• 
    

    
    

      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

      ?

      miR-145-5p靶向TPM3通過(guò)ERK信號(hào)通路抑制甲狀腺癌TPC-1細(xì)胞侵襲和轉(zhuǎn)移

      2021-02-22 09:35:44蘭天劉巍夢(mèng)鄒陽(yáng)邱平
      關(guān)鍵詞:熒光素酶癌細(xì)胞甲狀腺癌

      蘭天,劉巍夢(mèng),鄒陽(yáng),邱平

      ·論著·

      miR-145-5p靶向TPM3通過(guò)ERK信號(hào)通路抑制甲狀腺癌TPC-1細(xì)胞侵襲和轉(zhuǎn)移

      蘭天,劉巍夢(mèng),鄒陽(yáng),邱平

      610500 成都醫(yī)學(xué)院第一附屬醫(yī)院內(nèi)分泌代謝科(蘭天、邱平);617023 四川,攀枝花市第六人民醫(yī)院婦科(劉巍夢(mèng));400700 重慶市第九人民醫(yī)院內(nèi)科(鄒陽(yáng))

      探討 miR-145-5p 對(duì)甲狀腺癌細(xì)胞侵襲和轉(zhuǎn)移的影響。將miR-145-5p mimic 質(zhì)粒、mimic-NC 質(zhì)粒、TPM3 質(zhì)粒分別或聯(lián)合轉(zhuǎn)入甲狀腺癌細(xì)胞,RT-qPCR 檢測(cè) miR-145-5p 與 TPM3 mRNA 的表達(dá),雙熒光素酶報(bào)告檢測(cè)靶向關(guān)系,Transwell 法檢測(cè)細(xì)胞侵襲能力,劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力,Western blot 檢測(cè) ERK1/2 和 p-ERK1/2 的表達(dá);采用裸鼠右后肢腹側(cè)皮下注射構(gòu)建甲狀腺癌移植瘤模型,測(cè)定移植瘤體積和重量,免疫組化檢測(cè) Vimentin 的表達(dá),Western blot 檢測(cè) TPM3、ERK1/2 和 p-ERK1/2 的表達(dá)。miR-145-5p 在甲狀腺癌細(xì)胞中低表達(dá),miR-145-5p 靶向抑制 TPM3 表達(dá);miR-145-5p 過(guò)表達(dá)能夠明顯降低甲狀腺癌細(xì)胞的侵襲數(shù)目和傷口愈合率,并下調(diào) p-ERK 表達(dá),而 TPM3 過(guò)表達(dá)和 ERK 通路激活劑均可以逆轉(zhuǎn)這些現(xiàn)象;miR-145-5p 過(guò)表達(dá)會(huì)使體內(nèi)移植瘤的重量和體積明顯減少,并使移植瘤的 Vimentin 陽(yáng)性細(xì)胞比率明顯減少,TPM3 和 p-ERK1/2 蛋白表達(dá)明顯下調(diào)。miR-145-5p 靶向 TPM3 通過(guò) ERK 信號(hào)通路抑制甲狀腺癌細(xì)胞侵襲和轉(zhuǎn)移。

      miR-145-5p; 甲狀腺癌; 原肌球蛋白 3; ERK; 侵襲; 遷移

      甲狀腺癌是最常見(jiàn)的內(nèi)分泌腫瘤,在女性腫瘤發(fā)病率中高居第五位,發(fā)病率呈逐年上升趨勢(shì),全世界每年近 4 萬(wàn)人死于甲狀腺癌[1]。雖然大多數(shù)患者預(yù)后良好,10 年生存率約為 90%,但有相當(dāng)比例的患者在 10 年內(nèi)出現(xiàn)局部復(fù)發(fā)或遠(yuǎn)處轉(zhuǎn)移[2]。因此,在甲狀腺癌的發(fā)生、發(fā)展過(guò)程中尋找新的分子靶點(diǎn)是迫切需要的。miR-145 被認(rèn)為是一種腫瘤抑制因子,在多種癌癥細(xì)胞中均表現(xiàn)為下調(diào),如結(jié)腸癌、乳腺癌、前列腺癌等,已有研究表明其在甲狀腺癌細(xì)胞中也表現(xiàn)為下調(diào)[3]。原肌球蛋白(TPMs)是一系列存在于骨骼肌、平滑肌和一些非肌肉組織中的肌動(dòng)蛋白結(jié)合蛋白[4]。在骨骼肌中,TPMs 介導(dǎo)肌動(dòng)蛋白對(duì)鈣離子的反應(yīng),并參與細(xì)胞骨架微絲的穩(wěn)定[5]。已有研究表明,非肌肉性 TPMs 可能參與了癌癥的發(fā)展。TPM1 作為一種腫瘤抑制因子,在抑制腫瘤發(fā)展中發(fā)揮作用[6]。而 TPM3是一種重要的致癌基因,已被報(bào)道參與造血腫瘤、黑色素瘤、膠質(zhì)瘤等腫瘤的遷移和侵襲[7-8]。本文主要探討 miR-145-5p 對(duì)甲狀腺癌細(xì)胞侵襲和轉(zhuǎn)移的影響。

