王貞麗,王曉雯,付曉菁,張海霞,趙文文,陳雪紅
[摘要]目的? ?探討隱丹參酮(CPT)對(duì)肝癌細(xì)胞凋亡和內(nèi)質(zhì)網(wǎng)應(yīng)激的影響。 方法? ?HepG2細(xì)胞分別用不同濃度的CPT處理48 h,采用MTT法、乳酸脫氫酶釋放法和細(xì)胞劃痕法檢測(cè)細(xì)胞的生長和遷移能力,Annexin V FITC-PI染色后流式細(xì)胞分析術(shù)檢測(cè)細(xì)胞凋亡率,Western blot法檢測(cè)caspase3、重鏈結(jié)合蛋白(BIP)和蛋白激酶R樣內(nèi)質(zhì)網(wǎng)激酶(p-PERK)的表達(dá)水平。 結(jié)果? ?與陰性對(duì)照組相比,CPT作用后HepG2細(xì)胞的生長和遷移能力減弱(F=41.10~126.40,P<0.01),凋亡蛋白caspase3的活化水平升高(F=156.90,P<0.01)。15 μmol/L CPT可以使HepG2細(xì)胞中內(nèi)質(zhì)網(wǎng)應(yīng)激蛋白BIP和p-PERK的表達(dá)水平明顯下降(F=57.20、49.92,P<0.01)。結(jié)論? ?CPT對(duì)肝癌細(xì)胞的生長和遷移有抑制作用,并且能抑制內(nèi)質(zhì)網(wǎng)應(yīng)激和促進(jìn)凋亡。
[關(guān)鍵詞]隱丹參酮;Hep G2細(xì)胞;細(xì)胞增殖;細(xì)胞運(yùn)動(dòng);細(xì)胞凋亡;內(nèi)質(zhì)網(wǎng)應(yīng)激
[中圖分類號(hào)]R284;R329.25
[文獻(xiàn)標(biāo)志碼]A
[文章編號(hào)]2096-5532(2022)04-0602-05
doi:10.11712/jms.2096-5532.2022.58.128[HT]
[開放科學(xué)(資源服務(wù))標(biāo)識(shí)碼(OSID)]
[網(wǎng)絡(luò)出版]https://kns.cnki.net/kcms/detail/37.1517.R.20220822.1543.002.html;[JY]2022-08-2309:49:42
EFFECT OF CRYPTOTANSHINONE ON APOPTOSIS AND ENDOPLASMIC RETICULUM STRESS IN HEPATOMA CELLS
WANG Zhenli, WANG Xiaowen, FU Xiaojing, ZHANG Haixia, ZHAO Wenwen, CHEN Xuehong
(Quality Management Section of Qingdao Special Service Sanatorium Center of PLA Navy, Qingdao 266071, China)
[ABSTRACT]Objective To investigate the effect of cryptotanshinone (CPT) on apoptosis and endoplasmic reticulum stress in hepatoma cells.
Methods HepG2 cells were treated with different concentrations of CPT for 48 h. MTT assay, lactate dehydrogenase release assay, and wound healing assay were used to measure cell growth and migration abilities; flow cytometry was used to measure cell apoptosis rate after Annexin V FITC-PI staining; Western blot was used to measure the expression levels of caspase3, heavy chain-binding protein (BIP), and protein kinase R-like endoplasmic reticulum kinase (p-PERK).
Results Compared with the negative control group, the CPT treatment group had significant reductions in the growth and migration abilities of HepG2 cells (F=41.10-126.40,P<0.01) and a significant increase in the activation level of the apoptotic protein caspase3 (F=156.90,P<0.01). CPT at a concentration of 15 μmol/L significantly reduced the expression levels of the endoplasmic reticulum stress proteins BIP and p-PERK in HepG2 cells (F=57.20,49.92;P<0.01).
Conclusion CPT can inhibit the growth and migration of HepG2 cells, and it can also inhibit endoplasmic reticulum stress and promote apoptosis.
