)自美國(guó)Bio-Tek公司;蛋白電泳儀、化學(xué)發(fā)光成像儀、流式細(xì)胞儀均購(gòu)自美國(guó)BIO-RAD公司。

1.2 方法

1.2.1 外泌體提取分離 將人臍帶間充質(zhì)干細(xì)胞用專用培養(yǎng)基培養(yǎng),當(dāng)細(xì)胞生長(zhǎng)至融合率達(dá)80%時(shí)進(jìn)行1∶2傳代培養(yǎng)。收集3~4代的間充質(zhì)干細(xì)胞培養(yǎng)上清液,置于50 mL離心管中,4 ℃、2 000×g離心20 min去除雜質(zhì)細(xì)胞,留取上清液,0.22 μm過(guò)濾篩除去雜質(zhì),轉(zhuǎn)移至超離心轉(zhuǎn)子的多聚異體管中,4 ℃、120 000×g離心70 min,棄上清液留取沉淀,用70 μL PBS重懸沉淀獲取外泌體溶液。外泌體蛋白濃度采用BCA蛋白含量檢測(cè)試劑盒進(jìn)行蛋白定量,得到的外泌體溶液置于-80 ℃保存。

1.2.2 外泌體鑒定 (1)透射電鏡(TEM)觀測(cè)外泌體形態(tài)。吸取稀釋10倍的外泌體樣品10 μL滴在親水碳膜銅網(wǎng)上,靜置5 min,自然干燥使外泌體吸附在銅網(wǎng)上。吸取10 μL磷鎢酸負(fù)染1 min,濾紙吸去多余液體,水洗3次,每次5 min。室溫干燥5 min,采用TEM在120 kV下觀察并拍照。(2)納米粒徑分析法(NTA)檢測(cè)外泌體粒徑。取稀釋5 000倍的外泌體樣品1 mL,ZetaView上機(jī),待其布朗運(yùn)動(dòng)60 s后,測(cè)定粒徑大小、納米粒子濃度和尺寸分布。(3)Western blot檢測(cè)外泌體特異標(biāo)記蛋白分子TSG101、CD63的表達(dá)。

1.2.3 細(xì)胞培養(yǎng)及分組 食管癌ECA109細(xì)胞培養(yǎng)于含10%小牛血清以及100 mg/L青/鏈霉素的RPMI1640培養(yǎng)基中,37 ℃、5%CO2培養(yǎng)箱內(nèi)培養(yǎng),細(xì)胞長(zhǎng)至對(duì)數(shù)生長(zhǎng)期1∶2傳代,取3~4代的細(xì)胞用于實(shí)驗(yàn)。實(shí)驗(yàn)分組:MSC-Exo組,將外泌體用RPMI1640培養(yǎng)基稀釋終濃度為2×1010/mL,與食管癌ECA109細(xì)胞共培養(yǎng);空白對(duì)照組,用RPMI1640完全培養(yǎng)基培養(yǎng)食管癌ECA109細(xì)胞。每組設(shè)置3個(gè)復(fù)孔。

1.2.4 CCK-8增殖實(shí)驗(yàn)檢測(cè)細(xì)胞活性 將食管癌ECA109細(xì)胞以3×103個(gè)/孔接種于96孔板中,各組細(xì)胞設(shè)5個(gè)復(fù)孔,分別于24 h、48 h加入10%CCK-8溶液,37 ℃、5%CO2恒溫培養(yǎng)箱孵育40 min,采用酶標(biāo)儀于450 nm波長(zhǎng)處測(cè)定光密度(OD)值。實(shí)驗(yàn)重復(fù)3次。

1.2.5 劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力 取食管癌ECA109細(xì)胞,以6×105個(gè)/孔接種于6孔板中,37 ℃、5%CO2恒溫培養(yǎng)箱中培養(yǎng)。培養(yǎng)24 h后,用20 μL移液槍頭在孔中央劃出1條直線,將劃痕中細(xì)胞沖洗掉,于顯微鏡下采集細(xì)胞圖像。用Image J軟件定量各組劃痕面積并記為S1,各組加入低血清培養(yǎng)基(2.5%FBS)培養(yǎng),測(cè)量24 h、48 h劃痕面積,測(cè)量各組劃痕面積并記為S2,計(jì)算各組細(xì)胞遷移愈合率。遷移愈合率=(S1-S2)/S1×100%。重復(fù)實(shí)驗(yàn)3次。