      1 材料與方法

      1.1 主要材料

      DMEM 培養(yǎng)基購(gòu)自上海善然生物科技有限公司;胎牛血清購(gòu)自素爾生物科技有限公司;miR-145-5p mimic 質(zhì)粒、mimic-NC 質(zhì)粒、TPM3 質(zhì)粒及各引物均由上?;蛑扑幱邢薰驹O(shè)計(jì)并合成;Lipofectamine2000轉(zhuǎn)染試劑購(gòu)自上海恪敏生物科技有限公司;QIAzol 裂解試劑購(gòu)自北京鴻躍創(chuàng)新科技有限公司;cDNA 逆轉(zhuǎn)錄試劑盒購(gòu)自上海捷瑞生物工程有限公司;雙熒光素酶報(bào)告基因檢測(cè)試劑盒購(gòu)自北京原平皓生物技術(shù)有限公司;SYBR-Green PCR 試劑盒購(gòu)自賽默飛世爾科技公司;RIPA 裂解緩沖液購(gòu)自南京??藸柹锟萍加邢薰?;BCA 試劑盒購(gòu)自上海易色醫(yī)療科技有限公司;TPM3 抗體和 p-ERK1/2 抗體購(gòu)自上海鈺博生物科技有限公司;ERK1/2 抗體和 Vimentin 抗體購(gòu)自碧云天生物科技有限公司;12-O-14 烷酰醇-13-乙酸酯(TPA)購(gòu)自上海偉寰生物科技有限公司;裸鼠購(gòu)自四川夏派森醫(yī)藥科技有限公司,許可證:SYXK(川)2017-203。

      1.2 方法

      1.2.1 細(xì)胞及其培養(yǎng) 正常甲狀腺細(xì)胞系 Nthy-ori3-1和 BC-PAP、K1、TPC-1人甲狀腺癌細(xì)胞系均購(gòu)自 ATCC,于含 10% 胎牛血清的 DMEM 培養(yǎng)基中培養(yǎng),在 37 ℃、5% CO2孵育箱保存。

      1.2.2 分組及處理 將 TPC-1 細(xì)胞分為對(duì)照組、mimic-NC 組、miR-145-5p mimic 組、TPM3 組、TPM3 + miR-145-5p mimic 組,研究 miR-145-5p 對(duì)甲狀腺癌細(xì)胞的影響。mimic-NC 組、miR-145-5p mimic 組、TPM3 組、TPM3 + miR-145-5p mimic 組細(xì)胞在對(duì)數(shù)期時(shí),接種于 6 孔板(1 × 106個(gè)/孔)。當(dāng)達(dá)到 80% 融合,根據(jù) Lipofectamine2000說(shuō)明書(shū)將 100 nmol/L miR-145-5p mimic 質(zhì)粒、mimic-NC 質(zhì)粒、TPM3 質(zhì)粒分別或聯(lián)合轉(zhuǎn)染進(jìn)入 TPC-1 細(xì)胞。將 TPC-1 細(xì)胞分為對(duì)照組、miR-145-5p mimic 組、TPA 組、miR-145-5p mimic + TPA 組,研究 miR-145-5p 對(duì)甲狀腺癌細(xì)胞的作用通路。miR-145-5p mimic 組:根據(jù) Lipofectamine2000說(shuō)明書(shū)將 miR-145-5p mimic 質(zhì)粒轉(zhuǎn)染至TPC-1 細(xì)胞處理 6 h;TPA 組:50 μmol/L TPA 處理細(xì)胞 6 h;miR-145-5p mimic + TPA 組:根據(jù) Lipofectamine2000說(shuō)明書(shū)將 miR-145-5p mimic 質(zhì)粒用 50 μmol/L TPA 處理細(xì)胞 6 h。

      1.2.3 RT-qPCR 檢測(cè) miR-145-5p 與 TPM3 mRNA 的表達(dá) 采用 QIAzol 裂解試劑提取總RNA,采用 cDNA 逆轉(zhuǎn)錄試劑盒合成 cDNA。RT-qPCR 采用 SYBR-Green PCR 試劑盒說(shuō)明書(shū)操作進(jìn)行。GAPDH 作為 mRNA 的內(nèi)參,使用 2-ΔΔct法計(jì)算。miR-145-5p 的上游引物序列:5' GTCCAG TTTTCCCAGGAAT 3';miR-145-5p 的下游引物序列:5' TGGTGTCGTGGAGTCG 3'。TPM3 的上游引物序列:5' GCGGGAAGTGGAGGGAGAAA 3';TPM3 的下游引物序列:5' TAGACTCTGCCAGCT CGGCG 3'。GAPDH 的上游引物序列:5' ACAACTTT GGTATCGTGGAAGG 3';GAPDH 的下游引物序列:5' GCCATCACGCCACAGTTTC 3'。

