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

突变型KRAS在胰腺导管腺癌代谢中的作用

陈懿 谢黎 吴健

引用本文:
Citation:

突变型KRAS在胰腺导管腺癌代谢中的作用

DOI: 10.3969/j.issn.1001-5256.2022.12.043
基金项目: 

国家自然科学基金面上项目 (81871997);

国家自然科学基金面上项目 (82170624)

利益冲突声明:所有作者均声明不存在利益冲突。
作者贡献声明:陈懿对本文的思路和撰写有关键贡献;谢黎参与起草或修改文章关键内容;吴健负责文字的修改与定稿。
详细信息
    通信作者:

    吴健, jian.wu@fudan.edu.cn

Role of KRAS mutation in metabolism of pancreatic ductal adenocarcinoma

Research funding: 

National Science Foundation of China (81871997);

National Science Foundation of China (82170624)

More Information
  • 摘要: 胰腺癌作为一种致死率高、预后差的恶性肿瘤,肿瘤代谢相关的基因突变被认为是其进展和预后不良的基础。胰腺导管腺癌(PDAC)是最常见的胰腺癌类型,86%的PDAC患者被检测出KRAS突变,KRAS作为多种信号通路和转录因子上游的“分子开关”,对肿瘤细胞代谢过程起着重要的调控作用。突变型KRAS可影响营养物质和能量代谢重编程、巨胞饮作用和自噬,调节肿瘤微环境中的成分相互作用,维持癌细胞的生存和增殖。目前,临床上对PDAC的KRAS靶向治疗大多停留在试验阶段,未来能否真正应用于治疗仍需进一步临床研究。

     

  • 图  1  KRAS信号通路及其对细胞功能的作用

    注:KRAS在与GTP结合时被激活,启动下游多种信号通路,最终促进细胞存活、增殖、转化、侵袭、胞吞/胞吐功能。KRAS的突变破坏RAS的内在GTP酶活性,使GAP失去对GTP的失活作用,促进GTP向GDP转化,导致KRAS及其下游信号通路的持续激活。GEF:鸟苷酸交换因子;RALGEF:鸟嘌呤核苷酸交换因子;RALA:Ras样蛋白A;RALB:Ras样蛋白B;PLC:磷酯酶C;NK-κB:核因子κB;MEKK1/2/3/4:MAPK激酶1/2/3/4;JNK:c-Jun氨基末端激酶。

    Figure  1.  KRAS signaling pathway and its effects on cell function

    图  2  KRAS突变影响胰腺导管腺癌代谢重编程

    注:葡萄糖在胞液中可裂解为丙酮酸,是葡萄糖无氧氧化和有氧氧化的共同起始途径。在正常细胞中,氧供充足,丙酮酸主要进入线粒体,经历三羧酸循环,遵循糖的有氧氧化途径(黑色箭头路线所示)。(1)KRAS突变,致使葡萄糖转运体、糖酵解限速酶、糖代谢分支途径的关键酶(棕色)上调,糖代谢以无氧氧化为主(棕色箭头路线所示),从而促进葡萄糖摄取,增加糖酵解通量。(2)KRAS突变,刺激磷酸甘油酸激酶1向线粒体易位,致使癌细胞中丙酮酸脱氢酶激酶1磷酸化,抑制丙酮酸脱氢酶(蓝色箭头路线所示),从而抑制胰腺癌细胞中线粒体氧化磷酸化,产能减少。GFPT1:谷氨酰胺6-磷酸果糖转移酶;RPE:5-磷酸核丁糖-3-差向异构酶;RPIA:5-磷酸核丁糖异构酶。

    Figure  2.  KRAS mutation affects metabolic reprogramming in pancreatic ductal adenocarcinoma