[KEY WORDS] cryptotanshinone; Hep G2 cells; cell proliferation; cell movement; apoptosis; endoplasmic reticulum stress
肝癌是一種常見的惡性腫瘤[1]。目前常用的肝癌化療藥物5-氟尿嘧啶和鉑類(順鉑)經(jīng)過多年臨床應(yīng)用證實(shí)存在大量的副作用和不良反應(yīng)。目前針對(duì)肝癌治療的化學(xué)藥物和新的靶向藥物的研究層出不窮,但化療藥物耐藥性和預(yù)后不良的弊端仍然存在。近年來,中醫(yī)藥以其療效顯著、副作用小、原料經(jīng)濟(jì)方便等特點(diǎn),受到世界各國的高度關(guān)注[2]。因此,尋求療效優(yōu)越、不良反應(yīng)小的中藥是肝癌治療的潛在方向。丹參作為一種中藥,廣泛應(yīng)用于動(dòng)脈粥樣硬化、阿爾茨海默病、高脂血癥、肝纖維化、慢性腎衰竭和婦科疾病等多種疾病的治療,無嚴(yán)重副作用[3]。丹參的水溶性成分具有良好活性,隨著研究的深入,發(fā)現(xiàn)其脂溶性成分也具有抗結(jié)直腸癌、前列腺癌和肺癌等腫瘤的作用[4-5]。隱丹參酮(CPT)是丹參的主要脂溶性提取物。有研究表明,CPT對(duì)多種類型的癌細(xì)胞有直接的細(xì)胞毒性作用[6-8]。本研究旨在探討CPT對(duì)肝癌細(xì)胞凋亡和目前報(bào)道較多的內(nèi)質(zhì)網(wǎng)應(yīng)激的影響。
1材料與方法
1.1實(shí)驗(yàn)材料
CPT(晨光生物,中國陜西);RPMI 1640(基諾,中國江蘇);胎牛血清(Biological Industrie,以色列);乳酸脫氫酶(LDH)細(xì)胞毒性檢測(cè)試劑盒(碧云天,中國上海);內(nèi)質(zhì)網(wǎng)應(yīng)激抗體試劑盒、cleaved caspase3、caspase3(CST,美國);MTT細(xì)胞增殖及細(xì)胞毒性檢測(cè)試劑盒、PBS緩沖液、衣霉素(索萊寶,中國北京);Annexin V FITC-PI凋亡試劑盒(BD,美國)。
1.2實(shí)驗(yàn)方法
1.2.1細(xì)胞培養(yǎng)肝癌HepG2細(xì)胞(來源于青島大學(xué)基礎(chǔ)醫(yī)學(xué)院細(xì)胞凍存庫)用含體積分?jǐn)?shù)0.10胎牛血清和青霉素/鏈霉素溶液的RPMI 1640培養(yǎng)液,在37 ℃、含體積分?jǐn)?shù)0.05 CO2的無菌培養(yǎng)箱中培養(yǎng)。
1.2.2MTT法檢測(cè)細(xì)胞活性將HepG2細(xì)胞接種于96孔板中,12 h后加CPT處理,加藥濃度分別為0、1、5、10、15和30 μmol/L。CPT處理48 h后,加入MTT溶液,繼續(xù)培養(yǎng)3 h后加入Formazan溶解液,用酶標(biāo)儀檢測(cè)570 nm波長處每孔吸光度。
1.2.3LDH釋放法檢測(cè)細(xì)胞毒性HepG2細(xì)胞加CPT處理(其濃度分別為0、1和15 μmol/L),作用48 h后檢測(cè)LDH釋放量,用酶標(biāo)儀檢測(cè)490 nm波長處每孔吸光度。
1.2.4細(xì)胞劃痕實(shí)驗(yàn)將HepG2細(xì)胞接種于背面畫好橫線的6孔板中,培養(yǎng)12 h后用槍頭垂直于直線劃痕,以PBS清洗,加CPT處理(CPT用無血清培養(yǎng)液稀釋,濃度分別為0、1和15 μmol/L)。置于37 ℃、含體積分?jǐn)?shù)0.05 CO2無菌環(huán)境中培養(yǎng),分別于培養(yǎng)0、48 h時(shí)取樣,拍照。
1.2.5Western blot法檢測(cè)凋亡和內(nèi)質(zhì)網(wǎng)應(yīng)激相關(guān)蛋白表達(dá)收集CPT(0、1和(或)15 μmol/L)作用48 h后的HepG2細(xì)胞,提取蛋白。取30 μg蛋白,于搖床上用1 g/L脫脂奶粉室溫封閉2 h,分別加入cleaved caspase3、重鏈結(jié)合蛋白(BIP)、蛋白激酶R樣內(nèi)質(zhì)網(wǎng)激酶(p-PERK)一抗(1∶1 000),4 ℃過夜,以PBST清洗3次,加入二抗(1∶10 000),室溫?fù)u床孵育2 h,以PBST清洗,滴加ECL化學(xué)發(fā)光液在顯影儀中進(jìn)行化學(xué)發(fā)光成像。