      1.2.4 雙熒光素酶報(bào)告檢測(cè)靶向關(guān)系 收集生長(zhǎng)至對(duì)數(shù)期的 TPC-1 細(xì)胞,鋪于 96 孔板,每孔約4 × 103個(gè)細(xì)胞,24 h 后,分別轉(zhuǎn)染mimic-NC + TPM3 WT、mimic-NC + TPM3 MUT、miR-145-5pmimic + TPM3 WT、miR-145-5p mimic + TPM3 MUT,根據(jù)雙熒光素酶報(bào)告基因試劑盒說(shuō)明進(jìn)行測(cè)定,用螢火蟲(chóng)熒光素酶活性和腎熒光素酶活性比值表示熒光素酶的相對(duì)活性。

      1.2.5 Transwell 法檢測(cè)細(xì)胞侵襲能力 培養(yǎng) 48 h后,將細(xì)胞消化成單細(xì)胞懸液(5 × 104個(gè)/ml)。在 Transwell 小室上室用 Matrigel 包被,下室加入含有 10% 胎牛血清的 DMEM 細(xì)胞培養(yǎng)液 1 ml。孵育 24 h 后,除去膜上層細(xì)胞,將通過(guò)膜侵襲的細(xì)胞固定染色,在顯微鏡下觀察。侵入細(xì)胞數(shù)表示為每視野的平均細(xì)胞數(shù)。

      1.2.6 劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力 將各組 TPC-1 細(xì)胞消化鋪滿單層后用小號(hào)槍頭垂直劃痕,加入無(wú)血清培養(yǎng)基,在5% CO2、37 ℃恒溫的培養(yǎng)箱培養(yǎng)24 h 后,分別在顯微鏡下拍照,用 Image J 軟件分析。

      1.2.7 Western blot 檢測(cè) TPM3、ERK1/2 和 p-ERK1/2 的表達(dá) 用 RIPA 裂解液提取各組 TPC-1 細(xì)胞總蛋白,并用 BCA 試劑盒檢測(cè)總蛋白濃度,然后經(jīng) SDS-PAGE 分離蛋白后,用半干轉(zhuǎn)膜儀轉(zhuǎn)移蛋白質(zhì)至 PVDF 膜,并用脫脂牛奶室溫封閉蛋白 2 h,再加入一抗(TPM3:1:400;ERK1/2:1:1000;p-ERK1/2:1:200)在 4 ℃封閉過(guò)夜,接著加入對(duì)應(yīng)二抗室溫封閉 1 h,最后滴 ECL 曝光。

      1.2.8 移植瘤實(shí)驗(yàn) 將裸鼠隨機(jī)分為 TPC-1 組和 miR-145-5p mimic 組,每組 10 只,雌雄各半。在各組裸鼠右后肢腹側(cè)皮下分別注射 0.2 ml TPC-1 細(xì)胞和轉(zhuǎn)染 miR-145-5p mimic 的 TPC-1 細(xì)胞懸液。然后,繼續(xù)在無(wú)特定病原(SPF)條件下正常飲食飼養(yǎng),觀察裸鼠皮下成瘤情況,測(cè)定移植瘤體積。第 30 天頸椎脫位法處死裸鼠,完整取出皮下移植瘤,電子天平稱(chēng)重,Western blot 檢測(cè) TPM3、ERK1/2 和 p-ERK1/2 的表達(dá)。

      1.2.9 免疫組化檢測(cè) Vimentin的表達(dá) 經(jīng)常規(guī) 10% 中性甲醛溶液固定,石蠟包埋切片,脫蠟水化過(guò)氧化物酶阻斷內(nèi)源性過(guò)氧化物酶活性,非免疫性動(dòng)物血清阻斷非特異性反應(yīng)。再分別加入鼠抗人 Vimentin抗體(1:100),4 ℃過(guò)夜。滴加生物素標(biāo)記二抗,DAB 顯色,蒸餾水沖洗,蘇木素復(fù)染,梯度酒精脫水,二甲苯透明。最后,封片觀察統(tǒng)計(jì)。免疫組化陽(yáng)性細(xì)胞其細(xì)胞核有棕色顆粒沉著,顯微鏡下隨機(jī)選取 5 個(gè)視野,每個(gè)視野陽(yáng)性率 = 陽(yáng)性細(xì)胞數(shù)/總細(xì)胞數(shù) × 100%。

      1.3 統(tǒng)計(jì)學(xué)處理

      2 結(jié)果

      2.1 miR-145-5p 在甲狀腺癌細(xì)胞中下調(diào)

      RT-qPCR 檢測(cè)正常甲狀腺細(xì)胞系 Nthy-ori3-1 和 BC-PAP、K1、TPC-1 等 3 個(gè)甲狀腺癌細(xì)胞中 miR-145-5p 的表達(dá)發(fā)現(xiàn),與正常甲狀腺細(xì)胞系 Nthy-ori3-1 相比,BC-PAP、K1、TPC-1 等 3 個(gè)甲狀腺癌細(xì)胞中miR-145-5p mRNA 的表達(dá)明顯下調(diào)(< 0.01),且以TPC-1 下調(diào)最顯著(圖 1),故選擇甲狀腺癌細(xì)胞系 TPC-1 作為后續(xù)實(shí)驗(yàn)研究對(duì)象。

