中文English
ISSN 1001-5256 (Print)
ISSN 2097-3497 (Online)
CN 22-1108/R

留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

外泌体在肝内胆管癌中的作用

唐晋元 杨陈凤麟 梁冬乐 罗雨豪

引用本文:
Citation:

外泌体在肝内胆管癌中的作用

DOI: 10.12449/JCH240130
基金项目: 

国家自然科学基金 (81903000);

四川省自然科学基金 (2023NSFSC1846)

利益冲突声明:本文不存在任何利益冲突。
作者贡献声明:唐晋元负责课题设计与论文撰写;杨陈凤麟、梁冬乐负责绘制表格与图片;罗雨豪负责思路设计,文章修改和最后定稿。
详细信息
    通信作者:

    罗雨豪, luoyuhao1992@swmu.edu.cn (ORCID: 0000-0002-7489-5770)

Role of exosomes in intrahepatic cholangiocarcinoma

Research funding: 

National Natural Science Foundation of China (81903000);

Natural Science Foundation of Sichuan Province (2023NSFSC1846)

More Information
  • 摘要: 肝内胆管癌(ICC)是一种特殊类型的肝癌,其早期临床症状不典型,大多数患者初诊时已处于中晚期。由于缺乏有效的分子标志物和治疗手段,ICC患者5年生存率极低。外泌体是一种细胞分泌的囊泡,包含蛋白质、RNA、脂质等,是细胞间通讯的重要载体。近期研究显示外泌体在ICC发生发展过程中扮演重要角色,本文就外泌体在ICC中的诊断、治疗作用及其机制进行综述,并展望外泌体的治疗前景与潜在的临床应用。

     

  • 图  1  ICC的常见基因组改变及其频率

    Figure  1.  Common genomic changes and frequency of ICC

    图  2  ICC的分子发病机制

    注: EVCOX,环加氧酶;CTLA-4,细胞毒性T淋巴细胞抗原4;CXCL,C-X-C基序趋化因子配体;EGF,表皮生长因子;ERK,细胞外信号调节激酶;Hh,刺猬通路;IDH,异柠檬酸脱氢酶;iNOS,诱导氮氧化物合酶;MMP,基质金属蛋白酶;NF-κB,核因子κB;PDGF,血小板衍生生长因子;PD-1,程序性死亡蛋白1;PD-L1,程序性死亡配体1;PGE,前列腺素E;PI3K,磷脂酰肌醇3-激酶;STAT3,信号传导和转录因子3;TAM,肿瘤相关巨噬细胞;TAN,肿瘤相关性中性粒细胞。

    Figure  2.  Molecular pathogenesis of cholangiocarcinoma

    图  3  ICC源性外泌体调节肿瘤进展的机制

    Figure  3.  ICC derived exosomes can regulate tumor progression through a variety of mechanisms

    图  4  外泌体在ICC与CAF细胞通信中的作用

    Figure  4.  Effects of exosomes on the interaction between ICC and CAF cells

    表  1  外泌体与其他传统细胞外囊泡的区别

    Table  1.   Differences between exosomes and other traditional extracellular vesicles

    特征 直径(nm) 密度(g/mL) 形状 组成 标志物 细胞内起源
    外泌体 40~100 1.13~1.19 杯状 胆固醇,鞘磷脂,神经酰胺,脂筏,暴露PPS CD63,CD9,Alix, TSG101 内部隔室 (核内体)
    微泡 100~1 000 不规则形状 暴露PPS 整合素,选择素 和CD40配体 质膜
    核外颗粒体 50~200 双圆形 胆固醇,二酰基甘油,PPS CR1和蛋白水解酶 质膜
    膜颗粒 50~80 1.04~1.07 圆形 CD133 质膜
    外泌体样颗粒 20~50 1.1 不规则形状 无脂筏 TNFR I
    凋亡小泡 50~500 1.16~1.28 异形 组蛋白
    下载: 导出CSV
  • [1] YANG KG, WANG WW, WANG Y, et al. Proteomic analysis of serum and serum exosomes, and their application in intrahepatic cholangiocarcinoma[J]. Chin J Chromatogr, 2021, 39( 11): 1191- 1202. DOI: 10.3724/SP.J.1123.2021.04009.

