張思敏, 杜文輝, 王敏, 陳芬, 蔡小芳△
lncRNA FEZF1-AS1調控miR-363-3p影響LPS誘導的血管內皮細胞活力及凋亡*
張思敏1, 杜文輝1, 王敏1, 陳芬2, 蔡小芳1△
(華中科技大學同濟醫(yī)學院1附屬武漢兒童醫(yī)院急診科,2附屬協(xié)和醫(yī)院心內科,湖北 武漢 430022)
探討長鏈非編碼RNA (lncRNA) FEZF1-AS1調控微小RNA-363-3p (miR-363-3p)影響脂多糖(LPS)誘導的血管內皮細胞活力及凋亡的作用機制。體外培養(yǎng)人臍靜脈血管內皮細胞(HUVECs),分別將pcDNA-NC、pcDNA-FEZF1-AS1、anti-miR-NC、anti-miR-363-3p、miR-NC及miR-363-3p mimics轉染至HUVECs,并給予LPS刺激24 h;采用RT-qPCR檢測FEZF1-AS1和miR-363-3p的表達量;MTT法檢測細胞活力;流式細胞術檢測細胞凋亡率;雙螢光素酶報告基因實驗驗證FEZF1-AS1與miR-363-3p的靶向調控作用; Western blot法檢測細胞周期蛋白D1 (cyclin D1)、cleaved caspase-3和Ki67的表達量。與control組比較, LPS組FEZF1-AS1的表達水平顯著降低(<0.05), miR-363-3p的表達水平顯著升高(<0.05);與pcDNA-NC+LPS組比較,pcDNA-FEZF1-AS1+LPS組細胞存活率顯著升高(<0.05),細胞凋亡率顯著降低(<0.05), cyclin D1和Ki67蛋白水平顯著升高(<0.05), cleaved caspase-3蛋白水平顯著降低(<0.05);與anti-miR-NC+LPS組比較, anti-miR-363-3p+LPS組細胞存活率顯著升高(<0.05),細胞凋亡率顯著降低(<0.05), cyclin D1和Ki67蛋白水平顯著升高(<0.05), cleaved caspase-3蛋白水平顯著降低(<0.05);雙螢光素酶報告基因實驗證實FEZF1-AS1可靶向結合miR-363-3p;與miR-NC+pcDNA-FEZF1-AS1+LPS組比較, miR-363-3p+pcDNA-FEZF1-AS1+LPS組細胞存活率顯著降低(<0.05),細胞凋亡率顯著升高(<0.05), cyclin D1和Ki67蛋白水平顯著降低(<0.05), cleaved caspase-3蛋白水平顯著升高(<0.05)。過表達FEZF1-AS1可抑制miR-363-3p表達從而降低LPS誘導的血管內皮細胞凋亡率,增加其細胞活力。
長鏈非編碼RNA;FEZF1-AS1;微小RNA-363-3p;膿毒癥;血管內皮細胞;細胞活力;細胞凋亡
膿毒癥是臨床常見疾病之一,近年來,膿毒癥發(fā)病率與死亡率逐年上升,已嚴重威脅人類生命安全,研究表明脂多糖(lipopolysacharide, LPS)是引起膿毒癥的重要原因之一,并可促進多器官衰竭等發(fā)生[1-3]。長鏈非編碼RNA(long noncoding RNA, lncRNA)表達異??捎绊懭遂o脈內皮細胞增殖及凋亡[4]。lncRNA FEZF1-AS1表達下調可影響人晶狀體上皮細胞增殖及遷移[5]。通過生物信息學分析顯示微小RNA-363-3p (microRNA-363-3p, miR-363-3p)可能是FEZF1-AS1的靶基因,研究表明miR-363-3p在骨關節(jié)炎軟骨細胞中表達上調,同時可促進軟骨細胞凋亡[6]。但FEZF1-AS1是否可通過調控miR-363-3p表達從而影響膿毒癥時血管內皮細胞活力及凋亡尚未可知。因此,本研究采用LPS處理人血管內皮細胞,探討FEZF1-AS1對細胞活力及凋亡的影響及其對miR-363-3p的靶向調控作用。