1.2.6 流式細(xì)胞儀檢測(cè)細(xì)胞凋亡和細(xì)胞周期 MSC-Exos和食管癌細(xì)胞ECA109共培養(yǎng)48 h后,無(wú)EDTA胰酶消化并收集細(xì)胞,4 ℃預(yù)冷PBS洗滌3次,2 000×g離心5 min,按照BD細(xì)胞凋亡、周期試劑盒方法處理細(xì)胞:(1)加入500 μL緩沖液懸浮細(xì)胞,分別加入5 μL Annexin V-FITC和PI吹打混勻,室溫孵育15 min,1 h內(nèi)流式細(xì)胞儀檢測(cè)細(xì)胞凋亡率。(2)逐滴加入預(yù)冷的70%乙醇5 mL,混勻細(xì)胞4 ℃過(guò)夜,2 000×g離心5 min,去上清液,4 ℃預(yù)冷PBS洗滌3次,加入0.5 mL PI/RNase染液,室溫孵育15 min,1 h內(nèi)流式細(xì)胞儀檢測(cè)細(xì)胞周期。

1.2.7 Western blot檢測(cè)TSG101、CD63、磷酸化PI3K(p-PI3K)、磷酸化Akt(p-Akt)及β-catenin蛋白的表達(dá) 將6×105個(gè)細(xì)胞接種至6孔板,貼壁后分別用MSC-Exo、1640完全培養(yǎng)基處理細(xì)胞48 h,每組5個(gè)復(fù)孔。收集細(xì)胞,加入RIPA裂解緩沖液,冰上裂解30 min,低溫離心。取上清液,BCA蛋白定量試劑盒測(cè)定蛋白濃度。將20 μg蛋白上樣至SDS-PAGE膠加樣孔內(nèi),經(jīng)10%聚丙烯酰胺凝膠電泳(恒壓100 V)分離40~60 min。然后以0.65 mA/cm2恒流電轉(zhuǎn)移1 h將蛋白自凝膠轉(zhuǎn)印至PVDF膜,室溫5%脫脂奶粉封閉PVDF膜2 h后,加入1∶400稀釋的兔抗人TSG101、CD63、PI3K、Akt、" " " " "β-catenin一抗,4 ℃孵育過(guò)夜。TBST漂洗3遍,加入1∶400稀釋的羊抗兔IgG二抗孵育1 h,漂洗后用ECL化學(xué)發(fā)光試劑盒增強(qiáng)發(fā)光。X線膠片顯影,采用Image J系統(tǒng)分析計(jì)算蛋白相對(duì)含量。以β-actin蛋白作為內(nèi)參。實(shí)驗(yàn)重復(fù)3次。

1.3 統(tǒng)計(jì)學(xué)方法 采用SPSS 26.0軟件進(jìn)行數(shù)據(jù)分析。計(jì)量資料以[[x] ±s]表示,2組間比較采用獨(dú)立樣本t檢驗(yàn)。P<0.05為差異有統(tǒng)計(jì)學(xué)意義。

2 結(jié)果

2.1 MSC-Exo鑒定 透射電鏡下可見MSC-Exo呈圓盤狀,具有雙層膜,符合外泌體的形態(tài)特征結(jié)構(gòu)。外泌體平均直徑峰值(132.8±59.1)nm,濃度為5.0×1011 Particles/mL。Western blot實(shí)驗(yàn)結(jié)果顯示MSC-Exo表達(dá)特異性抗體TSG101、CD63,見圖1。

2.2 MSC-Exo對(duì)食管癌細(xì)胞增殖和遷移能力的影響 細(xì)胞培養(yǎng)0 h時(shí),2組細(xì)胞增殖能力和遷移愈合率差異均無(wú)統(tǒng)計(jì)學(xué)意義(P>0.05);培養(yǎng)24 h時(shí),MSC-Exo組細(xì)胞增殖能力低于空白對(duì)照組(P<0.05),遷移愈合率與空白對(duì)照組差異無(wú)統(tǒng)計(jì)學(xué)意義(P>0.05);培養(yǎng)48 h時(shí),MSC-Exo組細(xì)胞增殖能力和遷移愈合率均低于空白對(duì)照組(P<0.05),見圖2、表1。

2.3 MSC-Exo對(duì)食管癌細(xì)胞凋亡和細(xì)胞周期的影響 流式細(xì)胞術(shù)實(shí)驗(yàn)結(jié)果顯示,MSC-Exos與食管癌細(xì)胞ECA109共培養(yǎng)48 h后,MSC-Exo組細(xì)胞凋亡率(Q2+Q4期)高于空白對(duì)照組,G2+S期細(xì)胞比例低于空白對(duì)照組(P<0.05),見表2,圖3、4。