    表  1  KRAS突变对PDAC代谢的影响

    Table  1.   Effects of KRAS mutations on PDAC metabolism

    影响类型 涉及机制 分子水平改变 最终效应
    肿瘤细胞代谢重编程 糖代谢 上调:葡萄糖转运体、己糖激酶、磷酸果糖激酶1、乳酸脱氢酶、谷氨酰胺6-磷酸果糖转移酶、5-磷酸核丁糖-3-差向异构酶、5-磷酸核丁糖异构酶 葡萄糖摄取↑乳酸产量↑糖酵解通量↑
    磷酸甘油酸激酶1线粒体易位 氧化磷酸化↓能量产生↓
    脂代谢 以细胞外脂质供应占主导 避免损耗,限制细胞死亡
    过度表达酰基辅酶A合成酶长链3 抑制细胞自噬
    氨基酸代谢 诱导:磷酸戊糖途径的代谢重组、天冬氨酸转氨酶的表达 谷氨酰胺代谢↑
    抑制:苹果酸脱氢酶1精氨酸甲基化、谷氨酸脱氢酶的表达、线粒体解耦连蛋白2的表达 维持肿瘤细胞活力
    巨胞饮与自噬作用 巨胞饮 突变型KRAS与肿瘤细胞表面的αvβ3和半乳糖凝集素3形成复合物 癌细胞巨胞饮作用↑
    促进关键效应因子V-ATPase向质膜易位 维持癌细胞的氧化还原平衡
    自噬 激活YAP-TAZ途径及其下游JAK-STAT3信号转导 诱导细胞自噬相关的MHC-I降解
    下载: 导出CSV
  • [1] SIEGEL RL, MILLER KD, FUCHS HE, et al. Cancer statistics, 2021[J]. CA Cancer J Clin, 2021, 71(1): 7-33. DOI: 10.3322/caac.21654.
    [2] 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. DOI: 10.3322/caac.21492.
    [3] HUANG L, JANSEN L, BALAVARCA Y, et al. Resection of pancreatic cancer in Europe and USA: an international large-scale study highlighting large variations[J]. Gut, 2019, 68(1): 130-139. DOI: 10.1136/gutjnl-2017-314828.
    [4] HENRIKSEN A, DYHL-POLK A, CHEN I, et al. Checkpoint inhibitors in pancreatic cancer[J]. Cancer Treat Rev, 2019, 78: 17-30. DOI: 10.1016/j.ctrv.2019.06.005.
    [5] INDINI A, RIJAVEC E, GHIDINI M, et al. Targeting KRAS in solid tumors: Current challenges and future opportunities of novel KRAS inhibitors[J]. Pharmaceutics, 2021, 13(5): 653. DOI: 10.3390/pharmaceutics13050653.
    [6] HOSEIN AN, BREKKEN RA, MAITRA A. Pancreatic cancer stroma: an update on therapeutic targeting strategies[J]. Nat Rev Gastroenterol Hepatol, 2020, 17(8): 487-505. DOI: 10.1038/s41575-020-0300-1.
    [7] SUN H, ZHANG B, LI H. The Roles of frequently mutated genes of pancreatic cancer in regulation of tumor microenvironment[J]. Technol Cancer Res Treat, 2020, 19: 1533033820920969. DOI: 10.1177/1533033820920969.
    [8] HOLLINSHEAD K, PARKER SJ, EAPEN VV, et al. Respiratory supercomplexes promote mitochondrial efficiency and growth in severely hypoxic pancreatic cancer[J]. Cell Rep, 2020, 33(1): 108231. DOI: 10.1016/j.celrep.2020.108231.
    [9] DEY P, LI J, ZHANG J, et al. Oncogenic KRAS-Driven metabolic reprogramming in pancreatic cancer cells utilizes cytokines from the tumor microenvironment[J]. Cancer Discov, 2020, 10(4): 608-625. DOI: 10.1158/2159-8290.CD-19-0297.
    [10] BUSCAIL L, BOURNET B, CORDELIER P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer[J]. Nat Rev Gastroenterol Hepatol, 2020, 17(3): 153-168. DOI: 10.1038/s41575-019-0245-4.
    [11] ENCARNACIÓN-ROSADO J, KIMMELMAN AC. Harnessing metabolic dependencies in pancreatic cancers[J]. Nat Rev Gastroenterol Hepatol, 2021, 18(7): 482-492. DOI: 10.1038/s41575-021-00431-7.
    [12] NAGDAS S, KASHATUS JA, NASCIMENTO A, et al. Drp1 promotes KRas-driven metabolic changes to drive pancreatic tumor growth[J]. Cell Rep, 2019, 28(7): 1845-1859.e5. DOI: 10.1016/j.celrep.2019.07.031.
