摘要:目的 探究Go-ichi-ni-san復(fù)合物亞基1(GINS1)在肝細(xì)胞癌(HCC)進(jìn)展和化療耐藥中的作用及相關(guān)機(jī)制。方法 通過腫瘤數(shù)據(jù)庫GEPIA2網(wǎng)站檢索并分析GINS1在HCC患者和健康人群中的表達(dá)差異。收集2017年5月—2021年1月新疆醫(yī)科大學(xué)附屬腫瘤醫(yī)院及第一附屬醫(yī)院收治的40例HCC患者病理組織,通過免疫組化染色檢測(cè)GINS1在HCC組織和對(duì)應(yīng)癌旁組織中的表達(dá)差異,分析GINS1表達(dá)水平與HCC臨床TNM分期之間的關(guān)系。Western Blot檢測(cè)GINS1在HCC細(xì)胞系Huh7、Hep3B、Li-7、MHCC97H 和人正常肝細(xì)胞系 QSG7701 中的表達(dá)差異。通過慢病毒轉(zhuǎn)染細(xì)胞的方法構(gòu)建穩(wěn)定敲低GINS1的MHCC97H細(xì)胞株及其陰性對(duì)照細(xì)胞株。通過CCK-8實(shí)驗(yàn)和克隆形成實(shí)驗(yàn)檢測(cè)細(xì)胞增殖能力,劃痕實(shí)驗(yàn)檢測(cè)細(xì)胞遷移能力,Transwell實(shí)驗(yàn)檢測(cè)細(xì)胞侵襲能力,使用奧沙利鉑處理細(xì)胞以檢測(cè)細(xì)胞對(duì)化療藥物的敏感性。構(gòu)建裸鼠負(fù)瘤模型,觀察GINS1敲低對(duì)HCC體內(nèi)生長的影響。Western Blot檢測(cè)各組細(xì)胞Notch通路和JAK/STAT通路蛋白表達(dá)水平。加入Notch受體激動(dòng)劑Jagged-1處理細(xì)胞,分析GINS1與Notch/JAK/STAT通路之間的關(guān)系。計(jì)量資料兩組間比較采用成組t檢驗(yàn),多組間比較采用單因素方差分析,進(jìn)一步兩兩比較采用LSD-t檢驗(yàn)。結(jié)果 GINS1在HCC患者、HCC組織和HCC細(xì)胞系中均表達(dá)上調(diào)(P值均lt;0.05)。GINS1表達(dá)水平與HCC臨床TNM分期正相關(guān)(r=0.822,P=0.011)。與陰性對(duì)照組細(xì)胞相比,敲低GINS1的MHCC97H細(xì)胞增殖、遷移和侵襲活性均降低(P值均lt;0.01),對(duì)奧沙利鉑敏感性增強(qiáng)(Plt;0.01)。與對(duì)照組裸鼠相比,GINS1敲低導(dǎo)致裸鼠形成的腫瘤質(zhì)量和體積均受到顯著抑制(P值均lt;0.001)。與陰性對(duì)照組細(xì)胞相比,在敲低GINS1的MHCC97H細(xì)胞內(nèi),Notch1、Notch3、p-JAK2和p-STAT3表達(dá)水平明顯降低(P值均lt;0.05),JAK2和STAT3總體表達(dá)水平無明顯差異(P值均gt;0.05)。Jagged-1處理后,敲低GINS1的MHCC97H細(xì)胞的增殖、遷移和侵襲活性均有所增加,而細(xì)胞對(duì)奧沙利鉑敏感性有所減弱,p-JAK2、p-STAT3水平升高(P值均lt;0.05)。結(jié)論 GINS1在HCC中表達(dá)上調(diào),并且能夠通過Notch/JAK2/STAT3通路促進(jìn)HCC進(jìn)展和腫瘤細(xì)胞化療耐藥。
關(guān)鍵詞:GINS復(fù)合物;癌,肝細(xì)胞;抗藥性,腫瘤;信號(hào)傳導(dǎo)
基金項(xiàng)目:新疆維吾爾自治區(qū)自然科學(xué)基金(2022D01C245);新疆維吾爾自治區(qū)產(chǎn)學(xué)合作協(xié)同育人項(xiàng)目(MRHT1000023042108)
Effect of Go-Ichi-Ni-San complex subunit 1 on disease progression and chemotherapy resistance in hepatocellular carcinoma
HUO Yishan 1 ,LI Dawei 2 ,DUAN Xiangbing 1 ,MA Yuyu 1 ,ZHANG Guojun 3 ,ZHANG Kainan 4 ,MA Xiumin 1
1. Department of Clinical Laboratory,Clinical Laboratory Center,Tumor Hospital Affiliated to Xinjiang Medical University,Urumqi830000,China;2. Department of Clinical Laboratory,The Fifth Affiliated Hospital of Xinjiang Medical University,Urumqi830000,China;3. Experimental Diagnostic Center,Beijing Tiantan Hospital,Capital Medical University,Beijing 100070,China;4. Department of Clinical Laboratory,Xinjiang Uygur Autonomous Region People’s Hospital,Urumqi 830000,China
Corresponding author:MA Xiumin,maxiumin1210@sohu.com (ORCID:0000-0001-8011-7513)