      2.2 miR-145-5p 靶向抑制 TPM3 表達(dá)

      根據(jù) TargetScan 數(shù)據(jù)庫(kù)的預(yù)測(cè),TPM3 3'UTR 區(qū)與 miR-145-5p 存在結(jié)合位點(diǎn)(圖2A)。為了進(jìn)一步驗(yàn)證 miR-145-5p 直接作用于 TPM3,進(jìn)行了熒光素酶報(bào)告基因?qū)嶒?yàn),結(jié)果如圖 2B 所示。miR-145-5p 高表達(dá)明顯抑制了含有野生型 TPM3 質(zhì)粒的熒光素酶活性,但對(duì)突變型 TPM3 質(zhì)粒的熒光素酶活性無(wú)影響(< 0.01)。為了進(jìn)一步證實(shí)轉(zhuǎn)染效果,RT-qPCR 檢測(cè)各組細(xì)胞 TPM3 mRNA 的表達(dá),結(jié)果如圖 2C 所示,與 control 組細(xì)胞相比,mimic-NC 組細(xì)胞中 TPM3 mRNA 的表達(dá)無(wú)明顯變化,miR-145-5p mimic 組細(xì)胞中TPM3 mRNA 表達(dá)明顯下調(diào)(< 0.01),TPM3 組細(xì)胞中 TPM3 mRNA 表達(dá)明顯上調(diào)(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組細(xì)胞中 TPM3 mRNA 表達(dá)明顯上調(diào)(< 0.01)。因此,miR-145-5p 直接靶向抑制 TPM3。

      miR-145-5p mRNA 表達(dá)水平Expression of miR-145-5p mRNA1.2 1.0 0.8 0.6 0.4 0.2 0 Nthy-ori3-1 BC-PAP K1 TPC-1

      Figure 1 Expression of miR-145-5p in each group of cells was detected by RT-qPCR (**< 0.01 vs Nthy-ori3-1)

      A

      Figure 2 miR-145-5p targeted inhibition of TPM3 (A: The binding site of the TPM3 3'UTR region to miR-145-5p is predicted by the TargetScan database; B: miR-145-5p and TPM3 targeting relationship was detected by a dual luciferase reporter gene assay,**< 0.01 compared with the mimic-NC group; C: Expression of TPM3 mRNA in each group of cells was detected by RT-qPCR,**< 0.01 compared with the control group,##< 0.01 compared with the miR-145-5p mimic group)

      2.3 miR-145-5p 靶向 TPM3 抑制甲狀腺癌細(xì)胞侵襲和遷移

      Transwell 法檢測(cè)各組細(xì)胞侵襲能力,結(jié)果如圖 3A 所示。與 control 組細(xì)胞相比,mimic-NC 組每視野侵襲數(shù)目無(wú)明顯變化,miR-145-5p mimic 組每視野侵襲數(shù)目明顯降低(< 0.01),TPM3 組每視野侵襲數(shù)目增高(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組每視野侵襲數(shù)目明顯增高(< 0.01)。因此,miR-145-5p 靶向TPM3 抑制甲狀腺癌細(xì)胞侵襲。

      劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力,結(jié)果如圖 3B 所示,與 control 組細(xì)胞相比,mimic-NC 組細(xì)胞傷口愈合率無(wú)明顯變化,miR-145-5p mimic 組細(xì)胞傷口愈合率明顯降低(< 0.01),TPM3 組細(xì)胞傷口愈合率明顯增高(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組細(xì)胞傷口愈合率明顯增高(< 0.01)。因此,miR-145-5p 靶向 TPM3 抑制甲狀腺癌細(xì)胞遷移。

      2.4 miR-145-5p 靶向 TPM3 抑制 ERK 信號(hào)通路

      Western blot 檢測(cè)各組細(xì)胞 ERK 和磷酸化 ERK 蛋白表達(dá)量,結(jié)果如圖 4A 所示。與 control 組相比,mimic-NC 組細(xì)胞中 ERK 和磷酸化 ERK 蛋白表達(dá)均無(wú)明顯變化,miR-145-5p mimic 組細(xì)胞中 ERK 蛋白表達(dá)無(wú)明顯變化、磷酸化 ERK 蛋白表達(dá)明顯下調(diào)(< 0.01),TPM3 組細(xì)胞中 ERK 蛋白表達(dá)無(wú)明顯變化、磷酸化 ERK 蛋白表達(dá)明顯上調(diào)(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組細(xì)胞中 ERK 蛋白表達(dá)無(wú)明顯變化、磷酸化 ERK 蛋白表達(dá)明顯上調(diào)(< 0.01)。因此,miR-145-5p 靶向 TPM3 對(duì)甲狀腺癌的作用可能與抑制 ERK 信號(hào)通路有關(guān)。