    杨凯歌, 王薇薇, 王彦, 等. 血清和血清外泌体的蛋白质组分析及其在肝内胆管癌中的应用[J]. 色谱, 2021, 39( 11): 1191- 1202. DOI: 10.3724/SP.J.1123.2021.04009.
    [2] CAO LP, HONG JW, WU J. Potential of extracellular vesicles and exosomes as diagnostic markers for cholangiocarcinoma[J]. Hepatobiliary Surg Nutr, 2022, 11( 3): 436- 438. DOI: 10.21037/hbsn-2022-02.
    [3] ARAI Y, TOTOKI Y, HOSODA F, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma[J]. Hepatology, 2014, 59( 4): 1427- 1434. DOI: 10.1002/hep.26890.
    [4] SIA D, HOSHIDA Y, VILLANUEVA A, et al. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes[J]. Gastroenterology, 2013, 144( 4): 829- 840. DOI: 10.1053/j.gastro.2013.01.001.
    [5] ANDERSEN JB, SPEE B, BLECHACZ BR, et al. Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors[J]. Gastroenterology, 2012, 142( 4): 1021- 1031.e15. DOI: 10.1053/j.gastro.2011.12.005.
    [6] NEPAL C, O’ROURKE CJ, OLIVEIRA DVNP, et al. Genomic perturbations reveal distinct regulatory networks in intrahepatic cholangiocarcinoma[J]. Hepatology, 2018, 68( 3): 949- 963. DOI: 10.1002/hep.29764.
    [7] LIN JZ, SHI JP, GUO HL, et al. Alterations in DNA damage repair genes in primary liver cancer[J]. Clin Cancer Res, 2019, 25( 15): 4701- 4711. DOI: 10.1158/1078-0432.CCR-19-0127.
    [8] MAYNARD H, STADLER ZK, BERGER MF, et al. Germline alterations in patients with biliary tract cancers: A spectrum of significant and previously underappreciated findings[J]. Cancer, 2020, 126( 9): 1995- 2002. DOI: 10.1002/cncr.32740.
    [9] JUSAKUL A, CUTCUTACHE I, YONG CH, et al. Whole-genome and epigenomic landscapes of etiologically distinct subtypes of cholangiocarcinoma[J]. Cancer Discov, 2017, 7( 10): 1116- 1135. DOI: 10.1158/2159-8290.CD-17-0368.
    [10] CHAN-ON W, NAIRISMÄGI ML, ONG CK, et al. Exome sequencing identifies distinct mutational patterns in liver fluke-related and non-infection-related bile duct cancers[J]. Nat Genet, 2013, 45( 12): 1474- 1478. DOI: 10.1038/ng.2806.
    [11] ONG CK, SUBIMERB C, PAIROJKUL C, et al. Exome sequencing of liver fluke-associated cholangiocarcinoma[J]. Nat Genet, 2012, 44( 6): 690- 693. DOI: 10.1038/ng.2273.
    [12] RODRIGUES PM, OLAIZOLA P, PAIVA NA, et al. Pathogenesis of cholangiocarcinoma[J]. Annu Rev Pathol, 2021, 16: 433- 463. DOI: 10.1146/annurev-pathol-030220-020455.
    [13] XING C, LI H, LI RJ, et al. The roles of exosomal immune checkpoint proteins in tumors[J]. Mil Med Res, 2021, 8( 1): 56. DOI: 10.1186/s40779-021-00350-3.
    [14] HELWA I, CAI JW, DREWRY MD, et al. A comparative study of serum exosome isolation using differential ultracentrifugation and three commercial reagents[J]. PLoS One, 2017, 12( 1): e0170628. DOI: 10.1371/journal.pone.0170628.
    [15] LOBB RJ, BECKER M, WEN SW, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma[J]. J Extracell Vesicles, 2015, 4: 27031. DOI: 10.3402/jev.v4.27031.
    [16] HMMIER A, O’BRIEN ME, LYNCH V, et al. Proteomic analysis of bronchoalveolar lavage fluid(BALF) from lung cancer patients using label-free mass spectrometry[J]. BBA Clin, 2017, 7: 97- 104. DOI: 10.1016/j.bbacli.2017.03.001.
    [17] YÁÑEZ-MÓ M, SILJANDER PRM, ANDREU Z, et al. Biological properties of extracellular vesicles and their physiological functions[J]. J Extracell Vesicles, 2015, 4: 27066. DOI: 10.3402/jev.v4.27066.
    [18] GUO J, ZHONG XX, TAN QL, et al. miR-301a-3p induced by endoplasmic reticulum stress mediates the occurrence and transmission of trastuzumab resistance in HER2-positive gastric cancer[J]. Cell Death Dis, 2021, 12( 7): 696. DOI: 10.1038/s41419-021-03991-3.
    [19] ZHOU DJ, GE W, CAO DD. Advances in the study of exosomes in the development and diagnosis of hepatocellular liver cancer[J]. Chin Med Herald, 2023, 20( 10): 42- 44, 54. DOI: 10.20047/j.issn1673-7210.2023.10.08.