Abstract:Objective To investigate the role and mechanism of Go-Ichi-Ni-San complex subunit 1 (GINS1) in the progression of hepatocellular carcinoma (HCC) and the development of chemotherapy resistance. Methods The tumor database GEPIA2 was used to analyze the differential expression of GINS1 between HCC patients and healthy individuals,and pathological tissue samples were collected from 40 HCC patients who were admitted to The Affiliated Tumor Hospital of Xinjiang Medical University and the First Affiliated Hospital of Xinjiang Medical University from May 2017 to January 2021. Immunohistochemical staining was used to measure the difference in the expression of GINS1 between HCC tissue and corresponding adjacent tissue,and the correlation between the expression level of GINS1 and the clinical TNM stage of HCC was analyzed. Western blot was also used to measure the difference in the expression of GINS1 between HCC Huh7/Hep3B/Li-7/MHCC97H cell lines and normal human QSG7701hepatocytes. The method of lentivirus transfection was used to establish the MHCC97H cell line with stable GINS1 knockdown and its negative control cell line. CCK-8 assay and colony formation assay were used to measure cell proliferative capacity;scratch assay was used to measure cell migration ability;Transwell assay was used to measure cell invasion ability;cells were treated with oxaliplatin to measure their sensitivity to chemotherapy drugs. Nude mice were used to establish a tumor-bearing model and observe the effect of GINS1 knockdown on the growth of HCC in vivo. Western Blot was used to measure the expression levels of the proteins associated with the Notch pathway and the JAK/STAT pathway. The cells were treated with the Notch receptor agonist Jagged-1 to analyze the association between GINS1 and the Notch/JAK/STAT pathway. The independent-samples t test was used for comparison of continuous data between two groups;a one-way analysis of variance was used for comparison between multiple groups,and the least significant difference t-test was used for further comparison between two groups. Results The expression of GINS1 was upregulated in HCC patients,HCC tissue,and HCC cell lines (all Plt;0.05),and the expression level of GINS1 was positively correlated with the clinical TNM stage of HCC (r=0.822,P=0.011). Compared with the negative control cells,the GINS1-knockdown MHCC97H cells showed significant reductions in proliferation,migration,and invasion activities (all Plt;0.01) and a significantly enhanced sensitivity to oxaliplatin (Plt;0.01). Compared with the nude mice in the control group,GINS1 knockdown caused significant inhibition of tumor weight and volume in vivo in nude mice (all Plt;0.001). Compared with the negative control cells,the GINS1-knockdown MHCC97H cells showed significant reductions in the expression levels of Notch1,Notch3,p-JAK2,and p-STAT3 (all Plt;0.05),while there were no