1.2.6流式細(xì)胞術(shù)檢測(cè)細(xì)胞凋亡分別用濃度為1、15 μmol/L的CPT處理HepG2細(xì)胞48 h,然后用預(yù)冷PBS輕洗細(xì)胞2次,消化細(xì)胞。取1×104個(gè)細(xì)胞用Binding Buffer重懸后置于1.5 mL EP管中,每管加入5 μL FITC輕輕混勻,避光孵育15~30 min。加入5 μL PI和400 μL Binding Buffer,上機(jī)檢測(cè)調(diào)亡細(xì)胞。最后使用Flow Jo軟件分析細(xì)胞凋亡情況。
1.3統(tǒng)計(jì)學(xué)分析
采用Prism 5.0軟件進(jìn)行統(tǒng)計(jì)學(xué)分析。本研究所有實(shí)驗(yàn)均進(jìn)行3次獨(dú)立實(shí)驗(yàn),取均值。所得數(shù)據(jù)以[AKx-D]±s形式表示,組間比較采用單因素方差分析。統(tǒng)計(jì)學(xué)檢驗(yàn)均為雙側(cè)檢驗(yàn),以P<0.05為差異有統(tǒng)計(jì)學(xué)意義。
2結(jié)果
2.1CPT對(duì)HepG2細(xì)胞生長和遷移的作用
MTT法檢測(cè)結(jié)果顯示,CPT對(duì)HepG2細(xì)胞生長的抑制作用是呈梯度的。與陰性對(duì)照組相比,10、15、30 μmol/L CPT組HepG2細(xì)胞的生存率明顯下降,差異有統(tǒng)計(jì)學(xué)意義(F=126.40,P<0.001)(圖1A)。LDH釋放法檢測(cè)結(jié)果顯示,15 μmol/L CPT組HepG2細(xì)胞的LDH釋放量明顯升高,與陰性對(duì)照組相比,差異具有統(tǒng)計(jì)學(xué)意義(F=41.10,P<0.001)(圖1B)。細(xì)胞劃痕實(shí)驗(yàn)結(jié)果顯示,細(xì)胞劃痕48 h后,與陰性對(duì)照組相比,15 μmol/L CPT組的閉合率顯著降低,差異具有統(tǒng)計(jì)學(xué)意義(F=96.18,P<0.001)(圖1C)。
2.2CPT對(duì)HepG2細(xì)胞凋亡的影響
Western blot檢測(cè)結(jié)果顯示,與陰性對(duì)照組和1 μmol/L CPT組相比,15 μmol/L CPT組cleaved caspase3的表達(dá)水平增高,差異具有統(tǒng)計(jì)學(xué)意義(F=156.90,P<0.001)(圖2A)。流式細(xì)胞分析術(shù)檢測(cè)結(jié)果顯示,與陰性對(duì)照組相比,15 μmol/L CPT組細(xì)胞凋亡率明顯升高,差異具有統(tǒng)計(jì)學(xué)意義(F=1 090.00,P<0.001)(圖2B、表1)。
2.3CPT對(duì)HepG2細(xì)胞內(nèi)質(zhì)網(wǎng)應(yīng)激的影響
Western blot檢測(cè)結(jié)果顯示,與陰性對(duì)照組相比,15 μmol/L CPT組細(xì)胞中p-PERK和BIP蛋白的表達(dá)水平明顯降低,差異具有統(tǒng)計(jì)學(xué)意義(F=57.20、49.92,P<0.001);與1 μmol/L CPT組相比較,15 μmol/L CPT組細(xì)胞中BIP蛋白的表達(dá)水平明顯降低,差異均具有統(tǒng)計(jì)學(xué)意義(F=49.92,P<0.001)(圖3A)。在15 μmol/L CPT作用下,與未經(jīng)衣霉素預(yù)處理組相比,經(jīng)衣霉素預(yù)處理組HepG2細(xì)胞中BIP的表達(dá)水平明顯升高,差異具有統(tǒng)計(jì)學(xué)意義(F=41.09,P<0.001)(圖3B)。
3討論
研究發(fā)現(xiàn),CPT作為從丹參中提取的脂溶性單體,對(duì)腫瘤有很好的療效[7-9]。本文體外細(xì)胞學(xué)研究結(jié)果表明,CPT可以抑制HepG2細(xì)胞的生長,與陰性對(duì)照組相比,10、15、30 μmol/L CPT組HepG2細(xì)胞的生存率明顯下降。15 μmol/L的CPT可以破壞HepG2細(xì)胞的細(xì)胞膜,引起LDH釋放量明顯增加。另外,在細(xì)胞劃痕實(shí)驗(yàn)中,相對(duì)于陰性對(duì)照組,15 μmol/L CPT組HepG2細(xì)胞的遷移水平降低,差異具有統(tǒng)計(jì)學(xué)意義。上述結(jié)果證實(shí)CPT可以減弱HepG2細(xì)胞的生長和遷移能力。
