上海交通大学学报(医学版), 2023, 43(9): 1107-1114 doi: 10.3969/j.issn.1674-8115.2023.09.005

论著 · 基础研究

ASGR1在肝细胞癌中的意义及机制研究

李倩玉,1, 郭文韵1, 钱逸斐1, 李松玲2, 朱子俊1, 刘艳丰,1,3

1.上海交通大学医学院附属仁济医院临床干细胞研究中心,上海 200127

2.上海交通大学生物医学工程学院Med-X研究院,上海 200030

3.上海交通大学医学院附属仁济医院肝外科,上海 200127

Study on the significance and mechanism of ASGR1 in hepatocellular carcinoma

LI Qianyu,1, GUO Wenyun1, QIAN Yifei1, LI Songling2, ZHU Zijun1, LIU Yanfeng,1,3

1.Renji-Med X Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China

2.School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China

3.Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China

通讯作者: 刘艳丰,电子信箱:lyf7858188@163.com

编委: 徐敏

收稿日期: 2023-08-16   接受日期: 2023-09-18   网络出版日期: 2023-09-28

基金资助: 国家自然科学基金.  82073190
上海交通大学医学院“双百人”项目.  20221704

Corresponding authors: LIU Yanfeng, E-mail:lyf7858188@163.com.

Received: 2023-08-16   Accepted: 2023-09-18   Online: 2023-09-28

作者简介 About authors

李倩玉(1997—),女,硕士生;电子信箱:qianyu_lli@163.com。 E-mail:qianyu_lli@163.com

摘要

目的·探究去唾液酸糖蛋白受体1(asialoglycoprotein receptor 1,ASGR1)在肝细胞癌(hepatocellular carcinoma,HCC)中的意义及潜在机制。方法·通过R语言分析癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库中ASGR1在肝癌患者中的表达情况并绘制相关生存曲线。利用人类蛋白质图谱(The Human Protein Atlas,HPA)数据库获得人体正常肝组织和肝癌组织的免疫组织化学(immunohistochemistry,IHC)数据来分析ASGR1的蛋白表达情况。利用流体动力学尾静脉注射(hydrodynamic tail vein injection,HTVI)递送方法,在免疫完全的小鼠肝脏中敲除Asgr1探究其在体内的致瘤功能,并通过蛋白免疫印迹法(Western blotting,WB)验证基因敲除效率。利用R语言进行京都基因与基因组百科全书(Kyoto Encyclopedia of Genes and Genomes,KEGG)通路富集分析及相关性分析,利用基因探针富集(Gene Set Enrichment Analysis,GSEA)软件进行GSEA hallmark相关通路分析,利用实时荧光定量PCR(quantitative real-time PCR,qPCR)在小鼠肝癌组织中验证糖酵解关键基因表达水平。结果·ASGR1在肝癌组织中显著低表达,在肝癌患者中ASGR1的低表达与患者较差的总体生存期(overall survival,OS)、无疾病间隔(disease free interval,DFI)、无进展间隔期(progression free interval,PFI)和疾病特异性生存期(disease specific survival,DSS)相关;肿瘤分级程度越高的肝癌患者ASGR1基因表达水平越低。人体正常肝组织ASGR1蛋白的表达显著高于肝癌组织。在免疫完全的肝细胞癌小鼠模型中,小鼠内源性Asgr1敲除可增加肝组织中肿瘤结节的大小和数量。TCGA数据库中ASGR1低表达组肝癌患者富集到多条癌症及代谢相关通路,ASGR1表达与部分糖酵解关键基因表达呈负相关,Asgr1敲除组的小鼠肝癌组织中糖酵解水平高于对照组,提示ASGR1低表达很可能促进肝癌的生长发展,加强代谢重编程促进肿瘤的合成代谢发展。结论·ASGR1在肝癌患者中表达显著降低,与患者的预后呈正相关;小鼠体内敲除Asgr1可促进肝细胞癌的发生发展;ASGR1可以作为肝癌预后不良的潜在生物标志物和潜在治疗新靶点。