significant differences in the overall expression levels of JAK2 and STAT3 (Pgt;0.05). After Jagged-1 treatment,the GINS1-knockdown MHCC97H cells showed significant increases in proliferation,migration,and invasion activities and a significant reduction in sensitivity to oxaliplatin,as well as significant increases in the levels of p-JAK2 and p-STAT3 (all Plt;0.05). Conclusion GINS1 is upregulated in HCC and can promote HCC progression and chemotherapy resistance through the Notch/JAK2/STAT3 pathway.
Key words:Go-Ichi-Ni-San (GINS) Complex;Carcinoma,Hepatocellular;Drug Resistance,Neoplasm;Signal Transduction
Research funding:Natural Science Foundation of Xinjiang Uygur Autonomous Region (2022D01C245);Xinjiang Uygur Autonomous Region Industy-University Cooperation and Collaborative Education Project (MRHT1000023042108)
肝細(xì)胞癌(HCC)是全球最常見的癌癥之一。與其他常見癌癥(如乳腺癌、肺癌和前列腺癌)的相比,HCC的死亡率每年持續(xù)增加2%~3%,這是因?yàn)镠CC患者確診時(shí)往往處于晚期,而且晚期 HCC 尚無治愈方法[1]。HCC的早期診斷正成為一個(gè)巨大的挑戰(zhàn)。在過去的幾年里,HCC的早期診斷依賴于超聲監(jiān)測(cè)和甲胎蛋白血清學(xué)評(píng)估[2]。然而,這兩種方法的特異性和敏感性不足以準(zhǔn)確地診斷出早發(fā)性HCC[3]。因此,找到HCC早期診斷特異性靶點(diǎn)至關(guān)重要。Go-ichi-ni-san復(fù)合物亞基1(GINS1)屬于異四聚體復(fù)合物GINS的一部分[4]。研究表明,酵母中的GINS復(fù)合物能夠與其他分子組裝成更大的復(fù)合物來參與調(diào)節(jié)細(xì)胞周期的每個(gè)階段和DNA復(fù)制過程[5]。GINS復(fù)合物與哺乳動(dòng)物細(xì)胞中參與DNA復(fù)制的蛋白質(zhì)結(jié)合,以協(xié)助DNA復(fù)制過程[6]。GINS1是GINS復(fù)合物中的關(guān)鍵組成部分,在整個(gè)細(xì)胞周期中維持細(xì)胞增殖活性[7-9]。先前的研究發(fā)現(xiàn),在人類多種腫瘤中GINS1水平升高,例如膠質(zhì)瘤、乳腺癌和膀胱癌[7,10-11]。然而,其在HCC發(fā)展中的作用尚不清楚。本研究通過構(gòu)建穩(wěn)定敲低GINS1的 HCC細(xì)胞系,初步探究了 GINS1對(duì) HCC進(jìn)展的促進(jìn)作用及相關(guān)分子機(jī)制。
1 材料和方法
1.1 細(xì)胞系、實(shí)驗(yàn)動(dòng)物和病理組織切片 人HCC細(xì)胞系 Huh7、Hep3B、Li-7、MHCC97H 購自武漢普諾賽生物公司,人正常肝細(xì)胞系 QSG7701 購自合肥萬物生物公司。所有細(xì)胞使用含10%胎牛血清、1%青霉素/鏈霉素的RPMI-1640培養(yǎng)基,在含5% CO 2 的37 ℃孵箱內(nèi)培養(yǎng)。12 只 4 周齡 BALB/C 雄性裸鼠購自北京斯貝福生物公司,飼養(yǎng)于醫(yī)院潔凈動(dòng)物房內(nèi)。40例HCC患者(T1~T4期各10例)病理組織切片及對(duì)應(yīng)癌旁組織切片來自新疆醫(yī)科大學(xué)附屬腫瘤醫(yī)院及第一附屬醫(yī)院,樣本收集前患者均未接受新輔助化療。
1.2 生物信息學(xué)分析 通過檢索GEPIA2(http://gepia2.cancer-pku.cn/#index)數(shù)據(jù)庫,獲得GINS1在HCC患者和健康人群中的表達(dá)情況,下載差異表達(dá)結(jié)果圖。
1.3 免疫組化染色 本課題組委托上海奧特多生物科技公司制作了組織微陣列(tissue microarray,TMA),該陣列包含 40個(gè) HCC組織樣本和 40個(gè)對(duì)應(yīng)癌旁組織樣本,這些樣本收集于2017年5月—2021年1月。TMA切片用乙醇水化,然后用澄清劑二甲苯使其透明,再嵌入石蠟中。然后使用 H 2 O 2 抑制內(nèi)源性過氧化物酶功能。按照制造商的指示孵育一抗和二抗。使用倒置顯微鏡觀察每張載玻片。免疫組化染色評(píng)估由兩名經(jīng)驗(yàn)豐富的病理學(xué)家獨(dú)立進(jìn)行。測(cè)量腫瘤組織中免疫染色的強(qiáng)度用于評(píng)估GINS1表達(dá),陽性信號(hào)被描述為深棕色。
1.4 Western Blot檢測(cè) 對(duì)于細(xì)胞樣本,將細(xì)胞接種于6孔板內(nèi),長滿后用含蛋白酶抑制劑、磷酸酶抑制劑的RIPA裂解液(上海碧云天公司)裂解細(xì)胞獲得總蛋白。蛋白在金屬浴中煮沸變性,然后通過SDS-PAGE凝膠電泳分離,再轉(zhuǎn)移到聚偏二氟乙烯膜(美國Millipore公司)上。使用5%脫脂牛奶封閉膜2 h,膜與一抗在4 ℃下孵育過夜。用PBS沖洗膜3次,膜與二抗在室溫下孵育1 h。最后,使用ECL化學(xué)發(fā)光液對(duì)蛋白條帶進(jìn)行顯影。對(duì)于組織樣本,首先將組織切成小塊,按每30 mg組織需100 μL裂解液的比例,使用裂解液裂解組織,然后組織在冷凍研磨機(jī)上充分研磨。將組織勻漿靜置10 min,取上清用以后續(xù)試驗(yàn)。抗GAPDH、GINS1、Notch1、Notch3抗體購自美國Proteintech公司,抗Bax、Bad、JAK2、p-JAK2、STAT3和p-STAT3抗體購自美國abcam公司,羊抗兔和羊抗鼠二抗購自上海碧云天公司。