    [13] HOU P, KAPOOR A, ZHANG Q, et al. Tumor microenvironment remodeling enables bypass of oncogenic KRAS dependency in pancreatic cancer[J]. Cancer Discov, 2020, 10(7): 1058-1077. DOI: 10.1158/2159-8290.CD-19-0597.
    [14] SANTANA-CODINA N, ROETH AA, ZHANG Y, et al. Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis[J]. Nat Commun, 2018, 9(1): 4945. DOI: 10.1038/s41467-018-07472-8.
    [15] LI X, JIANG Y, MEISENHELDER J, et al. Mitochondria-translocated PGK1 functions as a protein kinase to coordinate glycolysis and the TCA cycle in tumorigenesis[J]. Mol Cell, 2016, 61(5): 705-719. DOI: 10.1016/j.molcel.2016.02.009.
    [16] ROZEVELD CN, JOHNSON KM, ZHANG L, et al. KRAS controls pancreatic cancer cell lipid metabolism and invasive potential through the lipase HSL[J]. Cancer Res, 2020, 80(22): 4932-4945. DOI: 10.1158/0008-5472.CAN-20-1255.
    [17] ROSSI SEBASTIANO M, POZZATO C, SALIAKOURA M, et al. ACSL3-PAI-1 signaling axis mediates tumor-stroma cross-talk promoting pancreatic cancer progression[J]. Sci Adv, 2020, 6(44): eabb9200. DOI: 10.1126/sciadv.abb9200.
    [18] ROSSI SEBASTIANO M, KONSTANTINIDOU G. Targeting long chain Acyl-CoA synthetases for cancer therapy[J]. Int J Mol Sci, 2019, 20(15): 3624. DOI: 10.3390/ijms20153624.
    [19] SALIAKOURA M, SEBASTIANO MR, NIKDIMA I, et al. Restriction of extracellular lipids renders pancreatic cancer dependent on autophagy[J]. J Exp Clin Cancer Res, 2022, 41(1): 16. DOI: 10.1186/s13046-021-02231-y.
    [20] WANG YP, ZHOU W, WANG J, et al. Arginine methylation of MDH1 by CARM1 inhibits glutamine metabolism and suppresses pancreatic cancer[J]. Mol Cell, 2016, 64(4): 673-687. DOI: 10.1016/j.molcel.2016.09.028.
    [21] RAHO S, CAPOBIANCO L, MALIVINDI R, et al. KRAS-regulated glutamine metabolism requires UCP2-mediated aspartate transport to support pancreatic cancer growth[J]. Nat Metab, 2020, 2(12): 1373-1381. DOI: 10.1038/s42255-020-00315-1.
    [22] ZHANG Y, COMMISSO C. Macropinocytosis in cancer: A complex signaling network[J]. Trends Cancer, 2019, 5(6): 332-334. DOI: 10.1016/j.trecan.2019.04.002.
    [23] RAMIREZ C, HAUSER AD, VUCIC EA, et al. Plasma membrane V-ATPase controls oncogenic RAS-induced macropinocytosis[J]. Nature, 2019, 576(7787): 477-481. DOI: 10.1038/s41586-019-1831-x.
    [24] KINSEY CG, CAMOLOTTO SA, BOESPFLUG AM, et al. Publisher correction: Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers[J]. Nat Med, 2019, 25(5): 861. DOI: 10.1038/s41591-019-0433-3.
    [25] HUMPTON TJ, ALAGESAN B, DENICOLA GM, et al. Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer[J]. Cancer Discov, 2019, 9(9): 1268-1287. DOI: 10.1158/2159-8290.CD-18-1409.
    [26] YAMAMOTO K, VENIDA A, YANO J, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I[J]. Nature, 2020, 581(7806): 100-105. DOI: 10.1038/s41586-020-2229-5.