关键词: 去唾液酸糖蛋白受体1(ASGR1 ; 肝细胞癌 ; 流体动力学尾静脉注射 ; 治疗靶点

Abstract

Objective ·To explore the significance and mechanism of asialoglycoprotein receptor 1 (ASGR1) in hepatocellular carcinoma. Methods ·The expression of ASGR1 in patients with liver cancer in The Cancer Genome Atlas (TCGA) database was analyzed by R language and the related survival curves were drawn. The Human Protein Atlas (HPA) database was used to obtain the immunohistochemistry (IHC) data of normal human liver tissue and liver cancer tissue to analyze the protein expression of ASGR1. By using the hydrodynamic tail vein injection (HTVI) delivery method, Asgr1 was knocked out in the liver of fully immune mice to explore its tumorigenic function invivo. Gene knockout efficiency was verified by Western blotting (WB). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and correlation analysis were performed by using R language. The GSEA hallmark correlation pathway analysis was performed by using Gene Set Enrichment Analysis (GSEA) software. The expression level of key genes of glycolysis in mouse liver cancer tissue was verified by quantitative real-time PCR (qPCR). Results ·ASGR1 was significantly low-expressed in liver cancer tissue, and the low expression of ASGR1 in liver cancer patients was associated with poorer overall survival (OS), disease-free interval (DFI), progression-free interval (PFI), and disease-specific survival (DSS). The higher the degree of tumor grade, the lower the expression level of ASGR1 in patients with liver cancer. Immunohistochemistry showed that the protein expression of ASGR1 in normal human liver tissue was significantly higher than that in liver cancer tissue. In an immunocompetent mouse model of hepatocellular carcinoma, knockout of endogenous Asgr1 in mice increased the size and number of tumor nodules in liver tissue. In the TCGA database, patients with liver cancer in the ASGR1 low-expression group were enriched in multiple cancer and metabolic pathways. The expression of ASGR1 was negatively correlated with some key genes of glycolysis. The level of glycolysis in liver cancer tissues of mice in the Asgr1 knockout group was higher than that in the control group. It was suggested that the low expression of ASGR1 be likely to promote the growth and development of liver cancer and strengthen metabolic reprogramming to promote the anabolic development of tumors. Conclusion ·The expression of ASGR1 is significantly reduced in patients with liver cancer, which is positively correlated with the prognosis of patients. Knocking out Asgr1 in mice can promote the occurrence and development of hepatocellular carcinoma. ASGR1 can be used as a potential biomarker for poor prognosis of liver cancer and a new target for potential treatment.

Keywords: asialoglycoprotein receptor 1 (ASGR1) ; hepatocellular carcinoma ; hydrodynamic tail vein injection ; therapeutic target

PDF (6113KB) 元数据 多维度评价 相关文章 导出 EndNote| Ris| Bibtex  收藏本文

本文引用格式

李倩玉, 郭文韵, 钱逸斐, 李松玲, 朱子俊, 刘艳丰. ASGR1在肝细胞癌中的意义及机制研究. 上海交通大学学报(医学版)[J], 2023, 43(9): 1107-1114 doi:10.3969/j.issn.1674-8115.2023.09.005

LI Qianyu, GUO Wenyun, QIAN Yifei, LI Songling, ZHU Zijun, LIU Yanfeng. Study on the significance and mechanism of ASGR1 in hepatocellular carcinoma. Journal of Shanghai Jiao Tong University (Medical Science)[J], 2023, 43(9): 1107-1114 doi:10.3969/j.issn.1674-8115.2023.09.005

肝癌(liver cancer)是世界上最常见的癌症之一,原发性肝癌(primary liver cancer)在全球癌症死因中位于第3位,严重威胁着人类健康1-2。原发性肝癌有多种类型,其中肝细胞癌(hepatocellular carcinoma,HCC)占全球肝癌病例的75%~85%,是最常见的一种肝癌类型。肝细胞癌的发生发展是一个多步骤的病理过程,发病的危险因素也多种多样,如乙型肝炎病毒和丙型肝炎病毒的慢性感染、黄曲霉毒素的感染、长期饮酒、2型糖尿病等3-4。虽然目前对肝细胞癌的研究已取得一定进展,但是其发生和发展机制尚未清晰,了解肝细胞癌的潜在分子机制对于未来的治疗至关重要。