1.5 實(shí)時(shí)熒光定量PCR(qRT-PCR) 使用Trizol試劑(美國Invitrogen公司)提取細(xì)胞RNA,使用SuperScript Ⅳ試劑盒(美國Thermo Fisher公司)將RNA逆轉(zhuǎn)錄合成cDNA,在CFX96 PCR 儀(美國 Bio-Rad 公司)上使用 ChamQ SYBR qPCR Master Mix試劑盒(南京Vazyme公司)對(duì)cDNA進(jìn)行RT-qPCR分析。以GAPDH基因轉(zhuǎn)錄水平歸一化處理各基因轉(zhuǎn)錄水平。各基因引物序列見表1。
1.6 CCK-8增殖實(shí)驗(yàn) 將細(xì)胞以 5 000個(gè)/孔的密度均勻接種在96孔板中。分別在第12、24、48和72 h向每孔加入10 μL CCK-8試劑(上海碧云天公司),繼續(xù)避光孵育2 h,并使用多功能微孔板讀數(shù)儀(新加坡PerkinElmer公司)測(cè)量450 nm處的吸光度。
1.7 克隆形成實(shí)驗(yàn) 將細(xì)胞按500個(gè)/孔的密度接種于6孔板內(nèi),完全培養(yǎng)基連續(xù)培養(yǎng)。待肉眼可見有明顯的細(xì)胞集落形成時(shí),撤去培養(yǎng)基,PBS洗滌3次,用4%多聚甲醛固定細(xì)胞,0.1%結(jié)晶紫染色細(xì)胞,PBS洗滌3次后,在顯微鏡下觀察并計(jì)數(shù)細(xì)胞集落數(shù)量。
1.8 劃痕實(shí)驗(yàn) 將細(xì)胞接種于6孔板內(nèi),待細(xì)胞匯合度達(dá)60%時(shí),撤去培養(yǎng)基,用10 μL移液槍頭對(duì)細(xì)胞進(jìn)行十字劃痕,制造直線“缺口”。以無血清培養(yǎng)基繼續(xù)培養(yǎng),分別在劃痕后0 h和48 h顯微鏡下觀察“缺口”面積變化。
1.9 Transwell侵襲實(shí)驗(yàn) 對(duì)于Transwell侵襲實(shí)驗(yàn),在冰上操作鋪好 Matrigel 膠。在 24 孔 Transwell 板(美國Corning公司)下室中加入含有 20%胎牛血清的完全培養(yǎng)基,上室中加入含有 1 000個(gè)細(xì)胞的無血清培養(yǎng)基。孵育48 h后,用4%多聚甲醛固定細(xì)胞,0.1%結(jié)晶紫染色,使用顯微鏡對(duì)侵襲細(xì)胞進(jìn)行定量分析。
1.10 化療耐藥敏感性檢測(cè) 將HCC細(xì)胞接種于96孔板中,加入終濃度為0、0.1、1、10、50、100 μmol/L的奧沙利鉑藥物(美國MCE公司),處理細(xì)胞48 h。使用CCK-8試劑盒測(cè)定細(xì)胞活性,計(jì)算半數(shù)生長抑制率(IC 50 ),使用GraphPad Prism 8.0 軟件進(jìn)行分析,比較不同處理細(xì)胞IC 50 值變化。以35 μmol/L濃度的奧沙利鉑培養(yǎng)細(xì)胞,觀察細(xì)胞敏感性變化。
1.11 慢病毒轉(zhuǎn)染細(xì)胞 包含GINS1基因表達(dá)干擾質(zhì)粒(shGINS1-#1,shGINS1-#2,shGINS1-#3)及其陰性對(duì)照質(zhì)粒(shNC)的慢病毒載體購自上海吉瑪基因公司。按照說明,將細(xì)胞按5 000個(gè)/孔的密度接種于24孔板內(nèi),分為GINS1敲低組和敲低陰性對(duì)照組,根據(jù)分組分別加入對(duì)應(yīng)的慢病毒-培養(yǎng)基混合液。病毒侵染細(xì)胞24 h后撤去培養(yǎng)基,更換為新鮮完全培養(yǎng)基。48 h后使用含10 mg/mL嘌呤霉素(上海碧云天公司)的完全培養(yǎng)基培養(yǎng)各組細(xì)胞,3天后存活細(xì)胞即為成功轉(zhuǎn)染了慢病毒的細(xì)胞。人類GINS1基因表達(dá)干擾質(zhì)粒中短發(fā)夾RNA(short hairpin RNA,shRNA)序列見表2。
1.12 裸鼠負(fù)瘤模型 12只裸鼠來自北京斯貝福生物公司[生產(chǎn)許可證號(hào):SCXK(京)2019-0010,合格證號(hào):1100111911047350],體質(zhì)量20~22 g。裸鼠在SPF級(jí)環(huán)境中適應(yīng)性飼養(yǎng) 1 周,環(huán)境相對(duì)濕度為 62%~70%,溫度為(23±1.5)℃,12 h/12 h間斷照明,提供充足飼料和無菌水,自由飲食。在隨機(jī)分配為 GINS1 敲低組和對(duì)照組(每組各6只)后,使用注射器在無菌環(huán)境中將細(xì)胞(2×10 6 個(gè)/只)注射到裸鼠背部。腫瘤細(xì)胞接種第21天從裸鼠身上取下腫瘤樣本,稱重、拍照并記錄。腫瘤體積計(jì)算公式:V=(長×寬2 )/2(mm 3 )。
1.13 統(tǒng)計(jì)學(xué)方法 使用 GraphPad Prism 8.0軟件進(jìn)行統(tǒng)計(jì)學(xué)分析。計(jì)量資料以 x ˉ ±s表示,兩組間比較采用成組t檢驗(yàn),多組間比較采用單因素方差分析,進(jìn)一步兩兩比較采用LSD-t檢驗(yàn)。Plt;0.05為差異有統(tǒng)計(jì)學(xué)意義。
2 結(jié)果