2.4 MSC-Exo對(duì)食管癌細(xì)胞p-PI3K、p-Akt、" " " " " "β-catenin蛋白表達(dá)的影響 MSC-Exo組p-PI3K、p-Akt和β-catenin蛋白表達(dá)水平均低于空白對(duì)照組(P<0.05),見表3、圖5。

3 討論

MSC-Exo中包含有脂質(zhì)、蛋白質(zhì)以及核酸成分,具有親代細(xì)胞的特性和功能,能夠模擬親代細(xì)胞體發(fā)揮免疫調(diào)節(jié)、組織修復(fù)及抗炎作用[5-6]。有研究發(fā)現(xiàn),來(lái)源于人臍帶間充質(zhì)干細(xì)胞的外泌體在體外可抑制胃癌細(xì)胞HGC-27的增殖和侵襲,circRNA測(cè)序結(jié)果提示有效分子可能是外泌體中的circ_0027599[7]。但也有研究顯示,從MSCs分化的脂肪細(xì)胞中分離出的外泌體與乳腺癌細(xì)胞MCF7共同培養(yǎng)后,外泌體能夠被MCF7細(xì)胞吸收攝取,促進(jìn)腫瘤細(xì)胞增殖和遷移[8-9]。目前MSC-Exo對(duì)腫瘤細(xì)胞發(fā)揮抑癌作用或促癌作用具有爭(zhēng)議性,分析其原因可能是MSC-Exo本身具有復(fù)雜多樣性[10],也有可能是由于MSC的培養(yǎng)條件不同等因素引起MSC-Exo分泌的生物活性因子發(fā)生改變,導(dǎo)致其在不同來(lái)源或組織環(huán)境下的功能和作用不同[11]。不同細(xì)胞來(lái)源(甚至同源不同代)的間充質(zhì)干細(xì)胞所分泌的生物活性因子亦存在差異,這可能是引起功能差異的重要原因之一[12]。有研究表明,MSC-Exo與人舌鱗癌細(xì)胞共培養(yǎng)后,細(xì)胞凋亡率明顯升高,提示MSC-Exo能有效誘導(dǎo)鱗癌細(xì)胞凋亡,細(xì)胞攝取的Exo可被癌細(xì)胞內(nèi)化,抑制其增殖并導(dǎo)致細(xì)胞死亡[13]。從人臍帶沃頓氏果凍間充質(zhì)干細(xì)胞中提取的Exo已被證明可抑制T24膀胱癌的生長(zhǎng)[14]。從正常人骨髓間充質(zhì)干細(xì)胞中分離的Exo可抑制細(xì)胞周期進(jìn)程并誘導(dǎo)肝癌(HepG2)和卡波西細(xì)胞凋亡[15]。本研究結(jié)果顯示,MSC-Exos與食管癌ECA109細(xì)胞分別共培養(yǎng)24 h、48 h后,細(xì)胞增殖能力明顯低于空白對(duì)照組;培養(yǎng)48 h,MSC-Exo組遷移愈合率顯著低于空白對(duì)照組,提示MSC-Exo能夠抑制食管癌ECA109細(xì)胞的增殖和遷移能力。MSC-Exos與食管癌細(xì)胞ECA109共培養(yǎng)48 h后,MSC-Exo組細(xì)胞凋亡率較空白對(duì)照組明顯升高,G2+S期細(xì)胞比例較空白對(duì)照組明顯降低,提示MSC-Exo可阻滯食管癌ECA109細(xì)胞的生長(zhǎng)周期,促進(jìn)食管癌細(xì)胞凋亡,對(duì)食管癌細(xì)胞生長(zhǎng)具有抑制作用。