去唾液酸糖蛋白受体1(asialoglycoprotein receptor 1,ASGR1)是一种C型凝集素,主要表达在肝脏,能特异性识别末端含有非还原半乳糖残基或N-乙酰半乳糖胺残基的糖蛋白或糖脂,介导多种唾液酸化糖蛋白的内吞作用和溶酶体降解5-7。然而,ASGR1在肝细胞癌中的功能和分子机制仍不十分清晰。本研究主要通过生物信息学分析和小鼠体内试验,探究ASGR1在肝细胞癌中的意义及潜在机制。

1 材料与方法

1.1 数据收集

通过UCSC Xena(xenabrowser.net)网站获取癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库的基因表达矩阵和患者临床数据。人体正常肝组织及肝癌组织相关的免疫组织化学(immunohistochemistry,IHC)数据来自人类蛋白质图谱(The Human Protein Atlas,HPA)数据库(https://www.proteinatlas.org/)。

1.2 生物信息学分析

使用R语言对基因表达矩阵进行癌和癌旁组织的表达量分析及肝癌患者不同基因表达之间的相关性分析。使用R语言survival包和survminer包进行生存分析。使用R语言limma包对基因表达矩阵按照ASGR1的中位值分为高低2组后进行差异基因分析,通过P值及错误发现率(false discovery rate,FDR)筛选差异基因,使用R语言ClusterProfiler包进行京都基因与基因组百科全书(Kyoto Encyclopedia of Genes and Genomes,KEGG)通路富集分析,使用基因探针富集(Gene Set Enrichment Analysis,GSEA)软件进行基因集富集分析。

1.3 流体动力学尾静脉注射

使用CRISPR-Cas9方法构建小鼠Asgr1-sgRNA质粒。将6周大的雄性C57BL/6小鼠随机分组,每组5只小鼠,利用流体动力学尾静脉注射(hydrodynamic tail vein injection,HTVI)方法,每只小鼠注射2 mL溶于生理盐水的质粒混合物。每2 mL质粒混合物包含12 μg pT3-EF1A-MYC-IRES-luciferase、10 μg pX330-sg-p53和6.5 μg CMV-SB13。试验组或对照组再分别加入12 μg lenti-CRISPR sgAsgr1或12 μg空载体lenti-CRISPR v2。在5~7 s内将溶于生理盐水的质粒混合物注射到小鼠的尾静脉中。通过腹部触诊监测小鼠,并在它们具有高肝肿瘤负荷时安乐死,取肝脏肿瘤进行分析。构建Asgr1-sgRNA质粒的引物序列见表1

表1   sgRNA序列(5′→3′)

Tab 1  Sequence for sgRNA (5′→3′)

sgRNAForward (5′→3′)Reverse (5′→3′)
Asgr1-sgRNA-1CACCGCGCTGCAAGAGCCGTGGAGTAAACACTCCACGGCTCTTGCAGCGC
Asgr1-sgRNA-2CACCGGACCATCATCAACTCCGGAGAAACCTCCGGAGTTGATGATGGTCC

新窗口打开| 下载CSV


1.4 蛋白免疫印迹法

液氮研磨小鼠肿瘤组织后,使用RIPA蛋白裂解液冰上裂解30 min,离心获得蛋白上清液,用酶标仪进行蛋白定量,后与蛋白上样缓冲液一起煮沸,获得蛋白样品。取适量蛋白样品上样后进行蛋白凝胶电泳,后将蛋白质转膜到PVDF膜上。用5%脱脂牛奶室温封闭1 h,洗膜后一抗4 ℃孵育过夜。次日洗膜后室温下孵育二抗1 h,洗膜后在凝胶成像仪中成像并记录。