2.1 GINS1在HCC中表達(dá)上調(diào) GEPIA2數(shù)據(jù)庫檢索結(jié)果顯示,GINS1在HCC患者中的表達(dá)水平顯著高于健康人群(圖1a)。隨后,在細(xì)胞層面進(jìn)行的蛋白免疫印跡分析結(jié)果表明,GINS1蛋白在4種人類HCC細(xì)胞系中的表達(dá)水平均顯著高于人正常肝細(xì)胞系QSG7701(Huh7:t=5.56,P=0.020;Hep3B:t=7.70,P=0.014;MHCC97H:t=8.62,P=0.010;Li-7:t=6.81,P=0.018),并且在具有高轉(zhuǎn)移潛能的MHCC97H細(xì)胞中表達(dá)水平最高(P=0.010)(圖1b)。為了驗(yàn)證細(xì)胞實(shí)驗(yàn)結(jié)果,本研究對(duì)包含40例HCC組織樣本和對(duì)應(yīng)癌旁組織樣本的TMA進(jìn)行免疫組化染色。結(jié)果表明,與癌旁組織相比,GINS1在HCC組織中表達(dá)水平顯著上調(diào)(P=0.003)(圖1c)。對(duì)T1~T4期HCC組織進(jìn)行蛋白印跡分析顯示,GINS1表達(dá)水平與HCC的T分期正相關(guān)(r=0.822,P=0.011))(圖 1d)。上述結(jié)果提示,GINS1高水平表達(dá)可能是促進(jìn)HCC發(fā)生和發(fā)展的重要因素。
2.2 下調(diào) GINS1表達(dá)抑制 HCC細(xì)胞增殖、遷移和侵襲活性 為了探究GINS1在HCC發(fā)生和發(fā)展過程中的作用,本研究構(gòu)建了穩(wěn)定敲低GINS1的MHCC97H細(xì)胞株(shGINS1-#1,shGINS1-#2,shGINS1-#3)及其陰性對(duì)照細(xì)胞株(shNC)。Western Blot和qRT-PCR結(jié)果表明,shGINS1-#3敲低效率最佳(Plt;0.001),所以后續(xù)GINS1敲低細(xì)胞株均為轉(zhuǎn)染了 shGINS1-#3 的 MHCC97H 細(xì)胞株(圖 2a、b)。CCK-8增殖實(shí)驗(yàn)和克隆形成實(shí)驗(yàn)結(jié)果顯示,與對(duì)照組相比,shGINS1-MHCC97H組細(xì)胞增殖活性顯著降低(P值均lt;0.01)(圖 2c、d)。本研究隨后分析了 GINS1敲低對(duì)MHCC97H細(xì)胞遷移和侵襲能力的影響。劃痕實(shí)驗(yàn)結(jié)果表明,shGINS1-#3組細(xì)胞的遷移能力顯著弱于shNC組細(xì)胞(Plt;0.01)(圖2e);Transwell實(shí)驗(yàn)結(jié)果顯示,shGINS1-#3組細(xì)胞在48 h內(nèi)發(fā)生侵襲的細(xì)胞數(shù)量顯著少于shNC組細(xì)胞(Plt;0.01)(圖2f)。這些結(jié)果表明,GINS1的表達(dá)能夠促進(jìn)HCC細(xì)胞增殖、遷移和侵襲。
2.3 GINS1表達(dá)缺失增強(qiáng)奧沙利鉑對(duì)HCC細(xì)胞的凋亡誘導(dǎo)作用 為了探究GINS1的表達(dá)是否與HCC細(xì)胞化療耐藥性的產(chǎn)生相關(guān),本研究使用不同濃度梯度奧沙利鉑處理MHCC97H細(xì)胞。如圖3a所示,shGINS1-#3組細(xì)胞對(duì)奧沙利鉑敏感性增強(qiáng),IC 50 值低于 shNC 細(xì)胞(Plt;0.01)。隨后,以35 μmol/L奧沙利鉑分別培養(yǎng)上述兩組細(xì)胞24 h。Western Blot檢測(cè)結(jié)果顯示,與shNC組相比,shGINS1-#3組細(xì)胞內(nèi)凋亡蛋白Bax和Bad表達(dá)水平顯著升高(P值均lt;0.001)(圖3b)。這說明,GINS1的表達(dá)有利于HCC細(xì)胞對(duì)奧沙利鉑化療藥物抗性的形成,GINS1表達(dá)缺失增強(qiáng)了奧沙利鉑對(duì)HCC細(xì)胞的凋亡誘導(dǎo)作用。
2.4 GINS1敲低抑制了HCC腫瘤體內(nèi)生長 為了驗(yàn)證體外實(shí)驗(yàn)的結(jié)果,本研究構(gòu)建了裸鼠HCC負(fù)瘤模型。在接種腫瘤細(xì)胞第21天處死動(dòng)物,完整取下腫瘤,觀察到GINS1敲低組裸鼠形成的腫瘤質(zhì)量和體積均顯著小于對(duì)照組(P值均lt;0.001)(圖4)。
2.5 GINS1通過Notch/JAK2/STAT3通路促進(jìn)HCC進(jìn)展和奧沙利鉑耐藥 Notch通路和JAK/STAT通路的異常表達(dá)與HCC的進(jìn)展密切相關(guān)。與shNC組細(xì)胞相比,在shGINS1-#3組細(xì)胞內(nèi),Notch通路中的Notch1、Notch3受體蛋白表達(dá)水平顯著降低(P值均lt;0.001),JAK/STAT通路中的JAK2、STAT3蛋白總體表達(dá)水平無顯著差異,而p-JAK2和p-STAT3蛋白表達(dá)水平顯著降低(P值均lt;0.001)(圖 5a)。加入 10 μg/mL 的 Notch 受體激動(dòng)劑 Jagged-1后,發(fā)現(xiàn)shGINS1-MHCC97H組細(xì)胞p-JAK2、p-STAT3表達(dá)水平均有所升高(P 值均lt;0.01)(圖 5b),提示 JAK2/STAT3通路受到Notch信號(hào)的直接調(diào)控。此外,克隆形成實(shí)驗(yàn)結(jié)果顯示,10 μg/mL的Jagged-1有效增強(qiáng)了shGINS1-MHCC97H組細(xì)胞增殖活性(Plt;0.01)(圖5c)。而劃痕實(shí)驗(yàn)和 Transwell 侵襲實(shí)驗(yàn)進(jìn)一步發(fā)現(xiàn),在 Jagged-1 作用下,因GINS1敲低而導(dǎo)致的MHCC97H細(xì)胞遷移和侵襲活性抑制得到部分逆轉(zhuǎn)(P值均lt;0.01)(圖5d、e)。最后,以35 μmol/L奧沙利鉑培養(yǎng)細(xì)胞24 h,Western Blot檢測(cè)發(fā)現(xiàn)添加Jagged-1的shGINS1-MHCC97H組細(xì)胞凋亡蛋白 Bax 和 Bad 表達(dá)水平均顯著低于未添加 Jagged-1 的shGINS1-MHCC97H組細(xì)胞(P值均lt;0.01)(圖5f)。以上結(jié)果表明,GINS1對(duì)HCC增殖、遷移、侵襲和耐藥性形成的作用是通過Notch/JAK2/STAT3通路介導(dǎo)的。