    周丁杰, 戈伟, 曹德东. 外泌体在肝细胞肝癌发展和诊断中的研究进展[J]. 中国医药导报, 2023, 20( 10): 42- 44, 54. DOI: 10.20047/j.issn1673-7210.2023.10.08.
    [20] WEERAPHAN C, PHONGDARA A, CHAIYAWAT P, et al. Phosphoproteome profiling of isogenic cancer cell-derived exosome reveals HSP90 as a potential marker for human cholangiocarcinoma[J]. Proteomics, 2019, 19( 12): e1800159. DOI: 10.1002/pmic.201800159.
    [21] IKEDA C, HAGA H, MAKINO N, et al. Utility of Claudin-3 in extracellular vesicles from human bile as biomarkers of cholangiocarcinoma[J]. Sci Rep, 2021, 11( 1): 1195. DOI: 10.1038/s41598-021-81023-y.
    [22] ARBELAIZ A, AZKARGORTA M, KRAWCZYK M, et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma[J]. Hepatology, 2017, 66( 4): 1125- 1143. DOI: 10.1002/hep.29291.
    [23] PAN Y, SHAO SJ, SUN H, et al. Bile-derived exosome noncoding RNAs as potential diagnostic and prognostic biomarkers for cholangiocarcinoma[J]. Front Oncol, 2022, 12: 985089. DOI: 10.3389/fonc.2022.985089.
    [24] HAN JY, AHN KS, KIM YH, et al. Circulating microRNAs as biomarkers in bile-derived exosomes of cholangiocarcinoma[J]. Ann Surg Treat Res, 2021, 101( 3): 140- 150. DOI: 10.4174/astr.2021.101.3.140.
    [25] GAO LL, YANG XP, ZHANG H, et al. Inhibition of miR-10a-5p suppresses cholangiocarcinoma cell growth through downregulation of Akt pathway[J]. Onco Targets Ther, 2018, 11: 6981- 6994. DOI: 10.2147/OTT.S182225.
    [26] LI O, JIANG B, YI WM, et al. LncRNA NEAT1 promotes cell proliferation, migration, and invasion via the miR-186-5p/PTP4A1 axis in cholangiocarcinoma[J]. Kaohsiung J Med Sci, 2021, 37( 5): 379- 391. DOI: 10.1002/kjm2.12354.
    [27] SUN ZP, TAN ZG, PENG C, et al. LncRNA SNHG3 facilitates the malignant phenotype of cholangiocarcinoma cells via the miR-3173-5p/ERG axis[J]. J Gastrointest Surg, 2022, 26( 4): 802- 812. DOI: 10.1007/s11605-021-05160-5.
    [28] CHEN Q, WANG HB, LI Z, et al. Circular RNA ACTN4 promotes intrahepatic cholangiocarcinoma progression by recruiting YBX1 to initiate FZD7 transcription[J]. J Hepatol, 2022, 76( 1): 135- 147. DOI: 10.1016/j.jhep.2021.08.027.
    [29] CHEN HW, CHENGALVALA V, HU HX, et al. Tumor-derived exosomes: Nanovesicles made by cancer cells to promote cancer metastasis[J]. Acta Pharm Sin B, 2021, 11( 8): 2136- 2149. DOI: 10.1016/j.apsb.2021.04.012.
    [30] LIU JY, REN LW, LI S, et al. The biology, function, and applications of exosomes in cancer[J]. Acta Pharm Sin B, 2021, 11( 9): 2783- 2797. DOI: 10.1016/j.apsb.2021.01.001.
    [31] LI L, ZHAO J, ZHANG QB, et al. Cancer cell-derived exosomes promote HCC tumorigenesis through hedgehog pathway[J]. Front Oncol, 2021, 11: 756205. DOI: 10.3389/fonc.2021.756205.
    [32] WANG YZ, YI J, CHEN XG, et al. The regulation of cancer cell migration by lung cancer cell-derived exosomes through TGF-β and IL-10[J]. Oncol Lett, 2016, 11( 2): 1527- 1530. DOI: 10.3892/ol.2015.4044.
    [33] LI XX, WANG SH, ZHU RJ, et al. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway[J]. J Hematol Oncol, 2016, 9: 42. DOI: 10.1186/s13045-016-0269-y.
    [34] LIN LY, DU LM, CAO K, et al. Tumour cell-derived exosomes endow mesenchymal stromal cells with tumour-promotion capabilities[J]. Oncogene, 2016, 35( 46): 6038- 6042. DOI: 10.1038/onc.2016.131.