      添加 ERK 信號(hào)通路激活劑TPA驗(yàn)證該預(yù)測(cè)。通過(guò) Transwell 法檢測(cè)各組細(xì)胞侵襲能力,結(jié)果見(jiàn)圖4B。與 control 組相比,miR-145-5p mimic組每視野 TPC-1 細(xì)胞侵襲數(shù)目明顯減少(< 0.01),TPA 組每視野 TPC-1 細(xì)胞侵襲數(shù)目明顯增多(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組每視野 TPC-1 細(xì)胞侵襲數(shù)目明顯增多(< 0.01),說(shuō)明 miR-145-5p 通過(guò) ERK 信號(hào)通路抑制甲狀腺癌細(xì)胞侵襲。通過(guò)劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力,結(jié)果如圖 4C 所示。與 control 組相比,miR-145-5p mimic 組傷口愈合率明顯降低(< 0.01),TPA 組傷口愈合率明顯升高(< 0.01);與 miR-145-5p mimic 組相比,TPM3 + miR-145-5p mimic 組傷口愈合率明顯升高(< 0.01)。因此,確證miR-145-5p 通過(guò) ERK 信號(hào)通路抑制甲狀腺癌細(xì)胞遷移。

      2.5 miR-145-5p 抑制甲狀腺癌細(xì)胞移植瘤發(fā)生和發(fā)展

      為了進(jìn)一步研究 miR-145-5p 對(duì)甲狀腺癌發(fā)生和發(fā)展影響,進(jìn)行了甲狀腺癌細(xì)胞移植瘤實(shí)驗(yàn)。如圖 5A、B、C 所示,與 control 組相比,miR-145-5p mimic 組大鼠體內(nèi)移植瘤的重量和體積明顯減少(< 0.01)。如圖 5D 所示,與 control 組相比,miR-145-5p mimic 組大鼠體內(nèi)移植瘤 Vimentin 的陽(yáng)性細(xì)胞比率明顯減少(< 0.01)。如圖 5E 所示,與 control 組相比,miR-145-5p mimic 組大鼠體內(nèi)移植瘤 TPM3、p-ERK 蛋白表達(dá)明顯下調(diào)(< 0.01)。因此,miR-145-5p 抑制甲狀腺癌細(xì)胞移植瘤發(fā)生和發(fā)展。

      3 討論

      癌癥的發(fā)生和發(fā)展是一個(gè)極其復(fù)雜的生物學(xué)過(guò)程,涉及數(shù)以百萬(wàn)計(jì)的分子和生物學(xué)途徑,其中 miRNAs 近年來(lái)受到廣泛關(guān)注。已有眾多證據(jù)表明 miRNAs與前列腺癌、宮頸癌、肝癌和肺癌等多種癌癥的發(fā)生和發(fā)展密切相關(guān)[9-12]。過(guò)表達(dá) miR-145 能夠抑制結(jié)腸癌、食管鱗狀細(xì)胞癌、肺癌等癌癥的發(fā)生[13-15]。本文通過(guò)研究發(fā)現(xiàn),與正常甲狀腺細(xì)胞相比,miR-145 在甲狀腺癌細(xì)胞中表達(dá)明顯下調(diào)。因此,過(guò)表達(dá) miR-145 有望抑制甲狀腺癌的發(fā)生和發(fā)展。本研究通過(guò) TargetScan 數(shù)據(jù)庫(kù)預(yù)測(cè)到 TPM3 3'UTR 區(qū)與 miR-145-5p 存在結(jié)合位點(diǎn),且通過(guò)熒光素酶報(bào)告基因?qū)嶒?yàn)發(fā)現(xiàn) miR-145-5p 直接靶向 TPM3。已有研究表明,在高轉(zhuǎn)移性小鼠黑色素瘤細(xì)胞系中,TPM3 的表達(dá)水平高于低轉(zhuǎn)移性小鼠黑色素瘤細(xì)胞[8];TPM3 敲除能顯著抑制膠質(zhì)瘤細(xì)胞的侵襲和遷移潛能[16]。本文研究發(fā)現(xiàn),過(guò)表達(dá) miR-145-5p 能夠靶向抑制 TPM3 的表達(dá),從而抑制甲狀腺癌的侵襲和遷移。

      甲狀腺癌患者治療存在的最大問(wèn)題就是局部復(fù)發(fā)或遠(yuǎn)處轉(zhuǎn)移,因此,抑制甲狀腺癌細(xì)胞的侵襲和遷移能力對(duì)于甲狀腺癌的治療至關(guān)重要。Liu 等[17]發(fā)現(xiàn),miR-145-5p 通過(guò)靶向信號(hào)素 3A 抑制脂肪源性干細(xì)胞的成骨分化。Ding 等[18]報(bào)道 miR-145 通過(guò)調(diào)節(jié) TGF-β1 表達(dá)抑制乳腺癌細(xì)胞的增殖和遷移。Sathyanarayanan 等[19]發(fā)現(xiàn) miR-145 通過(guò)靶向人宮頸癌細(xì)胞 SIP1,調(diào)節(jié)上皮-間質(zhì)轉(zhuǎn)化,抑制增殖、遷移和侵襲,因此,miR-145 有望抑制甲狀腺癌細(xì)胞的侵襲和遷移能力。本文通過(guò) Transwell 法和劃痕實(shí)驗(yàn)發(fā)現(xiàn),miR-145-5p 靶向 TPM3 抑制甲狀腺癌細(xì)胞侵襲和遷移,這與其他癌癥細(xì)胞的結(jié)果相一致。