CPT誘導(dǎo)的細(xì)胞生長和增殖抑制主要是由于細(xì)胞凋亡[10-11]。有研究表明,CPT可以誘導(dǎo)HeLa、DU145和Hep3B細(xì)胞凋亡,其機(jī)制主要是誘導(dǎo)凋亡蛋白caspase3的活化和抑制抗凋亡蛋白Bcl-xL等的表達(dá)[6,12-13]。CPT被普遍認(rèn)為是一種有效的信號(hào)轉(zhuǎn)導(dǎo)和轉(zhuǎn)錄激活因子3(STAT3)抑制劑。相關(guān)文獻(xiàn)報(bào)道,CPT可以快速抑制DU145細(xì)胞中STAT3 Tyr705的磷酸化,而后與細(xì)胞質(zhì)中的STAT3分子共定位,并通過直接結(jié)合到STAT3的SH2域來抑制STAT3二聚體的形成[12]。有文獻(xiàn)報(bào)道,STAT3可以調(diào)節(jié)細(xì)胞的生長和凋亡[13]。本研究結(jié)果表明,CPT可以活化細(xì)胞凋亡蛋白caspase3,從而誘導(dǎo)HepG2細(xì)胞發(fā)生凋亡。
內(nèi)質(zhì)網(wǎng)是一種多功能細(xì)胞器,可以參與維持細(xì)胞穩(wěn)態(tài)和支持細(xì)胞存活[14-15]。外界的刺激可以使其內(nèi)部錯(cuò)誤蛋白不斷積累,造成內(nèi)質(zhì)網(wǎng)應(yīng)激[16]。當(dāng)這種積累超過臨界閾值時(shí),未折疊蛋白反應(yīng)(UPR)補(bǔ)救信號(hào)轉(zhuǎn)導(dǎo)途徑會(huì)被激活,該途徑主要涉及3個(gè)內(nèi)質(zhì)網(wǎng)跨膜傳感器(IRE1α、PERK和ATF6)介導(dǎo)的通路[17-18]。相關(guān)研究表明,腫瘤細(xì)胞通過UPR適應(yīng)低糖環(huán)境,促進(jìn)腫瘤細(xì)胞的生存[19-21]。此外,有研究證實(shí),UPR通過抑制炎癥和凋亡信號(hào)通路進(jìn)行細(xì)胞保護(hù)[22-25]。本研究結(jié)果表明,CPT可以有效地抑制HepG2細(xì)胞中內(nèi)質(zhì)網(wǎng)應(yīng)激的發(fā)生。
綜上所述,CPT對(duì)HepG2細(xì)胞的生長和遷移有抑制作用,在促進(jìn)HepG2細(xì)胞凋亡的同時(shí)可抑制內(nèi)質(zhì)網(wǎng)應(yīng)激。本研究為CPT作為一種有潛力的抗肝癌藥物提供了證據(jù)。
[參考文獻(xiàn)]
[1]MILLER K D, FIDLER-BENAOUDIA M, KEEGAN T H, et al. Cancer statistics for adolescents and young adults, 2020[J]. CA: A Cancer Journal for Clinicians, 2020,70(6):443-459.
[2]ONG M, PENG J, JIN X L, et al. Chinese herbal medicine for the optimal management of polycystic ovary syndrome[J]. The American Journal of Chinese Medicine, 2017,45(3):405-422.
[3]ZHOU L M, ZUO Z, CHOW M S S. Danshen: an overview of its chemistry, pharmacology, pharmacokinetics, and clinical use[J]. The Journal of Clinical Pharmacology, 2005,45(12):1345-1359.
[4]張蒲容,呂青. 丹參酮ⅡA抗乳腺癌作用機(jī)制的實(shí)驗(yàn)研究[J]. 四川大學(xué)學(xué)報(bào)(醫(yī)學(xué)版), 2009,40(2):245-249.
[5]CAI Y E, ZHANG W J, CHEN Z R, et al. Recent insights into the biological activities and drug delivery systems of tanshinones[J]. International Journal of Nanomedicine, 2016,11:121-130.
[6]葉因濤,徐文清,仲巍. 隱丹參酮對(duì)宮頸癌Hela細(xì)胞增殖及細(xì)胞凋亡的影響[J]. 中國中藥雜志, 2010,35(1):118-121.