3 討論
HCC發(fā)病率和病死率均較高,通常預(yù)后不良,全球每年有近60萬人死于該?。?2-13]。因此,尋找HCC早期診斷和靶向治療的特異性標(biāo)志物有重要意義。
GINS1是四聚體復(fù)合物GINS的一部分,其編碼基因在進(jìn)化上非常保守。它是復(fù)制解旋酶機(jī)制的組成部分,并被認(rèn)為參與DNA復(fù)制。研究發(fā)現(xiàn),GINS1的缺失導(dǎo)致內(nèi)細(xì)胞團(tuán)增殖受損,造成胚胎死亡[14]。多項(xiàng)研究表明,GINS1不僅在正常細(xì)胞中有重要作用,而且在癌細(xì)胞中也發(fā)揮作用。GINS復(fù)合物在乳腺癌、肺癌等腫瘤中被發(fā)現(xiàn)高表達(dá),并且GINS1高轉(zhuǎn)錄活性與腫瘤細(xì)胞高增殖和轉(zhuǎn)移活性、高轉(zhuǎn)移潛能相關(guān)[15]。在本研究中,GINS1被證明在HCC患者、HCC組織和HCC細(xì)胞系中高表達(dá),并且促進(jìn)HCC進(jìn)展和化療耐藥。
研究發(fā)現(xiàn),Notch信號(hào)通路在進(jìn)化上較為保守,Notch受體(Notch1~4)經(jīng)過三次剪切后轉(zhuǎn)位至細(xì)胞核內(nèi)調(diào)控靶基因的轉(zhuǎn)錄[16]。Notch信號(hào)深度參與多種組織器官的發(fā)育和穩(wěn)態(tài),異常表達(dá)可導(dǎo)致癌性和非癌性疾病的發(fā)生。但近期研究表明,Notch信號(hào)的結(jié)果是多變的,且高度依賴于環(huán)境。Notch信號(hào)既可以促進(jìn)多種癌癥的發(fā)展,也可以抑制腫瘤進(jìn)展[17]。據(jù)報(bào)道,Notch1的表達(dá)上調(diào)可以增強(qiáng)HCC細(xì)胞干性和化療耐藥性[18]。另外,Notch1受體的激活可以引發(fā)肝祖細(xì)胞驅(qū)動(dòng)的HCC發(fā)生和腫瘤肺轉(zhuǎn)移[19]。Notch3受體在HCC中表達(dá)異常,并且與患者低生存率相關(guān)[20-21]。多項(xiàng)研究發(fā)現(xiàn),抑制Notch3可以有效增強(qiáng)化療藥物對(duì) HCC 的治療作用[22-24]。因此,Notch1 和 Notch3受體在HCC中均展現(xiàn)出重要作用。
JAK 非受體酪氨酸激酶家族由四種蛋白質(zhì)組成:JAK1、JAK2、JAK3和 TYK2。STAT蛋白是 JAK下游信號(hào)分子。STAT家族成員包括STAT1、STAT2、STAT3、STAT4、STAT5A、STAT5B和STAT6。JAK/STAT信號(hào)通路是細(xì)胞內(nèi)普遍表達(dá)的信號(hào)轉(zhuǎn)導(dǎo)通路,參與細(xì)胞增殖、分化、凋亡、免疫調(diào)節(jié)等許多重要的生物學(xué)過程[25-26]。多項(xiàng)研究表明,JAK/STAT信號(hào)通路與HCC發(fā)生和發(fā)展相關(guān)。使用干擾素-α(IFN-α)來驅(qū)動(dòng)宿主的抗病毒反應(yīng)是目前治療慢性乙型肝炎的一線方法,并已被證實(shí)可以減緩肝纖維化的進(jìn)展,甚至延緩HCC的發(fā)生。作為一種重要的免疫相關(guān)細(xì)胞因子,IFN-α可激活JAK/STAT信號(hào),誘導(dǎo)各種具有抗病毒和免疫調(diào)節(jié)功能的IFN刺激基因[27]。此外,由外來信號(hào)引起的HCC細(xì)胞內(nèi)JAK2/STAT3通路的激活能夠促進(jìn)HCC進(jìn)展[28]。在本研究中,GINS1的表達(dá)被證明能夠通過激活JAK2/STAT3通路促進(jìn)HCC進(jìn)展和化療耐藥。
總之,本研究進(jìn)一步證實(shí)了GINS1在HCC中的關(guān)鍵作用,并且與Notch/JAK2/STAT3通路的激活相關(guān)。這些發(fā)現(xiàn)可能為臨床HCC診治提供新的思路。然而,由于本研究缺少關(guān)于GINS1過表達(dá)的相關(guān)實(shí)驗(yàn),因此GINS1在HCC發(fā)生和發(fā)展中的作用還需要進(jìn)一步探索。
倫理學(xué)聲明:本研究中臨床樣本相關(guān)研究由新疆醫(yī)科大學(xué)第一附屬醫(yī)院倫理委員會(huì)審批,批號(hào):K202311-01,患者均簽署知情同意書。本研究中所有動(dòng)物操作均經(jīng)醫(yī)院動(dòng)物保護(hù)和使用倫理委員會(huì)批準(zhǔn),批號(hào):A240301-236。
利益沖突聲明:本文不存在任何利益沖突。
作者貢獻(xiàn)聲明:霍怡杉負(fù)責(zé)設(shè)計(jì)論文框架,起草論文;段相冰、馬玉玉負(fù)責(zé)實(shí)驗(yàn)操作,研究過程的實(shí)施;李大偉、張凱楠負(fù)責(zé)樣本數(shù)據(jù)收集,統(tǒng)計(jì)學(xué)分析和繪制圖表;張國軍負(fù)責(zé)論文修改;馬秀敏負(fù)責(zé)擬定寫作思路,指導(dǎo)撰寫文章并最后定稿。
參考文獻(xiàn):
[1] WANG Y, DENG BC. Hepatocellular carcinoma: Molecular mecha?nism, targeted therapy, and biomarkers[J]. Cancer Metastasis Rev,2023, 42(3): 629-652. DOI: 10.1007/s10555-023-10084-4.