      Control mimic-NC miR-145-5p mimic TPM3 TPM3 + miR-145-5p mimic

      侵襲細(xì)胞數(shù)目Number of invading cells500 400 300 200 100 0 Control mimic-NC miR-145-5p mimic TPM3 TPM3 +miR-145-5p mimicA

      Control mimic-NC miR-145-5p mimic TPM3 TPM3 + miR-145-5p mimic 0 h 24 h

      劃痕閉合率(%)Scratch closure rate (%)100 80 60 40 20 0 Control mimic-NC miR-145-5p mimic TPM3 TPM3 +miR-145-5p mimicB

      Figure 3 Effect of miR-145-5p targeting TPM3 on invasion and migration of thyroid cancer cell (A: Invasive ability of each group was detected by Transwell method,**< 0.01 compared with the control group,##< 0.01 compared with the miR-145-5p mimic group; B: Cell migration ability was detected by scratch test,**< 0.01 compared with the control group,##< 0.01 compared with the miR-145-5p mimic group)

      p-ERK1/2 ERK1/2 GAPDH蛋白表達(dá)水平Expressed of protein0.6 0.4 0.2 0 Controlmimic-NCmiR-145-5p mimicTPM3TPM3 + miR-145-5p mimic Control mimic-NC miR-145-5p mimic TPM3 TPM3 +miR-145-5p mimic ERK1/2 p-ERK1/2 A

      Figure 4 miR-145-5p inhibits ERK signaling pathway by targeting TPM3 (A: Expression levels of ERK and p-ERK protein in each group were detected by Western blot,**compared with control group,< 0.01,##compared with miR-145-5p mimic group,< 0.01; B: Invasive ability of each group was detected by Transwell method,**< 0.01 compared with the control group,##< 0.01 compared with the miR-145-5p mimic group; C: Cell migration ability was detected by scratch test,**< 0.01 compared with the control group,##< 0.01 compared with the miR-145-5p mimic group)

      移植瘤體積(mm3)Transplanted tumor volume (mm3)800 600 400 200 0 Control miR-145-5p mimic A 3 6 9 12 15 18 21 24 27 30 時(shí)間(h)Time (h)B

      Figure 5 Effect of miR-145-5p on thyroid carcinoma xenografts (A: Picture of transplanted tumor; B: Volume of transplanted tumor,**< 0.01 compared with the control group; C: Weight of the transplanted tumor,**< 0.01 compared with the control group; D: Expression of Vimentin+in transplanted tumors was detected by immunohistochemistry,**< 0.01 compared with the control group; E: Expression of TPM3, ERK and p-ERK protein in the transplanted tumor was detected by Western blot,**< 0.01 compared with the control group)

      本研究發(fā)現(xiàn),miR-145-5p 靶向 TPM3 抑制磷酸化 ERK 的表達(dá),說(shuō)明 miR-145-5p 靶向TPM3 對(duì)甲狀腺癌的侵襲和遷移能力的抑制作用可能與抑制 ERK 信號(hào)通路有關(guān)。為了驗(yàn)證這一猜測(cè),添加 ERK 信號(hào)通路激活劑 TPA 后通過(guò) Transwell 法和劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞侵襲和遷移能力。結(jié)果發(fā)現(xiàn),miR-145-5p 通過(guò) ERK 信號(hào)通路抑制甲狀腺癌細(xì)胞侵襲和遷移。ERK 信號(hào)通路是自然界生物體內(nèi)普遍存在的信號(hào)轉(zhuǎn)導(dǎo)系統(tǒng)之一,參與細(xì)胞的增殖、分化及凋亡等不同生理功能。已有研究表明激活ERK 信號(hào)通路途徑有助于甲狀腺癌細(xì)胞的侵襲和遷移[20],抑制 ERK 信號(hào)通路途徑能夠抑制甲狀腺癌細(xì)胞的侵襲和遷移[21],這與本文研究結(jié)果相統(tǒng)一。

      通過(guò)體外實(shí)驗(yàn)可知,miR-145-5p 過(guò)表達(dá)靶向 TPM3 通過(guò) ERK 信號(hào)通路抑制甲狀腺癌細(xì)胞的侵襲和遷移。生物機(jī)體內(nèi)環(huán)境是極其復(fù)雜多變的,為了更好地了解 miR-145-5p 對(duì)甲狀腺癌細(xì)胞的影響,本文進(jìn)行了裸鼠體內(nèi)移植瘤實(shí)驗(yàn)。通過(guò)研究發(fā)現(xiàn),miR-145-5p 過(guò)表達(dá)會(huì)使體內(nèi)移植瘤的重量和體積明顯減少,并使移植瘤的 Vimentin 陽(yáng)性細(xì)胞比率明顯減少,TPM3、ERK、p-ERK蛋白表達(dá)明顯下調(diào),說(shuō)明 miR-145-5p 過(guò)表達(dá)抑制甲狀腺癌移植瘤的侵襲和遷移,并抑制 ERK 信號(hào)通路的激活。