人臍靜脈血管內皮細胞(human umbilical vein endothelial cells,HUVECs)購自ATCC。RPMI-1640培養(yǎng)液和胎牛血清購自Gibco; LPS購自Sigma; Lipofectamine 2000和pcDNA3.1購自Invitrogen; Trizol、反轉錄及實時熒光定量PCR試劑盒均購自TaKaRa; miR-363-3p特異性寡核苷酸抑制物(anti-miR-363-3p)及其陰性對照(anti-miR-NC)、miR-363-3p寡核苷酸模擬物(miR-363-3p mimics)及其陰性對照序列(miR-NC)、FEZF1-AS1小分子干擾RNA(si-FEZF1-AS1)和亂序無意義陰性對照序列(si-NC)購自上海吉瑪制藥技術有限公司; MTT購自上海晶抗生物工程有限公司; RIPA裂解液和Annexin V-FITC/PI細胞凋亡試劑盒購自Sigma;雙螢光素酶報告基因檢測試劑盒購自Promega;兔抗人細胞周期蛋白D1 (cyclin D1)、增殖標記蛋白Ki67和cleaved caspase-3抗體購自CST;辣根過氧化物酶標記的山羊抗兔IgG Ⅱ抗購自武漢博士德生物工程有限公司。
2.1實驗分組血管內皮細胞培養(yǎng)于含10%胎牛血清的RPMI-1640培養(yǎng)液中,置于37℃、5% CO2恒溫培養(yǎng)箱內培養(yǎng),待細胞生長至80%融合度時進行傳代培養(yǎng),取對數(shù)生長期內皮細胞,0.25%胰蛋白酶消化,調整細胞密度為1×108/L,按照每孔100 μL接種于96孔板,使用100 μg/L的LPS處理24 h[7],記作LPS組。同時將正常培養(yǎng)的細胞作為對照(control)組。分別將pcDNA-NC、pcDNA-FEZF1-AS1、anti-miR-NC、anti-miR-363-3p、miR-NC和miR-363-3p mimics轉染至血管內皮細胞48 h,隨后使用100 μg/L LPS處理24 h,根據(jù)轉染序列不同分別分為pcDNA-NC+LPS組、pcDNA-FEZF1-AS1+LPS組、anti-miR-NC+LPS組、anti-miR-363-3p+LPS組、miR-NC+pcDNA-FEZF1-AS1+LPS組和miR-363-3p+pcDNA-FEZF1-AS1+LPS組。
2.2RT-qPCR檢測FEZF1-AS1和miR-363-3p的表達水平采用Trizol法提取細胞中總RNA,應用紫外分光光度計測定RNA濃度。根據(jù)反轉錄試劑盒說明書操作將總RNA反轉錄合成cDNA。FEZF1-AS1的正向引物序列為5’-GAAGACACGGCAGCTGTAAG-3’,反向引物序列為5’-AGCTCTCTTCCTTTCCACCC-3’;miR-363-3p的正向引物5’-AAATTGCACGGTATCCATCTGT-3’,反向引物序列為5’-AAACTTCGTGGGTAGTGTCAAG-3’;β-actin的正向引物序列為5’-TGCTGTCCCTGTATGCCTCT-3’,反向引物序列為5’-TGATGTCACGCACGATTT-3’;U6的正向引物序列為5’-ATTGGAACGATACAGAGAAGATT-3’,反向引物序列為5’-GGAACGCTTCACGAATTTG-3’,引物均由上海生工生物工程股份有限公司合成。反應體系:SYBR Green Master Mix 10 μL,正反向引物0.8 μL,cDNA 1 μL,ddH2O補足體系至20 μL。反應條件:95℃ 2 min; 95℃ 15 s, 60℃ 30 s, 72℃ 30 s,共循環(huán)40次。FEZF1-AS1以β-actin為內參照, miR-363-3p以U6為內參照,采用2-ΔΔCt法計算FEZF1-AS1和miR-363-3p的表達水平。
2.3MTT法檢測細胞存活率收集各組對數(shù)生長期血管內皮細胞接種至96孔板(每孔5×103個),每組設置3個復孔,每孔加入20 μL MTT試劑,繼續(xù)培養(yǎng)4 h,棄上清,每孔加入150 μL二甲基亞砜(dimethylsulfoxide,DMSO),應用酶標儀檢測各孔在波長為490 nm處的相對吸光度()值,計算細胞存活率(%)=(各實驗組值-空白對照組值)/(正常對照組值-空白對照組值)×100%。實驗重復3次。