抗体信息:ASGR1抗体(PTM BIO,PTM-6093),ACTB抗体(ABclonal,AC028)。

1.5 总RNA提取及实时荧光定量PCR

液氮研磨小鼠肿瘤组织后,加入适量TRIzol进行裂解,后加入体积为TRIzol体积1/5的三氯甲烷颠倒混匀。12 000×g低温离心15 min后收集上清液,加入等体积异丙醇后混匀。继续12 000×g低温离心10 min,弃上清液并加入75%乙醇。12 000×g低温离心5 min后弃上清液。移除乙醇并自然干燥RNA沉淀,及时溶解于DEPC水并测定RNA浓度。使用反转录试剂将RNA反转录为cDNA。后在罗氏LightCycler®384系统进行实时荧光定量PCR(quantitative real-time PCR,qPCR)反应。引物序列见表2

表2   PCR引物序列(5′→3′)

Tab 2  Primer sequence for PCR (5′→3′)

PrimerForward (5′→3′)Reverse (5′→3′)
ActbGGCTGTATTCCCCTCCATCGCCAGTTGGTAACAATGCCATGT
Hk2TGATCGCCTGCTTATTCACGGAACCGCCTAGAAATCTCCAGA
Pgk1ATGTCGCTTTCCAACAAGCTGGCTCCATTGTCCAAGCAGAAT
Pkm2GCCGCCTGGACATTGACTCCCATGAGAGAAATTCAGCCGAG

新窗口打开| 下载CSV


1.6 统计学方法

采用R软件进行生物信息学分析。试验数据使用GraphPad Prism 9软件进行分析,组间比较采用t检验,P<0.05表示差异具有统计学意义。

2 结果

2.1 ASGR1 在肝细胞癌中的表达及其与预后的关系

为了探究ASGR1与肝细胞癌发生发展的关系,我们在TCGA肝癌数据库中对临床患者的转录组测序数据进行生物信息学分析,结果显示:相比于癌旁组织,ASGR1在肝癌组织样本中低表达(图1A);随着肝癌进展,分级越高,ASGR1的表达越低(图1B)。根据TCGA数据库肝癌患者的临床信息绘制生存曲线,结果显示ASGR1的表达与肝癌患者的总体生存期(overall survival,OS)、无疾病间隔(disease-free interval,DFI)、无进展间隔期(progression-free interval,PFI)和疾病特异性生存期(disease-specific survival,DSS)均呈明显的正相关关系(图1C~F)。

图1

图1   ASGR1 在肝癌组织样本中的表达及其与预后的关系

Note: A. Expression of ASGR1 in tumor and normal tissues of liver cancer patients from TCGA database. B. Expression of ASGR1 in different stages of liver cancer patients from TCGA database. C‒F. The overall survival (C), disease-free interval (D), progression-free interval (E) and disease-specific survival (F) curves of liver cancer patients with low or high ASGR1 expression from TCGA database.

Fig 1   Expression of ASGR1 in liver cancer samples and its correlation with prognosis


2.2 ASGR1蛋白在肝癌组织中的表达情况

通过HPA数据库,我们获得了人体正常肝组织和肝癌组织相关的IHC数据,以比较两者之间ASGR1的蛋白表达水平。结果显示,ASGR1在肝癌组织中的表达显著低于正常肝组织(图2A、B)。

图2

图2   ASGR1在人体正常肝组织和肝癌组织的蛋白表达水平

Note: A. Immunohistochemical pictures of ASGR1-related protein expression levels in normal human liver tissue and liver cancer tissue from the HPA database. B. The proportion of different degrees of ASGR1 staining in normal human liver tissue and liver cancer tissue.

