145
Views
33
CrossRef citations to date
0
Altmetric
Review

The role of Clusterin in cancer metastasis

, , , , , & show all
Pages 2405-2414 | Published online: 27 Mar 2019

References

  • Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147(2):275–292. doi:10.1016/j.cell.2011.09.02422000009
  • Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–1564. doi:10.1126/science.120354321436443
  • Reinhard F, Zhang CC, Shipitsin M, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi:10.1016/j.cell.2008.03.02718485877
  • Blaschuk O, Burdzy K, Fritz IB. Purification and characterization of a cell-aggregating factor (Clusterin), the major glycoprotein in ram rete testis fluid. J Biol Chem. 1983;258(12):7714–7720. 6863260
  • Ischia J, So AI. The role of heat shock proteins in bladder cancer. Nat Rev Urol. 2013;10(7):386–395. doi:10.1038/nrurol.2013.10823670183
  • Nelson AR, Sagare AP, Zlokovic BV. Role of Clusterin in the brain vascular clearance of amyloid-β. Proc Natl Acad Sci U S A. 2017;114(33):8681–8682. doi:10.1073/pnas.171135711428765369
  • Oh SB, Kim MS, Park S, et al. Clusterin contributes to early stage of Alzheimer’s disease pathogenesis. Brain Pathol. 2018. doi:10.1111/bpa.12660
  • Guitart K, Loers G, Buck F, et al. Improvement of neuronal cell survival by astrocyte-derived exosomes under hypoxic and ischemic conditions depends on prion protein. Glia. 2016;64(6):896–910. doi:10.1002/glia.2296326992135
  • Fang F, Chang R, Yang L. Heat shock factor 1 promotes invasion and metastasis of hepatocellular carcinoma in vitro and in vivo. Cancer. 2012;118(7):1782–1794. doi:10.1002/cncr.2648222009757
  • Shapiro B, Tocci P, Haase G, et al. Clusterin, a gene enriched in intestinal stem cells, is required for L1-mediated colon cancer metastasis. Oncotarget. 2015;6(33):34389–34401. doi:10.18632/oncotarget.536026399194
  • Makridakis M, Roubelakis MG, Bitsika V, et al. Analysis of secreted proteins for the study of bladder cancer cell aggressiveness. J Proteome Res. 2010;9(6):3243–3259. doi:10.1021/pr100189d20423150
  • Lau SH, Sham JS, Xie D, et al. Clusterin plays an important role in hepatocellular carcinoma metastasis. Oncogene. 2006;25(8):1242–1250. doi:10.1038/sj.onc.120914116247463
  • Wang C, Jiang K, Kang X, et al. Tumor-derived secretory Clusterin induces epithelial-mesenchymal transition and facilitates hepatocellular carcinoma metastasis. Int J Biochem Cell Biol. 2012;44(12):2308–2320. doi:10.1016/j.biocel.2012.09.01223010347
  • Miyake H, Gleave ME, Arakawa S, et al. Introducing the Clusterin gene into human renal cell carcinoma cells enhances their metastatic potential. J Urol. 2002;167(5):2203–2208. 11956479
  • Shiota M, Zardan A, Takeuchi A, et al. Clusterin mediates TGF-β-induced epithelial-mesenchymal transition and metastasis via Twist1 in prostate cancer cells. Cancer Res. 2012;72(20):5261–5272. doi:10.1158/0008-5472.CAN-12-025422896337
  • Wang C, Jin G, Jin H, et al. Clusterin facilitates metastasis by EIF3I/Akt/MMP13 signaling in hepatocellular carcinoma. Oncotarget. 2015;6(5):2903–2916. doi:10.18632/oncotarget.309325609201
  • Chen X, Halberg RB, Ehrhardt WM, et al. Clusterin as a biomarker in murine and human intestinal neoplasia. Proc Natl Acad Sci U S A. 2003;100(16):9530–9535. doi:10.1073/pnas.123363310012886021