[2] National Health Commission of the People’s Republic of China.Standard for diagnosis and treatment of primary liver cancer (2024edition)[J]. J Clin Hepatol, 2024, 40(5): 893-918. DOI: 10.12449/JCH240508.中華人民共和國國家衛(wèi)生健康委員會(huì). 原發(fā)性肝癌診療指南(2024年版)[J]. 臨床肝膽病雜志, 2024, 40(5): 893-918. DOI: 10.12449/JCH240508.
[3] WANG WY, WEI C. Advances in the early diagnosis of hepatocellu?lar carcinoma[J]. Genes Dis, 2020, 7(3): 308-319. DOI: 10.1016/j.gendis.2020.01.014.
[4] YE Y, SONG YN, HE SF, et al. GINS2 promotes cell proliferation and inhibits cell apoptosis in thyroid cancer by regulating CITED2 and LOXL2[J]. Cancer Gene Ther, 2019, 26(3-4): 103-113. DOI: 10.1038/s41417-018-0045-y.
[5] SEKEDAT MD, FENY? D, ROGERS RS, et al. GINS motion reveals rep?lication fork progression is remarkably uniform throughout the yeast ge?nome[J]. Mol Syst Biol, 2010, 6: 353. DOI: 10.1038/msb.2010.8.
[6] OGINO H, ISHINO S, MAYANAGI K, et al. The GINS complex from the thermophilic archaeon, Thermoplasma acidophilum may func?tion as a homotetramer in DNA replication[J]. Extremophiles, 2011,15(4): 529-539. DOI: 10.1007/s00792-011-0383-2.
[7] YANG H, LIU XC, ZHU XL, et al. GINS1 promotes the proliferation and migration of glioma cells through USP15-mediated deubiquitina?tion of TOP2A[J]. iScience, 2022, 25(9): 104952. DOI: 10.1016/j.isci.2022.104952.
[8] BAXLEY RM, LEUNG W, SCHMIT MM, et al. Bi-allelic MCM10 vari?ants associated with immune dysfunction and cardiomyopathy cause telomere shortening[J]. Nat Commun, 2021, 12(1): 1626. DOI: 10.1038/s41467-021-21878-x.
[9] XU XL, CHEN E, MO LH, et al. BRCA1 represses DNA replication ini?tiation through antagonizing estrogen signaling and maintains ge?nome stability in parallel with WEE1-MCM2 signaling during preg?nancy[J]. Hum Mol Genet, 2019, 28(5): 842-857. DOI: 10.1093/hmg/ddy398.
[10] AHMAD M, HAMEED Y, KHAN M, et al. Up-regulation of GINS1highlighted a good diagnostic and prognostic potential of survival in three different subtypes of human cancer[J]. Braz J Biol, 2021, 84:e250575. DOI: 10.1590/1519-6984.250575.
[11] FU QQ, ZHENG H, WANG X, et al. GINS1 promotes the initiation and progression of bladder cancer by activating the AKT/mTOR/c-Myc signaling pathway[J]. Exp Cell Res, 2024, 440(1): 114125. DOI:10.1016/j.yexcr.2024.114125.
[12] WANG KC, WANG MD, YANG T. Key points in AASLD practice guid?ance on prevention, diagnosis, and treatment of hepatocellular car?cinoma(2023)[J]. J Clin Hepatol, 2023, 39(9): 2081-2086. DOI: 10.3969/j.issn.1001-5256.2023.09.008.王科淳, 王明達(dá), 楊田. 《2023年美國肝病學(xué)會(huì)實(shí)踐指導(dǎo): 肝細(xì)胞癌的預(yù)防、診斷和治療》意見要點(diǎn)[J]. 臨床肝膽病雜志, 2023, 39(9): 2081-2086. DOI: 10.3969/j.issn.1001-5256.2023.09.008.
[13] LI Z, ZHU JY. Interpretation of guidelines for the diagnosis and treat?ment of primary liver cancer(2024 edition)[J]. J Clin Hepatol, 2024,40(7): 1324-1327. DOI: 10.12449/JCH240707.李照, 朱繼業(yè). 《原發(fā)性肝癌診療指南(2024年版)》解讀[J]. 臨床肝膽病雜志, 2024, 40(7): 1324-1327. DOI: 10.12449/JCH240707.