Wnt/β-catenin信號(hào)通路與多種腫瘤的發(fā)生發(fā)展密切相關(guān)[16]。β-catenin是Wnt途徑中最為關(guān)鍵的轉(zhuǎn)導(dǎo)子,抑制異常激活的Wnt/GSK3/β-catenin信號(hào)通路轉(zhuǎn)導(dǎo)能延緩腫瘤進(jìn)展[17]。PI3K、Akt是PI3K/Akt信號(hào)通路上游的關(guān)鍵激酶,其蛋白磷酸化有利于腫瘤細(xì)胞的增殖和遷移[18]。有研究表明,臍帶間充質(zhì)干細(xì)胞外泌體的miR-146a可通過(guò)靶向?qū)应吗みB蛋白亞基γ2(LAMC2)來(lái)調(diào)節(jié)PI3K/Akt信號(hào)通路,從而降低卵巢癌細(xì)胞的生長(zhǎng)和化學(xué)抗性[19]。有研究將人舌鱗癌細(xì)胞用MSC-Exo處理24 h后,發(fā)現(xiàn)PI3K、Akt和mTOR的磷酸化水平均被顯著抑制,而總PI3K、Akt和mTOR的表達(dá)無(wú)顯著變化,提示MSC-Exo對(duì)SCC25細(xì)胞活化PI3K/Akt/mTOR信號(hào)通路具有抑制作用[13]。本研究結(jié)果顯示,MSC-Exos與食管癌細(xì)胞ECA109共培養(yǎng)48 h后,MSC-Exo組p-PI3K、p-Akt和β-catenin蛋白表達(dá)水平均低于空白對(duì)照組,提示MSC-Exo可抑制PI3K、AKT的活化及" " " " " β-catenin蛋白的表達(dá),與上述研究結(jié)果基本一致。提示MSC-Exo對(duì)PI3K/Akt、Wnt/β-catenin等信號(hào)通路均具有抑制作用,其可通過(guò)多條信號(hào)通路抑制腫瘤細(xì)胞的增殖與遷移。

綜上所述,與MSC相比,MSC-Exo具有低免疫源性、高安全性的治療優(yōu)勢(shì),且受到道德倫理的限制相對(duì)較小。但由于MSC-Exo的成分和功能因腫瘤類型而異,應(yīng)用MSC-Exo在不同類型腫瘤患者中的效果需要進(jìn)行具體分析。因此,對(duì)MSC-Exo進(jìn)行嚴(yán)格的質(zhì)控及更深入的研究和臨床實(shí)踐是必要的。

參考文獻(xiàn)

[1] XIA C,DONG X,LI H,et al. Cancer statistics in China and United States[J]. Chin Med J (Engl),2022,135(5):584-590. doi:10.1097/CM9.0000000000002108.

[2] ZHU H,MA X,YE T,et al. Esophageal cancer in China:Practice and research in the new era[J]. Int J Cancer,2023,152(9):1741-1751. doi:10.1002/ijc.34301.

[3] MENSHIKOV M,ZUBKOVA E,STAFEEV I,et al. Autophagy,mesenchymal stem cell differentiation,and secretion[J]. Biomedicines,2021,9(9):1178. doi:10.3390/biomedicines9091178.

[4] ARSHAD M,JALIL F,JALEELl H,et al. Bone marrow derived mesenchymal stem cells therapy for rheumatoid arthritis-a concise review of past ten years[J]. Mol Biol Rep,2023,50(5):4619-4629. doi:10.1007/s11033-023-08277-9.

[5] VAN GRIENSVEN M,BALMAYOR E R. Extracellularvesicles are key players in mesenchymal stem cells dual potential to regenerate and modulate the immune system[J]. Adv Drug Deliv Rev,2024,207:115203. doi:10.1016/j.addr.2024.115203.

[6] ZHU Y,CHEN X,LIAO Y. Mesenchymal stem cells derived apoptotic extracellular vesicles (ApoEVs):mechanism and application in tissue regeneration[J]. Stem Cells,2023,41(9):837-849. doi:10.1093/stmcls/sxad046.

[7] 褚小莉,陳林,朱棟煒. 臍帶間充質(zhì)干細(xì)胞來(lái)源的外泌體抑制胃癌細(xì)胞的增殖和侵襲[J]. 現(xiàn)代腫瘤醫(yī)學(xué),32(12):2140-2145. CHU X L,CHEN L,ZHU D W. Exosomes derived from umbilical cord mesenchymal stem cells inhibit proliferation and invasion of gastric cancer cells[J]. Modern Oncology,2024,32(12):2140-2145. doi:10.3969/j.issn.1672-4992.2024.12.002.

[8] VAN V S,KOURTI A,AUSLOOS E,et al. ATP13A4 upregulation drives the elevated polyamine transport system in the breast cancer cell line MCF7[J]. Biomolecules,2023,13(6):918. doi:10.3390/biom13060918.

[9] WANG S,SU X,XU M,et al. Exosomes secreted by mesenchymal stromal/stem cell-derived adipocytes promote breast cancer cell growth via activation of Hippo signaling pathway[J]. Stem Cell Res Ther,2019,10(1):117. doi:10.1186/ s13287-019-1220-2.