Fig 2   Protein expression level of ASGR1 in normal human liver tissue and liver cancer tissue


2.3 Asgr1 敲除在小鼠体内对肝细胞癌发生发展的影响

为了进一步探究ASGR1在肝细胞癌发生和发展中的作用,通过HTVI递送方法在免疫完全小鼠中通过注射pT3-EF1A-MYC-IRES-luciferase、pX330-sg-p53、CMV-SB13、lenti-CRISPR sgAsgr1(试验组)或lenti-CRISPR v2(对照组)来探究ASGR1的作用8图3)。

图3

图3   小鼠流体动力学高压尾静脉注射模型

Note: Delivery via HTVI in fully immune mice by injection of pT3-EF1A-MYC-IRES-luciferase, pX330-sg-p53, CMV-SB13, lenti-CRISPR sgAsgr1 (sgAsgr1 group) or lenti-CRISPR v2 (sgCtrl group).

Fig 3   Schematic diagram of mouse hydrodynamic tail vein injection model


小鼠肝癌模型注射后约30 d,我们收获了小鼠肝脏(图4A)。通过对小鼠肝癌组织中ASGR1蛋白水平进行检测,证实该模型实现了内源性Asgr1敲除(图4B)。对每一个肝脏的肿瘤形成情况进行统计分析,结果显示,Asgr1敲除组(sgAsgr1)相比于对照组(sgCtrl),小鼠肝重比有上升的趋势,肝脏所形成的肿瘤个数更多,最大直径更大,较大肿瘤的占比也更大(图4C~F)。

图4

图4   Asgr1 敲除验证及小鼠肝细胞癌的形成情况

Note: A. Representative pictures of hepatocellular carcinoma in sgCtrl and sgAsgr1 group. B. Protein expression of ASGR1 and ACTB in liver tumor tissues of mice in sgCtrl and sgAsgr1 group. C‒F. Liver weight ratio (C), tumor number (D), max size (E) and proportion of tumor numbers (F) with different sizes in sgCtrl and sgAsgr1 group.

Fig 4   Validation of Asgr1 knockout and formation of hepatocellular carcinoma in mice


2.4 ASGR1 相关的生物学功能及富集通路分析

为了分析ASGR1与肝细胞癌发生发展潜在可能的分子机制,我们对TCGA肝癌数据库进行了分析。根据P<0.05及FDR>0.5选取ASGR1低表达情况下上调的前557个基因进行KEGG通路富集分析。其中,富集到的排序靠前的通路包括ECM-受体相互作用通路、细胞周期通路、癌症通路等癌症相关的通路(图5A)。同时,我们还使用GSEA软件对以ASGR1中位值区分的高低2组进行了Hallmark通路的富集分析,结果显示ASGR1低表达组与G2M检查点通路、E2F信号通路、糖酵解通路、PI3K-AKT-MTOR信号通路有关(图5B)。

图5

图5   TCGA肝癌数据库中 ASGR1 表达相关的生物信息学分析及小鼠肝癌组织mRNA表达水平验证

Note: A. KEGG pathway enrichment analysis of genes negatively correlated with ASGR1. B. GSEA analysis of hallmark pathway enrichment between ASGR1 low and high group. C. Correlation analysis of expression of ASGR1 and glycolysis-related genes. D. The mRNA expression levels of glycolysis-related genes in mouse liver cancer tissue.

Fig 5   Bioinformatics analysis of ASGR1 expression in TCGA liver cancer database and verification of mRNA expression levels in mouse liver cancer tissue


根据分析的结果,我们在TCGA肝癌数据库中对糖酵解途径的关键基因与ASGR1进行了相关性分析,结果发现,ASGR1的表达与己糖激酶2(HK2)、磷酸甘油酸激酶1(PGK1)、丙酮酸激酶2(PKM2)均呈明显的负相关(图5C)。为了进一步证实上面的分析结果,我们检测了Asgr1体内敲除组和对照组小鼠肝癌组织的mRNA水平,结果显示,Hk2Pgk1Pkm2Asgr1敲除组上调(图5D)。

3 讨论

肝细胞癌是全球第六大常见癌症,是全球癌症相关死亡的第四大原因9,5年生存率仅为15%,并且肝细胞癌的发病率和死亡率逐年都在增加,预计到2030年,肝细胞癌将成为癌症相关死亡的第三大原因10。由于肝细胞癌早期诊断较为困难且肿瘤细胞容易转移扩散,患者的整体生存率很低11,肿瘤的复发和转移是肝细胞癌患者死亡的主要原因12。尽管随着外科手术切除、化学治疗、介入、靶向及免疫治疗等方式的发展13,肝细胞癌的治疗取得了较大的进步,但其转移复发率仍然很高。因此,深入了解调控肝细胞癌发生发展的分子机制,针对特定的基因或通路进行特定的诊断治疗可能是未来治疗肝细胞癌的有效手段。