2.4流式細胞術檢測細胞凋亡率收集各組對數(shù)生長期血管內皮細胞,預冷PBS洗滌,4℃條件下經15 984×離心10 min,棄上清,預冷PBS再次洗滌,棄上清,加入500 μL Binding Buffer懸浮細胞,參照細胞凋亡試劑盒說明書依次加入5 μL Annexin V-FITC與5 μL PI,充分混勻,室溫避光孵育10 min,應用流式細胞儀檢測細胞凋亡率。
2.5雙螢光素酶報告基因檢測FEZF1-AS1的靶基因StarBase預測顯示FEZF1-AS1與miR-363-3p存在靶向關系,構建野生型載體WT-FEZF1-AS1和突變型載體MUT-FEZF1-AS1,分別將miR-NC和miR-363-3p mimics與WT-FEZF1-AS1和MUT-FEZF1-AS1共轉染至LPS處理的血管內皮細胞,收集細胞并檢測螢光素酶活性。分別將pcDNA-NC、pcDNA-FEZF1-AS1、si-NC和si-FEZF1-AS1轉染至血管內皮細胞, RT-qPCR檢測細胞中miR-363-3p的表達水平。
2.6Western blot檢測cyclin D1、cleaved caspase-3和Ki67蛋白表達收集各組對數(shù)生長期血管內皮細胞,加入RIPA裂解液,冰上裂解30 min,4℃條件下經999×離心15 min,提取細胞總蛋白。采用BCA法檢測細胞蛋白濃度。取40 μg蛋白樣品與上樣緩沖液充分混勻后點樣,經SDS-PAGE分離蛋白,轉膜、封閉,加入cyclin D1、cleaved caspase-3、Ki67與β-actin Ⅰ抗(1∶1 000),4℃孵育24 h,TBST洗膜,加入Ⅱ抗(1∶5 000),TBST洗膜,ECL顯影,應用自動凝膠成像系統(tǒng)分析各蛋白條帶。
采用SPSS 21.0統(tǒng)計學軟件分析數(shù)據(jù),計量資料以( mean±SD)表示,且均符合正態(tài)分布,兩組間比較采用獨立樣本檢驗,多組間比較采用單因素方差分析,以<0.05為差異有統(tǒng)計學意義。
與control組比較, LPS組細胞中FEZF1-AS1的表達水平顯著降低(<0.05), miR-363-3p的表達水平顯著升高(<0.05),見表1。
表1 LPS處理的HUVECs中FEZF1-AS1和miR-363-3p的表達情況
*<0.05control group.
與control組比較, LPS組細胞存活率顯著降低(<0.05),細胞凋亡率顯著升高(<0.05), cyclin D1和Ki67蛋白水平顯著降低(<0.05), cleaved caspase-3蛋白水平顯著升高(<0.05);與pcDNA-NC+LPS組比較, pcDNA-FEZF1-AS1+LPS組細胞活力顯著升高(<0.05),細胞凋亡率顯著降低(<0.05), cyclin D1和Ki67蛋白水平顯著升高(<0.05), cleaved caspase-3蛋白水平顯著降低(<0.05); LPS組與pcDNA-NC+LPS組各項指標比較差異無統(tǒng)計學意義(0.05),見圖1、表2。
Figure 1. Effect of FEZF1-AS1 overexpression on ability and apoptosis of LPS-treated HUVECs. A: flow cytometry to detect apoptosis; B: Western blot detection of Ki67, cyclin D1 and cleaved caspase-3 protein expression. Mean±SD. n=9. *P<0.05 vs control group; #P<0.05 vs pcDNA-NC+LPS group.