[7]XU D F, LIN T H, LI S S, et al. Cryptotanshinone suppresses androgen receptor-mediated growth in androgen dependent and castration resistant prostate cancer cells[J]. Cancer Letters, 2012,316(1):11-22.
[8]CHEN X P, GUO J J, BAO J L, et al. The anticancer properties of Salvia miltiorrhiza Bunge (Danshen): a systematic review[J]. Medicinal Research Reviews, 2014,34(4):768-794.
[9]CAO L, HONG W P, CAI P H, et al. Cryptotanshinone strengthens the effect of gefitinib against non-small cell lung cancer through inhibiting transketolase[J]. European Journal of Pharmacology, 2021,890:173647.
[10]KIM J H, JEONG S J, KWON T R, et al. Cryptotanshinone enhances TNF-α-induced apoptosis in chronic myeloid leukemia KBM-5 cells[J]. Apoptosis: an International Journal on Programmed Cell Death, 2011,16(7):696-707.
[11]PARK I J, KIM M J, PARK O J, et al. Cryptotanshinone induces ER stress-mediated apoptosis in HepG2 and MCF7 cells[J]. Apoptosis: an International Journal on Programmed Cell Death, 2012,17(3):248-257.
[12]SHIN D S, KIM H N, SHIN K D, et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells[J]. Cancer Research, 2009,69(1):193-202.
[13]LEE W Y, CHEUNG C C, LIU K W, et al. Cytotoxic effects of tanshinones from Salvia miltiorrhiza on doxorubicin-resis-tant human liver cancer cells[J]. Journal of Natural Products, 2010,73(5):854-859.
[14]RON D, WALTER P. Signal integration in the endoplasmic Reticulum unfolded protein response[J]. Nature Reviews Molecular Cell Biology, 2007,8(7):519-529.
[15]LYNES E M, SIMMEN T. Urban planning of the endoplasmic Reticulum (ER): how diverse mechanisms segregate the many functions of the ER[J]. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, 2011,1813(10):1893-1905.
[16]HETZ C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond[J]. Nature Reviews Molecular Cell Biology, 2012,13(2):89-102.
[17]TABAS I, RON D. Integrating the mechanisms of apoptosis induced by endoplasmic Reticulum stress[J]. Nature Cell Bio-logy, 2011,13(3):184-190.
[18]HOLLIEN J, WEISSMAN J S. Decay of endoplasmic Reticulum-localized mRNAs during the unfolded protein response[J]. Science (New York, N Y), 2006,313(5783):104-107.
[19]CORAZZARI M, GAGLIARDI M, FIMIA G M, et al. Endoplasmic Reticulum stress, unfolded protein response, and cancer cell fate[J]. Frontiers in Oncology, 2017,7:78.
[20]GIAMPIETRI C, PETRUNGARO S, CONTI S, et al. Cancer microenvironment and endoplasmic Reticulum stress response[J]. Mediators of Inflammation, 2015,2015:417281.
[21]RYDER C, MCCOLL K, ZHONG F, et al. Acidosis promotes Bcl-2 family-mediated evasion of apoptosis: involvement of acid-sensing G protein-coupled receptor Gpr65 signaling to Mek/Erk[J]. The Journal of Biological Chemistry, 2012,287(33):27863-27875.
[22]ZHANG J, ZHOU Y, LI N, et al. Curcumol overcomes TRAIL resistance of non-small cell lung cancer by targeting NRH: quinone oxidoreductase 2 (NQO2)[J]. Advanced Science (Weinheim, Baden-Wurttemberg, Germany), 2020,7(22):2002306.
[23]OUTINEN P A, SOOD S K, PFEIFER S I, et al. Homocysteine-induced endoplasmic Reticulum stress and growth arrest leads to specific changes in gene expression in human vascular endothelial cells[J]. Blood, 1999,94(3):959-967.
[24]CUBILLOS-RUIZ J R, BETTIGOLE S E, GLIMCHER L H. Tumorigenic and immunosuppressive effects of endoplasmic Reticulum stress in cancer[J]. Cell, 2017,168(4):692-706.
[25]TIAN J X, MO J M, XU L, et al. Scoulerine promotes cell viability reduction and apoptosis by activating ROS-dependent endoplasmic Reticulum stress in colorectal cancer cells[J]. Chemico-Biological Interactions, 2020,327:109184.
(本文編輯馬偉平)
青島大學(xué)學(xué)報(bào)(醫(yī)學(xué)版)2022年4期