ASGR1,也被称为肝细胞半乳糖/N-乙酰基葡糖胺受体,或者Ashwell-Morell受体,主要表达于肝窦状间隙的肝实质细胞表面14,能够介导去唾液酸化的糖蛋白被肝细胞内吞降解15。目前已知的ASGR1在糖蛋白代谢16、调节凝血17、胆固醇代谢18等多个生理过程都发挥着重要的作用19。在肝病中,发现ASGR1在肝硬化组织中上调20。关于ASGR1在肝细胞癌中的作用,有研究21表明,ASGR1可能通过与LASS2相互作用来抑制V-ATPase的活性,从而抑制肝癌细胞的转移潜能。最新的研究22表明,ASGR1通过促进NLKSTAT3的结合、抑制STAT3磷酸化来抑制肝癌的进展。这些研究均提示ASGR1在肝细胞癌发生发展中发挥着重要的作用,然而关于ASGR1在肝细胞癌中的研究仍然很少。

本研究为了探究ASGR1在肝细胞癌中的意义及机制,首先分析TCGA数据库中ASGR1在肝癌患者中的表达情况并做了生存分析,发现ASGR1在肝癌组织中显著低表达,同时肿瘤分级程度越高,ASGR1基因表达水平越低;且ASGR1低表达的患者的OS、DFI、PFI、DSS均缩短。之后,又利用HPA数据库发现人体正常肝组织ASGR1蛋白的表达显著高于肝癌组织。以上结果说明,ASGR1基因很可能是一种肿瘤抑制因子,在肝细胞癌进展中发挥抗肿瘤作用。为了更有效地探究ASGR1在肝细胞癌发生发展中的作用,通过HTVI建立小鼠肝原位模型,在小鼠肝脏中敲除Asgr1,探究其在体内的致瘤的功能,结果表明,小鼠内源性敲除Asgr1可以增加肝组织中肿瘤结节的大小和数量,敲除Asgr1后肝细胞癌体内成瘤能力增强。通过对TCGA中肝癌的数据进行KEGG及GSEA分析,发现ASGR1的低表达激活了癌症相关通路以及细胞周期和细胞增殖等相关通路,提示ASGR1的低表达有助于肝细胞癌的生长和发展;同时,ASGR1低表达组与糖酵解通路、PI3K-AKT-MTOR信号通路有关,相关性分析表明ASGR1与糖酵解通路部分关键基因的表达水平呈显著的负相关。为了进一步证实分析的结果,我们在小鼠肝癌组织中对糖酵解通路部分关键基因在mRNA水平进行验证,验证结果与生物信息学分析一致,Asgr1敲除组小鼠肝癌组织的糖酵解水平高于对照组。肝脏作为代谢器官在维持机体代谢稳态中起着重要作用,在调节各种代谢途径方面具有重要作用23,代谢重编程被认为是肿瘤重要的代谢表型24-25,恶性肿瘤可以通过改变代谢途径来维持生长复制需要的高能量需求26。参与糖酵解、脂肪生成、氨基酸合成和核苷酸生物合成的几种代谢酶得到增强27,以加强代谢重编程过程中肿瘤的合成代谢发展。致癌信号转导通路PI3K-AKT-MTOR通路可增强肿瘤中的Warburg效应,促进癌细胞生长和转移28;该信号激活的癌症已被发现变得更具侵袭性,并且AKT通路激活已被证明是肝癌患者早期复发和预后不良的显著危险因素29-30。这些均提示ASGR1的低表达很可能促使肝细胞癌发生代谢重编程,进而促进肝细胞癌的发生发展。其具体机制还有待进一步探究。