      綜上所述,miR-145 在甲狀腺癌細(xì)胞中低表達(dá),高表達(dá) miR-145 靶向抑制 TPM3 來(lái)抑制甲狀腺癌細(xì)胞的侵襲和遷移,這種作用是通過(guò) ERK 信號(hào)通路來(lái)實(shí)現(xiàn)的。未來(lái)將進(jìn)一步挖掘 miR-145 在甲狀腺癌的診斷和預(yù)后中的價(jià)值。

      [1] Lin Y, Cheng K, Wang T, et al. miR-217 inhibits proliferation, migration, and invasion via targeting akt3 in thyroid cancer. Biomed Pharmacother, 2017, 95:1718-1724.

      [2] Pemayun TG. Current diagnosis and management of thyroid nodules. Acta Med Indones, 2016, 48(3):247-257.

      [3] Foroutan T, Farhadi A, Abroun S, et al. Adipose derived stem cells affect miR-145and p53 expressions of Co-cultured hematopoietic stem cells. Cell J, 2018, 19(4):654-659.

      [4] Jang WI, Jo YJ, Kim HC, et al. Non-muscle tropomyosin (Tpm3) is crucial for asymmetric cell division and maintenance of cortical integrity in mouse oocytes. Cell Cycle, 2016, 13(15):2359-2369.

      [5] Janco M, Rynkiewicz MJ, Li L, et al. Molecular integration of the anti-tropomyosin compound ATM-3507 into the coiled coil overlap region of the cancer-associated Tpm3.1. Sci Rep, 2019, 9(1):11262.

      [6] Zhu S, Si ML, Wu H, et al. Microrna-21 targets the tumor suppressor gene tropomyosin 1 (tpm1). J Biol Chem, 2017, 282(19):14328- 14336.

      [7] Kim TM, Yim SH, Shin SH, et al. Clinical implication of recurrent copy number alterations in hepatocellular carcinoma and putative oncogenes in recurrent gains on 1q. Int J Cancer, 2018, 123(12): 2808-2815.

      [8] Zhou S, Ma X, Wang ZJ, et al. Research on the establishment of a TPM3 monoclonal stable transfected PANC-1 cell line and the experiment of the EMT occurrence in human pancreatic cancer. Onco Targets Ther, 2019, 12(2):5577-5587.

      [9] Feng F, Liu H, Chen A, et al. miR-148-3p and miR-152-3p synergistically regulate prostate cancer progression via repressing KLF4. J Cell Biochem, 2019, 120(10):17228-17239.

      [10] Chen L, Shi X, Huo D, et al. miR-449b-5p regulates cell proliferation, migration and radioresistance in cervical cancer by interacting with the transcription suppressor FOXP1. Eur J Pharmacol, 2019, 856: 172399.

      [11] Wang S, Zhang S, He Y, et al. HOXA11-AS regulates JAK-STAT pathway by miR-15a-3p/STAT3 axis to promote the growth and metastasis in liver cancer. J Cell Biochem, 2019, 120(9):15941-15951.

      [12] Yang Y, Li H, Liu Y, et al. MIR-4319 hinders yap expression to restrain non-small cell lung cancer growth through regulation of LIN28-mediated RFX5 stability. Biomed Pharmacother, 2019, 115: 108956.

      [13] Sheng N, Tan G, You W, et al. Mir-145 inhibits human colorectal cancer cell migration and invasion via PAK4-dependent pathway. Cancer Med, 2017, 6(6):1331-1340.

      [14] Mei LL, Wang WJ, Qiu YT, et al. miR-145-5p suppresses tumor cell migration, invasion and epithelial to mesenchymal transition by regulating the Sp1/NF-κB signaling pathway in esophageal squamous cell carcinoma. Int J Mol Sci, 2017, 18(9):1833.

      [15] Pan Y, Ye C, Tian Q, et al. miR-145 suppresses the proliferation, invasion and migration of NSCLC cells by regulating the BAX/BCL-2 ratio and the caspase-3 cascade. Oncol Lett, 2018, 15(4):4337-4343.

      [16] Tao T, Shi Y, Han D, et al. TPM3, a strong prognosis predictor, is involved in malignant progression through MMP family members and EMT-like activators in gliomas. Tumor Biol, 2014, 35(9):9053-9059.

      [17] Liu X, Zhu W, Wang L, et al. miR-145-5p suppresses osteogenic differentiation of adipose-derived stem cells by targeting semaphorin 3A. In Vitro Cell Dev Biol Anim, 2019, 55(3):189-202.

      [18] Ding Y, Zhang C, Zhang J, et al. miR-145 inhibits proliferation and migration of breast cancer cells by directly or indirectly regulating TGF-β1 expression. Int J Oncol, 2017, 50(5):1701-1710.