表2 過表達FEZF1-AS1對LPS處理的血管內皮細胞活力和凋亡的影響
P<0.05control group;#<0.05pcDNA-NC+LPS group.
與anti-miR-NC組+LPS比較,anti-miR-363-3p+LPS組細胞存活率顯著升高(<0.05),細胞凋亡率顯著降低(<0.05), cyclin D1和Ki67蛋白水平顯著升高(<0.05), cleaved caspase-3蛋白水平顯著降低(<0.05),見圖2、表3。
Figure 2. Western blot detection of cyclin D1, Ki67 and cleaved caspase-3 protein expression. Mean±SD. n=9. *P<0.05 vs anti-miR-NC+LPS group.
表3 低表達miR-363-3p對LPS處理的血管內皮細胞活力和凋亡的影響
*<0.05anti-miR-NC+LPS group.
Starbase預測顯示FEZF1-AS1與miR-363-3p存在結合位點,見圖3。
Figure 3. Starbase predicted the binding sites between FEZF1-AS1 and miR-363-3p.
雙螢光素酶報告基因實驗結果顯示,共轉染野生型載體WT-FEZF1-AS1的細胞實驗中,與miR-NC+LPS組比較,miR-363-3p+LPS組螢光素酶活性顯著降低(<0.05);共轉染突變型載體MUT-FEZF1-AS1的細胞實驗中,miR-363-3p+LPS組螢光素酶活性與miR-NC+LPS組比較差異無統(tǒng)計學意義(0.05),見表4。
表4 miR-NC或miR-363-3p與FEZF1-AS1野生型及突變型報告質粒共轉染LPS處理的血管內皮細胞后雙螢光素酶活性檢測
*<0.05miR-NC+LPS group.
與pcDNA-NC+LPS組比較,pcDNA-FEZF1-AS1+LPS組細胞中miR-363-3p+LPS的表達水平顯著降低(<0.05);與si-NC+LPS組比較, si-FEZF1-AS1+LPS組細胞中miR-363-3p+LPS的表達水平顯著升高(<0.05),見表5。
表5 RT-qPCR檢測miR-363-3p的表達
P<0.05pcDNA-NC+LPS group;#<0.05si-FEZF1-AS1+LPS group.
與miR-NC+pcDNA-FEZF1-AS1+LPS組比較, miR-363-3p+pcDNA-FEZF1-AS1+LPS組細胞存活率顯著降低(<0.05),細胞凋亡率顯著升高(<0.05), cyclin D1和Ki67蛋白水平顯著降低(<0.05), cleaved caspase-3蛋白水平顯著升高(<0.05),見圖4、表6。
Figure 4. Western blot detection of cyclin D1, Ki67 and cleaved caspase-3 protein expression. Mean±SD. n=9. *P<0.05 vs miR-NC+pcDNA-FEZF1-AS1+LPS group.
表6 高表達miR-363-3p可以逆轉FEZF1-AS1高表達對LPS處理的血管內皮細胞活力和凋亡的影響
P<0.05miR-NC+pcDNA-FEZF1-AS1+LPS group.