综上所述,本研究主要通过生物信息学分析和小鼠体内尾静脉注射模型探究了ASGR1在肝细胞癌中的意义及机制,提出ASGR1基因具有作为肝细胞癌临床预后指标和治疗靶点的潜力,并初步分析验证了ASGR1低表达促进肝细胞癌发生发展的分子机制及信号通路。本研究可为肝细胞癌发生发展及治疗提供新的思路。

作者贡献声明

李倩玉、郭文韵参与试验设计;李倩玉、钱逸斐负责论文的写作和修改;李松玲、朱子俊参与文献检索;刘艳丰负责试验指导与论文审阅。所有作者均阅读并同意最终稿件的提交。

AUTHOR's CONTRIBUTIONS

The study was designed by LI Qianyu and GUO Wenyun. The manuscript was drafted and revised by LI Qianyu and QIAN Yifei. The document was searched by LI Songling and ZHU Zijun. The research was guided by LIU Yanfeng. All the authors have read the last version of paper and consented for submission.

利益冲突声明

所有作者声明不存在利益冲突。

COMPETING INTERESTS

All authors disclose no relevant conflict of interests.

参考文献

BRAY F, FERLAY J, SOERJOMATARAM I, et al. Global cancer statistics 2018: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2018, 68(6): 394-424.

[本文引用: 1]

SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3): 209-249.

[本文引用: 1]

SINGAL A G, LAMPERTICO P, NAHON P. Epidemiology and surveillance for hepatocellular carcinoma: new trends[J]. J Hepatol, 2020, 72(2): 250-261.

[本文引用: 1]

SINGH M K, DAS B K, CHOUDHARY S, et al. Diabetes and hepatocellular carcinoma: a pathophysiological link and pharmacological management[J]. Biomed Pharmacother, 2018, 106: 991-1002.

[本文引用: 1]

ASHWELL G, HARFORD J. Carbohydrate-specific receptors of the liver[J]. Annu Rev Biochem, 1982, 51: 531-554.

[本文引用: 1]

TANOWITZ M, HETTRICK L, REVENKO A, et al. Asialoglycoprotein receptor 1 mediates productive uptake of N-acetylgalactosamine-conjugated and unconjugated phosphorothioate antisense oligonucleotides into liver hepatocytes[J]. Nucleic Acids Res, 2017, 45(21): 12388-12400.

夏梦蝶, 廖韦, 向琼, 等. 去唾液酸糖蛋白受体1在动脉粥样硬化中的研究进展[J]. 中国动脉硬化杂志, 2022, 30(6): 541-545.

[本文引用: 1]

XIA M D, LIAO W, XIANG Q, et al. Research progress of asialoglycoprotein receptor 1 in atherosclerosis[J]. Chinese Journal of Arteriosclerosis, 2022, 30(6): 541-545.

[本文引用: 1]

DE GALARRETA M R, BRESNAHAN E, MOLINA-SÁNCHEZ P, et al. β-catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma[J]. Cancer Discov, 2019, 9(8): 1124-1141.

[本文引用: 1]

SIEGEL R L, MILLER K D, WAGLE N S, et al. Cancer statistics, 2023[J]. CA A Cancer J Clinicians, 2023, 73(1): 17-48.

[本文引用: 1]

RAHIB L, SMITH B D, AIZENBERG R, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States[J]. Cancer Res, 2014, 74(11): 2913-2921.

[本文引用: 1]

ZHOU J, SUN H C, WANG Z, et al. Guidelines for the diagnosis and treatment of hepatocellular carcinoma (2019 edition)[J]. Liver Cancer, 2020, 9(6): 682-720.

[本文引用: 1]

EL-SERAG H B. Hepatocellular carcinoma[J]. N Engl J Med, 2011, 365(12): 1118-1127.

[本文引用: 1]

LLOVET J M, KELLEY R K, VILLANUEVA A, et al. Hepatocellular carcinoma[J]. Nat Rev Dis Primers, 2021, 7(1): 6.