      [19] Sathyanarayanan A, Chandrasekaran KS, Karunagaran D. microRNA-145 modulates epithelial-mesenchymal transition and suppresses proliferation, migration and invasion by targeting SIP1 in human cervical cancer cells. Cell Oncol (Dordr), 2017, 40(2):119-131.

      [20] Guan H, Guo Z, Liang W, et al. Trop2 enhances invasion of thyroid cancer by inducing MMP2 through ERK and JNK pathways. BMC Cancer, 2017, 17(1):486.

      [21] Hong S, Yu S, Li J, et al. miR-20b displays tumor-suppressor functions in papillary thyroid carcinoma by regulating the MAPK/ERK signaling pathway. Thyroid, 2016, 26(12):1733-1743.

      miR-145-5p inhibits thyroid cancer TPC-1 cell invasion and metastasis via ERK signaling pathway by targeting TPM3

      LAN Tian, LIU Wei-meng, ZOU Yang, QIU Ping

      Department of Endocrinology and Metabolism, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China (LAN Tian, QIU Ping); Department of Gynecology, The Sixth People's Hospital of Panzhihua, Panzhihua 617023, China (LIU Wei-meng); Internal Medicine, The Ninth People's Hospital of Chongqing, Chongqing 400700, China(ZOU Yang)

      This work aims to study the effect of miR-145-5p on invasion and metastasis of thyroid cancer cells.miR-145-5p mimic plasmid, mimic-NC plasmid, and TPM3 plasmid were separately or jointly transfected into thyroid cancer cells by Lipofectamine2000. Expression of miR-145-5p and TPM3 mRNA was detected by RT-qPCR. The targeted relationship was detected by dual luciferase reporting assay. Cell invasion ability was detected by Transwell method. Cell migration ability was measured by a scratch test. Expression of ERK1/2, and p-ERK1/2 was detected by Western blot. A thyroid cancer xenograft model was constructed by subcutaneous injection of the right hind limb of nude mice. The volume and weight of the transplanted tumor were determined. Expression of vimentin was detected by immunohistochemistry. Expression of TPM3, ERK1/2 and p-ERK1/2 was detected by Western blot.Expression of miR-145-5p is decreased in thyroid cancer cells, and miR-145-5p was found to target and inhibit TPM3 expression. Overexpression of miR-145-5p significantly reduced the number of invasion and wound healing of thyroid cancer cells, and down-regulated p-ERK expression, whereas both TPM3 overexpression and ERK pathway activators reversed the miR-145-5p-induced effects (< 0.01). Overexpression of miR-145-5p significantly reduced the weight and volume of transplanted tumors, and significantly reduced the ratio of Vimentin-positive cells in transplanted tumors and down-regulated TPM3 and p-ERK1/2 protein expression (< 0.01).miR-145-5p inhibits thyroid cancer cell invasion and metastasis via ERK signaling pathway by targeting TPM3.

      miR-145-5p; thyroid cancer; TPM3; ERK; invasion; migration

      QIU Ping, Email: wxqiuping@163.com

      四川省教育廳自然類(lèi)課題(18Z065)

      邱平,Email:wxqiuping@163.com

      2020-09-10

      10.3969/j.issn.1673-713X.2021.01.005

      猜你喜歡
      熒光素酶癌細(xì)胞甲狀腺癌
      NNMT基因啟動(dòng)子雙熒光素酶報(bào)告系統(tǒng)的構(gòu)建及其與SND1靶向關(guān)系的驗(yàn)證
      不同雙熒光素酶方法對(duì)檢測(cè)胃癌相關(guān)miRNAs靶向基因TIAM1的影響
      分化型甲狀腺癌切除術(shù)后多發(fā)骨轉(zhuǎn)移一例
      分化型甲狀腺癌肺轉(zhuǎn)移的研究進(jìn)展
      重組雙熒光素酶報(bào)告基因質(zhì)粒psiCHECK-2-Intron構(gòu)建轉(zhuǎn)染及轉(zhuǎn)染細(xì)胞螢火蟲(chóng)熒光素酶和海腎熒光素酶表達(dá)
      癌細(xì)胞最怕LOVE
      假如吃下癌細(xì)胞
      癌細(xì)胞最怕Love
      奧秘(2017年5期)2017-07-05 11:09:30
      正常細(xì)胞為何會(huì)“叛變”? 一管血可測(cè)出早期癌細(xì)胞
      全甲狀腺切除術(shù)治療甲狀腺癌適應(yīng)證選擇及并發(fā)癥防治
      崇阳县| 广汉市| 鱼台县| 砚山县| 邓州市| 云安县| 博湖县| 蒲江县| 深州市| 西华县| 惠州市| 盐池县| 沁水县| 云林县| 和平县| 漳州市| 通渭县| 阳泉市| 柳河县| 沭阳县| 重庆市| 屏山县| 泾阳县| 廊坊市| 桐梓县| 黄山市| 东乡县| 广灵县| 太康县| 兴城市| 合水县| 平安县| 唐山市| 南丹县| 界首市| 卢湾区| 磴口县| 遵义市| 石河子市| 大兴区| 津市市|