膿毒癥發(fā)生時可促使血管內皮細胞損傷,炎癥和氧化應激等可促使心肌缺血或心肌損傷從而導致膿毒癥心肌損傷的發(fā)生,既往研究顯示部分基因或信號通路異常表達可參與膿毒癥血管內皮細胞增殖及凋亡等過程從而參與膿毒癥的發(fā)生及發(fā)展[8-10]。但膿毒癥發(fā)病率與死亡率仍逐年增加,因而本研究積極探尋新型lncRNA并分析其對機體膿毒癥時血管內皮細胞活力及凋亡的作用。
FEZF1-AS1在膿毒癥發(fā)生及發(fā)展過程中的作用機制尚未可知,本研究結果顯示LPS誘導的血管內皮細胞中FEZF1-AS1的表達量降低,既往研究顯示FEZF1-AS1在結直腸癌、卵巢癌和前列腺癌中表達異常并可調控細胞增殖及凋亡[11-13],提示FEZF1-AS1在膿毒癥發(fā)生過程中可能發(fā)揮重要調控作用。本研究進一步分析顯示FEZF1-AS1過表達可促進血管內皮細胞存活,抑制細胞凋亡,提示FEZF1-AS1過表達可減輕LPS誘導的血管內皮細胞損傷。研究表明cyclinD1可正向調控細胞周期,促進細胞增殖,cleaved caspase-3高表達可促進細胞凋亡[14-15]。本研究結果顯示FEZF1-AS1過表達后cyclin D1和Ki67蛋白水平升高,cleaved caspase-3蛋白水平降低,進一步證實FEZF1-AS1過表達可調控cyclin D1和cleaved caspase-3表達從而促進血管內皮細胞活力及抑制細胞凋亡。
為探究FEZF1-AS1在血管內皮細胞損傷中的作用機制,本研究通過雙螢光素酶報告基因實驗與qRT-PCR實驗證實FEZF1-AS1可靶向調控miR-363-3p的表達。miR-363-3p在肝癌、非小細胞肺癌和肺腺癌等腫瘤中異常表達并可參與癌癥發(fā)生及發(fā)展過程[16-18]。本研究結果顯示LPS處理后血管內皮細胞中miR-363-3p的表達上調,進一步分析顯示抑制miR-363-3p表達后細胞存活率升高,細胞凋亡率降低,cyclin D1和Ki67蛋白水平升高,cleaved caspase-3蛋白水平降低,提示抑制miR-363-3p表達可促進LPS誘導的血管內皮細胞活力,抑制細胞凋亡,進一步表明FEZF1-AS1可能通過靶向調控miR-363-3p的表達從而發(fā)揮作用。為進一步證實,本研究將FEZF1-AS1過表達聯(lián)合miR-363-3p過表達處理HUVECs,結果顯示細胞活力被顯著抑制,同時細胞凋亡率顯著升高,提示FEZF1-AS1過表達可能靶向抑制miR-363-3p表達從而促進膿毒癥時血管內皮細胞活力及抑制細胞凋亡。
綜上所述,LPS誘導的血管內皮細胞中FEZF1-AS1的表達水平降低,miR-363-3p的表達水平升高,F(xiàn)EZF1-AS1過表達可能靶向抑制miR-363-3p表達而促進血管內皮細胞活力及抑制細胞凋亡從而對血管內皮細胞發(fā)揮保護作用,可為進一步揭示膿毒癥發(fā)病機制提供參考資料。
[1]霍江,李強,于穎群,等. 右美托咪定對LPS所致膿毒癥小鼠急性肺損傷的保護作用[J]. 軍事醫(yī)學, 2015, 39(12):941-943.
Huo J, Li Q, Yu YQ, et al. Dexmedetomidine protects against LPS-induced ALI in septic mice[J]. Mil Med Sci, 2015, 39(12):941-943.
[2]王胤中,史克潔,穆心葦. 早期應用艾司洛爾對LPS誘導膿毒癥大鼠心功能的影響[J]. 安徽醫(yī)藥, 2015, 19(3):422-426.
Wang YZ, Shi KJ, Mu XW. Early influence of esmolol on the cardiac function of LPS induced sepsis rats[J]. Anhui Med Pharm J, 2015, 19(3):422-426.
[3]李林成,劉銘傳,曹夢遠,等. 褪黑素調控微小RNA-9抑制膿毒癥炎性反應的機制[J]. 中華實驗外科雜志, 2019, 36(5):823-826.
Li LC, Liu MC, Cao MY, et al. Melatonin regulates inflammation in sepsis via microRNA-9 and related mechanism[J]. Chin J Exp Surg, 2019, 36(5):823-826.
[4]陳海英,孟濤. LncRNA MALAT1對人臍靜脈內皮細胞HUVEC增殖與凋亡影響[J]. 中國公共衛(wèi)生, 2016, 32(10):1340-1342.