[本文引用: 1]

Gilbert Ashwell: sweet on science[J]. Nat Med, 2008, 14(6): 608.

[本文引用: 1]

GREWAL P K. The ashwell-morell receptor[J]. Methods Enzymol, 2010, 479: 223-241.

[本文引用: 1]

SUGAHARA K, TOGASHI H, TAKAHASHI K, et al. Separate analysis of asialoglycoprotein receptors in the right and left hepatic lobes using Tc-GSA SPECT[J]. Hepatology, 2003, 38(6): 1401-1409.

[本文引用: 1]

GREWAL P K, UCHIYAMA S, DITTO D, et al. The Ashwell receptor mitigates the lethal coagulopathy of sepsis[J]. Nat Med, 2008, 14(6): 648-655.

[本文引用: 1]

WANG J Q, LI L L, HU A, et al. Inhibition of ASGR1 decreases lipid levels by promoting cholesterol excretion[J]. Nature, 2022, 608(7922): 413-420.

[本文引用: 1]

RIGOPOULOU E I, ROGGENBUCK D, SMYK D S, et al. Asialoglycoprotein receptor (ASGPR) as target autoantigen in liver autoimmunity: lost and found[J]. Autoimmun Rev, 2012, 12(2): 260-269.

[本文引用: 1]

WITZIGMANN D, QUAGLIATA L, SCHENK S H, et al. Variable asialoglycoprotein receptor 1 expression in liver disease: implications for therapeutic intervention[J]. Hepatol Res, 2016, 46(7): 686-696.

[本文引用: 1]

GU D, JIN H, JIN G, et al. The asialoglycoprotein receptor suppresses the metastasis of hepatocellular carcinoma via LASS2-mediated inhibition of V-ATPase activity[J]. Cancer Lett, 2016, 379(1): 107-116.

[本文引用: 1]

ZHU X X, SONG G Y, ZHANG S Y, et al. Asialoglycoprotein receptor 1 functions as a tumor suppressor in liver cancer via inhibition of STAT3[J]. Cancer Res, 2022, 82(21): 3987-4000.

[本文引用: 1]

TONG M, WONG T L, ZHAO H, et al. Loss of tyrosine catabolic enzyme HPD promotes glutamine anaplerosis through mTOR signaling in liver cancer[J]. Cell Rep, 2021, 36(8): 109617.

[本文引用: 1]

FAUBERT B, SOLMONSON A, DEBERARDINIS R J. Metabolic reprogramming and cancer progression[J]. Science, 2020, 368(6487): eaaw5473.

[本文引用: 1]

YEH Y C, HO H L, WU Y C, et al. AKT1 internal tandem duplications and point mutations are the genetic hallmarks of sclerosing pneumocytoma[J]. Mod Pathol, 2020, 33(3): 391-403.

[本文引用: 1]

HÖNIGOVA K, NAVRATIL J, PELTANOVA B, et al. Metabolic tricks of cancer cells[J]. Biochim Biophys Acta BBA Rev Cancer, 2022, 1877(3): 188705.

[本文引用: 1]

CAI J, SUN X H, GUO H, et al. Non-metabolic role of UCK2 links EGFR-AKT pathway activation to metastasis enhancement in hepatocellular carcinoma[J]. Oncogenesis, 2020, 9(12): 103.

[本文引用: 1]

ICARD P, SIMULA L, WU Z R, et al. Why may citrate sodium significantly increase the effectiveness of transarterial chemoembolization in hepatocellular carcinoma?[J]. Drug Resist Updat, 2021, 59: 100790.

[本文引用: 1]

BUONTEMPO F, ERSAHIN T, MISSIROLI S, et al. Inhibition of Akt signaling in hepatoma cells induces apoptotic cell death independent of Akt activation status[J]. Invest New Drugs, 2011, 29(6): 1303-1313.

[本文引用: 1]

TIAN L Y, SMIT D J, JÜCKER M. The role of PI3K/AKT/mTOR signaling in hepatocellular carcinoma metabolism[J]. Int J Mol Sci, 2023, 24(3): 2652.

[本文引用: 1]

/