[10] WANG J,ZHANG A,HUANG F,et al. MSC-EXO and tempol ameliorate bronchopulmonary dysplasia in newborn rats by activating HIF-1α[J]. Pediatr Pulmonol,2023,58(5):1367-1379. doi:10.1002/ppul.26317.

[11] HU Z,YUAN Y,ZHANG X,et al. Human umbilical cord mesenchymal stem cell-derived exosomes attenuate oxygen-glucose deprivation/reperfusion-induced microglial pyroptosis by promoting FOXO3a-dependent mitophagy[J]. Oxid Med Cell Longev,2021,2021:6219715. doi:10.1155/2021/6219715.

[12] CAO M,ZHAO Y,CHEN T,et al. Adipose mesenchymal stem cell-derived exosomal microRNAs ameliorate polycystic ovary syndrome by protecting against metabolic disturbances[J]. Biomaterials,2022,288:121739. doi:10.1016/j.biomaterials.2022.121739.

[13] QIU Y,SUN J,QIU J,et al. Antitumor activity of cabazitaxel and MSC-TRAIL derived extracellular vesicles in drug-resistant oral squamous cell carcinoma[J]. Cancer Manag Res,2020,12:10809-10820. doi:10.2147/CMAR.S277324.

[14] SURAMAN M,KEDRACKA-KROK S,JANKOWSKA U,et al. Proteomic profiling of ectosomes derived from paired urothelial bladder cancer and normal cells reveals the presence of biologically-relevant molecules[J]. Int J Mol Sci,2021,22(13):6816. doi:10.3390/ijms22136816.

[15] CHANG W,CERIONE R,ANTONYAK M. Extracellular vesicles and their roles in cancer progression[J]. Methods Mol Biol,2021,2174:143-170. doi:10.1007/978-1-0716-0759-6_10.

[16] LIU J,XIAO Q,XIAO J,et al. Wnt/β-catenin signalling:function,biological mechanisms,and therapeutic opportunities[J]. Signal Transduct Target Ther,2022,7(1):3. doi:10.1038/s41392-021-00762-6.

[17] ZHAO Q,BI Y,ZHONG J,et al. Pristimerin suppresses colorectal cancer through inhibiting inflammatory responses and Wnt/β-catenin signaling[J]. Toxicol Appl Pharmacol,2020,386:114813. doi:10.1016/j.taap.2019.114813.

[18] WANG J,HU K,CAI X,et al. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis[J]. Acta Pharm Sin B,2022,12(1):18-32. doi:10.1016/j.apsb.2021.07.023.

[19] QIU L,WANG J,CHEN M,et al. Exosomal microRNA 146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2 mediated PI3K/Akt axis[J]. Int J Mol Med,2020,46(2):609-620. doi:10.3892/ijmm.2020.4634.

(2024-10-14收稿 2024-12-04修回)

(本文編輯 陳麗潔)

猜你喜歡
細(xì)胞增殖細(xì)胞凋亡外泌體
外泌體miRNA在肝細(xì)胞癌中的研究進(jìn)展
間充質(zhì)干細(xì)胞外泌體在口腔組織再生中的研究進(jìn)展
循環(huán)外泌體在心血管疾病中作用的研究進(jìn)展
外泌體在腫瘤中的研究進(jìn)展
木犀草素對(duì)對(duì)乙酰氨基酚誘導(dǎo)的L02肝細(xì)胞損傷的保護(hù)作用
miRAN—9對(duì)腫瘤調(diào)控機(jī)制的研究進(jìn)展
人類microRNA—1246慢病毒抑制載體的構(gòu)建以及鑒定
傳染性法氏囊病致病機(jī)理研究
科技視界(2016年15期)2016-06-30 12:27:37
G—RH2誘導(dǎo)人肺腺癌A549細(xì)胞凋亡的實(shí)驗(yàn)研究
有關(guān)“細(xì)胞增殖”一輪復(fù)習(xí)的有效教學(xué)策略
彰化市| 巴彦县| 雷州市| 蒲江县| 漳州市| 敖汉旗| 哈巴河县| 乌海市| 巫溪县| 廊坊市| 夏邑县| 神木县| 阜平县| 齐齐哈尔市| 双柏县| 舟曲县| 万荣县| 青州市| 崇义县| 庆安县| 瑞安市| 蒙城县| 漠河县| 历史| 阳西县| 临泉县| 视频| 龙川县| 临猗县| 临澧县| 承德县| 邵东县| 卢氏县| 巴楚县| 永定县| 元谋县| 扎赉特旗| 射洪县| 东源县| 西华县| 乌兰察布市|