Chen HY, Meng T. Effect of LncRNA MALAT1 on proliferation and apoptosis of human umbilical vein endothelial cells[J]. Chin J Publ Heal, 2016, 32(10):1340-1342.
[5] Wang Y, Chen L, Gu Y, et al. LncRNA FEZF1-AS1 promotes TGF-β2-mediated proliferation and migration in human lens epithelial cells SRA01/04[J]. J Ophthalmol, 2019, 2019:4736203.
[6] Zhang M, Wang Z, Li B, et al. Identification of microRNA?363?3p as an essential regulator of chondrocyte apoptosis in osteoarthritis by targeting NRF1 through the p53?signaling pathway[J]. Mol Med Rep, 2020, 21(3):1077-1088.
[7]何慧敏,李昂,張淑文,等. 組蛋白去乙?;敢种苿榷舅厮卵軆绕ぜ毎麚p傷的影響[J]. 中國危重病急救醫(yī)學, 2012, 23(10):602-604.
He HM, Li A, Zhang SW, et al. The effects of a histone deacetylase(HDAC) inhibitor on endotoxin-induced endothelial cell injury[J]. Chin Criti Care Med, 2012, 23(10):602-604.
[8] Zaky A, Deem S, Bendjelid K, et al. Characterization of cardiac dysfunction in sepsis[J]. Shock, 2014, 41(1): 12-24.
[9]魏雅念,王龍,王鑫,等. 神經調節(jié)蛋白-1對膿毒癥大鼠心肌組織炎癥和心肌細胞凋亡的影響[J]. 山西醫(yī)科大學學報, 2017, 48(8):763-768.
Wei YN, Wang L, Wang X, et al. Effects of neuregulin-1 on myocardial inflammation and cardiomyocyte apoptosis in rats with sepsis[J]. J Shanxi Med Univ, 2017, 48(8):763-768.
[10] 康文,王龍,王倩文,等. 神經調節(jié)蛋白-1對膿毒癥大鼠心肌血管內皮和心臟功能的影響[J]. 臨床急診雜志, 2019, 20(7):521-527.
Kang W, Wang L, Wang QW, et al. Effect ofneuregulin-1onmyocardial vascularendothelium andcardiac functionin septic rats[J]. J Clin Emerg (China), 2019, 20(7):521-527.
[11] Li J, Zhao LM, Zhang C, et al. The lncRNA FEZF1-AS1 promotes the progression of colorectal cancer through regulating OTX1 and targeting miR-30a-5p[J]. Oncol Res, 2020, 28(1):51-63.
[12] Zhao X, Cheng Z, Wang J. Long noncoding RNA FEZF1-AS1 promotes proliferation and inhibits apoptosis in ovarian cancer by activation of JAK-STAT3 pathway[J]. Med Sci Monit, 2018, 24(1):8088-8095.
[13] Zhu LF, Song LD, Xu Q, et al. Highly expressed long non-coding RNA FEZF1-AS1 promotes cells proliferation and metastasis through notch signaling in prostate cancer[J]. Eur Rev Med Pharmacol Sci, 2019, 23(12):5122-5132.
[14] 郭洪宇,趙玉嬌,高博,等. 中電導鈣激活鉀通道調節(jié)表皮生長因子誘導的人臍靜脈內皮細胞體外增殖[J]. 解剖學報, 2016, 47(2):209-215.
Guo HY, Zhao YJ, Gao B, et al. Intermediate conductance Ca2+activated K+channels 3.1 regulates theproliferation of human umbilical vein endothelial cells induced byepidermal growth factor[J]. Acta Anat Sin, 2016, 47(2):209-215.
[15] 林佳瓊,陳美姬,郭瑞鮮,等. 壞死性凋亡介導高糖引起的人臍靜脈內皮細胞損傷[J]. 中國病理生理雜志, 2016, 32(9):1608-1613.
Lin JQ, Chen MJ, Guo RX, et al. Necroptosis mediates high glucose-induced injury in human umbilical veinendothelial cells[J]. Chin J Pathophysiol, 2016, 32(9):1608-1613.