    [27] POMMIER A, ANAPARTHY N, MEMOS N, et al. Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases[J]. Science, 2018, 360(6394). DOI: 10.1126/science.aao4908.
    [28] DAI E, HAN L, LIU J, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein[J]. Autophagy, 2020, 16(11): 2069-2083. DOI: 10.1080/15548627.2020.1714209.
    [29] MONTERAN L, EREZ N. The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment[J]. Front Immunol, 2019, 10: 1835. DOI: 10.3389/fimmu.2019.01835.
    [30] HAMARSHEH S, GROβ O, BRUMMER T, et al. Immune modulatory effects of oncogenic KRAS in cancer[J]. Nat Commun, 2020, 11(1): 5439. DOI: 10.1038/s41467-020-19288-6.
    [31] WANG T, NOTTA F, NAVAB R, et al. Senescent carcinoma-associated fibroblasts upregulate IL8 to enhance prometastatic phenotypes[J]. Mol Cancer Res, 2017, 15(1): 3-14. DOI: 10.1158/1541-7786.MCR-16-0192.
    [32] HAFEZI S, SABER-AYAD M, ABDEL-RAHMAN WM. Highlights on the role of KRAS mutations in reshaping the microenvironment of pancreatic adenocarcinoma[J]. Int J Mol Sci, 2021, 22(19): 10219. DOI: 10.3390/ijms221910219.
    [33] NOLLMANN FI, RUESS DA. Targeting mutant KRAS in pancreatic cancer: Futile or promising?[J]. Biomedicines, 2020, 8(8): 281. DOI: 10.3390/biomedicines8080281.
    [34] CANON J, REX K, SAIKI AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity[J]. Nature, 2019, 575(7781): 217-223. DOI: 10.1038/s41586-019-1694-1.
    [35] HALLIN J, ENGSTROM LD, HARGIS L, et al. The KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients[J]. Cancer Discov, 2020, 10(1): 54-71. DOI: 10.1158/2159-8290.CD-19-1167.
    [36] HONG DS, FAKIH MG, STRICKLER JH, et al. KRASG12C inhibition with sotorasib in advanced solid tumors[J]. N Engl J Med, 2020, 383(13): 1207-1217. DOI: 10.1056/NEJMoa1917239.
    [37] JONCKHEERE N, VASSEUR R, VAN SEUNINGEN I. The cornerstone K-RAS mutation in pancreatic adenocarcinoma: From cell signaling network, target genes, biological processes to therapeutic targeting[J]. Crit Rev Oncol Hematol, 2017, 111: 7-19. DOI: 10.1016/j.critrevonc.2017.01.002.
    [38] DROSTEN M, BARBACID M. Targeting the MAPK pathway in KRAS-driven tumors[J]. Cancer Cell, 2020, 37(4): 543-550. DOI: 10.1016/j.ccell.2020.03.013.
    [39] TERRELL EM, MORRISON DK. Ras-mediated activation of the raf family kinases[J]. Cold Spring Harb Perspect Med, 2019, 9(1): a033746. DOI: 10.1101/cshperspect.a033746.
    [40] NAGASAKA M, POTUGARI B, NGUYEN A, et al. KRAS Inhibitors- yes but what next? Direct targeting of KRAS-vaccines, adoptive T cell therapy and beyond[J]. Cancer Treat Rev, 2021, 101: 102309. DOI: 10.1016/j.ctrv.2021.102309.
  • 加载中
图(2) / 表(1)
计量
  • 文章访问数:  1534
  • HTML全文浏览量:  1233
  • PDF下载量:  130
  • 被引次数: 0
出版历程
  • 收稿日期:  2022-05-24
  • 录用日期:  2022-08-10
  • 出版日期:  2022-12-20
  • 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