[14] UENO M, ITOH M, KONG LY, et al. PSF1 is essential for early em?bryogenesis in mice[J]. Mol Cell Biol, 2005, 25(23): 10528-10532.DOI: 10.1128/MCB.25.23.10528-10532.2005.
[15] NAGAHAMA Y, UENO M, MIYAMOTO S, et al. PSF1, a DNA replica?tion factor expressed widely in stem and progenitor cells, drives tu?morigenic and metastatic properties[J]. Cancer Res, 2010, 70(3):1215-1224. DOI: 10.1158/0008-5472.CAN-09-3662.
[16] MAI WH, CHEN CX, LIU QY, et al. Regulation of Notch signaling pathway in immune responses during infection[J]. Chin J Immunol,2024, 40(4): 872-879.麥文豪, 陳楚溪, 劉巧媛, 等. Notch信號(hào)通路在感染過程中對(duì)免疫應(yīng)答的調(diào)控作用[J]. 中國免疫學(xué)雜志, 2024, 40(4): 872-879.
[17] ZHOU BH, LIN WL, LONG YL, et al. Notch signaling pathway: Archi?tecture, disease, and therapeutics[J]. Signal Transduct Target Ther,2022, 7(1): 95. DOI: 10.1038/s41392-022-00934-y.
[18] ZHANG XY, SU TH, WU YF, et al. N6-methyladenosine reader YTHDF1 promotes stemness and therapeutic resistance in hepato?cellular carcinoma by enhancing NOTCH1 expression[J]. Cancer Res, 2024, 84(6): 827-840. DOI: 10.1158/0008-5472.CAN-23-1916.
[19] WU WR, SHI XD, ZHANG FP, et al. Activation of the Notch1-c-myc-VCAM1 signalling axis initiates liver progenitor cell-driven hepato?carcinogenesis and pulmonary metastasis[J]. Oncogene, 2022, 41(16): 2340-2356. DOI: 10.1038/s41388-022-02246-5.
[20] GRAMANTIERI L, GIOVANNINI C, LANZI A, et al. Aberrant Notch3and Notch4 expression in human hepatocellular carcinoma[J]. Liver Int, 2007, 27(7): 997-1007. DOI: 10.1111/j.1478-3231.2007.01544.x.
[21] HU L, XUE F, SHAO MH, et al. Aberrant expression of Notch3 pre?dicts poor survival for hepatocellular carcinomas[J]. Biosci Trends,2013, 7(3): 152-156.
[22] GIOVANNINI C, BAGLIONI M, BARON TOALDO M, et al. Notch3 in?hibition enhances sorafenib cytotoxic efficacy by promoting GSK3b phosphorylation and p21 down-regulation in hepatocellular carci?noma[J]. Oncotarget, 2013, 4(10): 1618-1631. DOI: 10.18632/on?cotarget.1221.
[23] GIOVANNINI C, SALZANO AM, BAGLIONI M, et al. Brivanib in com?bination with Notch3 silencing shows potent activity in tumour mod?els[J]. Br J Cancer, 2019, 120(6): 601-611. DOI: 10.1038/s41416-018-0375-4.
[24] ZHU WH, LIANG Q, YANG X, et al. Combination of sorafenib and Valproic acid synergistically induces cell apoptosis and inhibits he?patocellular carcinoma growth via down-regulating Notch3 and pAkt[J]. Am J Cancer Res, 2017, 7(12): 2503-2514.
[25] XIN P, XU XY, DENG CJ, et al. The role of JAK/STAT signaling path?way and its inhibitors in diseases[J]. Int Immunopharmacol, 2020,80: 106210. DOI: 10.1016/j.intimp.2020.106210.
[26] YI L, LI WD, WANG Y, et al. Effects of aloperine on proliferation,apoptosis and immune escape of colorectal cancer cells by regulat?ing IL-6/JAK1/STAT3 signaling pathway[J]. Chin J Immunol, 2024,40(7): 1436-1440.伊亮, 李偉東, 王有, 等. 苦豆堿調(diào)節(jié)IL-6/JAK1/STAT3信號(hào)通路對(duì)結(jié)直腸癌細(xì)胞增殖、凋亡和免疫逃逸的影響[J]. 中國免疫學(xué)雜志, 2024, 40(7): 1436-1440.
[27] CARAGLIA M, VITALE G, MARRA M, et al. Alpha-interferon and its effects on signalling pathways within cells[J]. Curr Protein Pept Sci,2004, 5(6): 475-485. DOI: 10.2174/1389203043379378.
[28] ZHAO ZW, SONG JJ, TANG BF, et al. CircSOD2 induced epigenetic alteration drives hepatocellular carcinoma progression through acti?vating JAK2/STAT3 signaling pathway[J]. J Exp Clin Cancer Res,2020, 39(1): 259. DOI: 10.1186/s13046-020-01769-7.
收稿日期:2024-07-20;錄用日期:2024-10-08
本文編輯:王瑩
引證本文:HUO YS, LI DW, DUAN XB, et al. Effect of Go-Ichi-Ni-San complex subunit 1 on disease progression and chemotherapy resistance in hepatocellular carcinoma[J]. J Clin Hepatol, 2025, 41(3): 485-492.
霍怡杉, 李大偉, 段相冰, 等 . Go-ichi-ni-san 復(fù)合物亞基 1(GINS1)對(duì)肝細(xì)胞癌進(jìn)展和化療耐藥的影響[J]. 臨床肝膽病雜志, 2025, 41(3): 485-492.