[16] Wang J, Liang H, Ge H, et al. MicroRNA?363?3p inhibits hepatocarcinogenesis by targeting HMGA2 and is associated with liver cancer stage[J]. Mol Med Rep, 2019, 19(2):935-942.
[17] Jiang C, Cao Y, Lei T, et al. MicroRNA-363-3p inhibits cell growth and invasion of non?small cell lung cancer by targeting HMGA2[J]. Mol Med Rep, 2018, 17(2):2712-2718.
[18] Rong H, Chen B, Wei X, et al. Long non-coding RNA XIST expedites lung adenocarcinoma progression through upregulating MDM2 expression via binding to miR-363-3p[J]. Thorac Cancer, 2020, 11(3):659-671.
Effect of lncRNA FEZF1-AS1 on viability and apoptosis of LPS-induced vascular endothelial cells by regulating miR-363-3p expression
ZHANG Si-min1, DU Wen-hui1, WANG Min1, CHEN Fen2, CAI Xiao-fang1
(1,,2,,,,430022,)
To investigate the mechanism of long noncoding RNA (lncRNA) FEZF1-AS1 regulating microRNA-363-3p (miR-363-3p) on the viability and apoptosis of lipopolysaocharide (LPS)-induced vascular endothelial cells.Human umbilical vein endothelial cells (HUVECs) were cultured. pcDNA-NC, pcDNA-FEZF1-AS1, anti-miR-NC, anti-miR-363-3p, miR-NC and miR-363-3p mimics were transfected into the HUVECs and LPS stimulation was applied for 24 h. RT-qPCR was used to detect the expression of FEZF1-AS1 and miR-363-3p. The cell viability was measured by MTT assay. The apoptotic rate was analyzed by flow cytometry. The dual-luciferase reporter experiment was used to verify the targeted regulation of FEZF1-AS1 and miR-363-3p. Western blot was used to determined the expression of cyclin D1, Ki67 and cleaved caspase-3.Compared with control group, the expression level of FEZF1-AS1 in LPS group was significantly reduced (<0.05), and the expression level of miR-363-3p was significantly increased (<0.05). Compared with pcDNA-NC+LPS group, the cell viability in pcDNA-FEZF1-AS1+LPS group was significantly increased (<0.05), the apoptotic rate was significantly reduced (<0.05), the protein levels of cyclin D1 and Ki67 were significantly increased (<0.05), and the protein level of cleaved caspase-3 was significantly reduced (<0.05). Compared with anti-miR-NC+LPS group, the cell viability in anti-miR-363-3p+LPS group was significantly increased (<0.05), the apoptotic rate was significantly reduced (<0.05),the protein levels of cyclin D1 and Ki67 were significantly increased (<0.05), and the protein level of cleaved caspase-3 was significantly reduced (<0.05). Dual-luciferase reporter experiment confirmed that FEZF1-AS1 targeted miR-363-3p. Compared with miR-NC+pcDNA-FEZF1-AS1+LPS group, the cell viability in miR-363-3p+pcDNA-FEZF1-AS1+LPS group was significantly reduced (<0.05), the apoptotic rate was significantly increased (<0.05), the protein levels of cyclin D1 and Ki67 were significantly reduced (<0.05), and the protein level of cleaved caspase-3 was significantly increased (<0.05).Over-expression of FEZF1-AS1 promotes the viability and inhibits apoptosis of LPS induced vascular endothelial cells by inhibiting the expression of miR-363-3p.
Long noncoding RNA; FEZF1-AS1; MicroRNA-363-3p; Sepsis; Vascular endothelial cells; Cell viability; Apoptosis
R363.2;R329.2+5
A
10.3969/j.issn.1000-4718.2020.11.004
1000-4718(2020)11-1945-07
2020-04-29
2020-06-28
國家自然科學基金項目(No.81300196)
Tel: 13343409966; E-mail: msnuz76@163.com
(責任編輯:林白霜,余小慧)