    [35] BECKER A, THAKUR BK, WEISS JM, et al. Extracellular vesicles in cancer: Cell-to-cell mediators of metastasis[J]. Cancer Cell, 2016, 30( 6): 836- 848. DOI: 10.1016/j.ccell.2016.10.009.
    [36] HE LQ, ZHU W, CHEN Q, et al. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis[J]. Theranostics, 2019, 9( 26): 8206- 8220. DOI: 10.7150/thno.37455.
    [37] TANG MKS, YUE PYK, IP PP, et al. Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface[J]. Nat Commun, 2018, 9( 1): 2270. DOI: 10.1038/s41467-018-04695-7.
    [38] PARK JE, DUTTA B, TSE SW, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift[J]. Oncogene, 2019, 38( 26): 5158- 5173. DOI: 10.1038/s41388-019-0782-x.
    [39] ZHANG PF, GAO C, HUANG XY, et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma[J]. Mol Cancer, 2020, 19( 1): 110. DOI: 10.1186/s12943-020-01222-5.
    [40] XIE FT, XU MX, LU J, et al. The role of exosomal PD-L1 in tumor progression and immunotherapy[J]. Mol Cancer, 2019, 18( 1): 146. DOI: 10.1186/s12943-019-1074-3.
    [41] YIN Z, YU M, MA TT, et al. Mechanisms underlying low-clinical responses to PD-1/PD-L1 blocking antibodies in immunotherapy of cancer: A key role of exosomal PD-L1[J]. J Immunother Cancer, 2021, 9( 1): e001698. DOI: 10.1136/jitc-2020-001698.
    [42] MOON B, CHANG S. Exosome as a delivery vehicle for cancer therapy[J]. Cells, 2022, 11( 3): 316. DOI: 10.3390/cells11030316.
    [43] WALKER S, BUSATTO S, PHAM A, et al. Extracellular vesicle-based drug delivery systems for cancer treatment[J]. Theranostics, 2019, 9( 26): 8001- 8017. DOI: 10.7150/thno.37097.
    [44] LI L, PIONTEK K, ISHIDA M, et al. Extracellular vesicles carry microRNA-195 to intrahepatic cholangiocarcinoma and improve survival in a rat model[J]. Hepatology, 2017, 65( 2): 501- 514. DOI: 10.1002/hep.28735.
    [45] SHA M, JEONG S, QIU BJ, et al. Isolation of cancer-associated fibroblasts and its promotion to the progression of intrahepatic cholangiocarcinoma[J]. Cancer Med, 2018, 7( 9): 4665- 4677. DOI: 10.1002/cam4.1704.
    [46] SIRICA AE, CAMPBELL DJ, DUMUR CI. Cancer-associated fibroblasts in intrahepatic cholangiocarcinoma[J]. Curr Opin Gastroenterol, 2011, 27( 3): 276- 284. DOI: 10.1097/MOG.0b013e32834405c3.
    [47] FABRIS L, SATO K, ALPINI G, et al. The tumor microenvironment in cholangiocarcinoma progression[J]. Hepatology, 2021, 73( Suppl 1): 75- 85. DOI: 10.1002/hep.31410.
    [48] YANG RJ, WANG D, HAN S, et al. Erratum: miR-206 suppresses the deterioration of intrahepatic cholangiocarcinoma and promotes sensitivity to chemotherapy by inhibiting interactions with stromal CAF: Erratum[J]. Int J Biol Sci, 2022, 18( 11): 4466- 4467. DOI: 10.7150/ijbs.75760.
    [49] OTA Y, TAKAHASHI K, OTAKE S, et al. Extracellular vesicle-encapsulated miR-30e suppresses cholangiocarcinoma cell invasion and migration via inhibiting epithelial-mesenchymal transition[J]. Oncotarget, 2018, 9( 23): 16400- 16417. DOI: 10.18632/oncotarget.24711.
  • 加载中
图(4) / 表(1)
计量
  • 文章访问数:  134
  • HTML全文浏览量:  35
  • PDF下载量:  25
  • 被引次数: 0
出版历程
  • 收稿日期:  2023-04-18
  • 录用日期:  2023-06-15
  • 出版日期:  2024-01-23
  • 分享
  • 用微信扫码二维码

    分享至好友和朋友圈

目录

    /

    返回文章
    返回