  • Kang YK, Hong SW, Lee H, et al. Overexpression of Clusterin in human hepatocellular carcinoma. Hum Pathol. 2004;35(11):1340–1346. doi:10.1016/j.humpath.2004.07.02115668890
  • Chen RX, Song HY, Dong YY, et al. Dynamic expression patterns of differential proteins during early invasion of hepatocellular carcinoma. PLoS One. 2014;9(3):e88543. doi:10.1371/journal.pone.008854324614035
  • Shimizu T, Choi E, Petersen CP, et al. Characterization of progressive metaplasia in the gastric corpus mucosa of Mongolian gerbils infected with Helicobacter pylori. J Pathol. 2016;239(4):399–410. doi:10.1002/path.473527125972
  • Yu SY, Hong LC, Feng J, et al. Integrative proteomics and transcriptomics identify novel invasive-related biomarkers of non-functioning pituitary adenomas. Tumour Biol. 2016;37(7):8923–8930. doi:10.1007/s13277-015-4767-226753958
  • Liu K, He Q, Liao G, et al. Identification of critical genes and gene interaction networks that mediate osteosarcoma metastasis to the lungs. Exp Ther Med. 2015;10(5):1796–1806. doi:10.3892/etm.2015.276726640552
  • Zamay GS, Kolovskaya OS, Zamay TN, et al. Aptamers selected to postoperative lung adenocarcinoma detect circulating tumor cells in human blood. Mol Ther. 2015;23(9):1486–1496. doi:10.1038/mt.2015.10826061649
  • Zhang F, Kumano M, Beraldi E, et al. Clusterin facilitates stress-induced lipidation of LC3 and autophagosome biogenesis to enhance cancer cell survival. Nat Commun. 2014;5:5775. doi:10.1038/ncomms597225503391
  • Zhang H, Kim JK, Edwards CA, et al. Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol. 2005;7(9):909–915. doi:10.1038/ncb129116113678
  • Park DC, Yeo SG, Wilson MR, et al. Clusterin interacts with Paclitaxel and confer Paclitaxel resistance in ovarian cancer. Neoplasia. 2008;10(9):964–972. 18714397
  • Chen X, Bode AM, Dong Z, Cao Y. The epithelial-mesenchymal transition (EMT) is regulated by oncoviruses in cancer. FASEB J. 2016;30(9):3001–3010. doi:10.1096/fj.201600388R27279361
  • Mao XY, Li QQ, Gao YF, Zhou HH, Liu ZQ, Jin WL. Gap junction as an intercellular glue: emerging roles in cancer EMT and metastasis. Cancer Lett. 2016;381(1):133–137. doi:10.1016/j.canlet.2016.07.03727490999
  • Beerling E, Seinstra D, de Wit E, et al. Plasticity between epithelial and mesenchymal states unlinks EMT from metastasis-enhancing stem cell capacity. Cell Rep. 2016;14(10):2281–2288. doi:10.1016/j.celrep.2016.02.03426947068
  • Chou TY, Chen WC, Lee AC, et al. Clusterin silencing in human lung adenocarcinoma cells induces a mesenchymal-to-epithelial transition through modulating the ERK/Slug pathway. Cell Signal. 2009;21(5):704–711. doi:10.1016/j.cellsig.2009.01.00819166932
  • Jolly MK, Ware KE, Gilja S, et al. EMT and MET: necessary or permissive for metastasis? Mol Oncol. 2017;11(7):755–769. doi:10.1002/1878-0261.1208328548345
  • Jin G, Howe PH. Transforming growth factor beta regulates Clusterin gene expression via modulation of transcription factor c-Fos. Eur J Biochem. 1999;263(2):534–542. 10406964
  • Lenferink AE, Cantin C, Nantel A, et al. Transcriptome profiling of a TGF-beta-induced epithelial-to-mesenchymal transition reveals extracellular Clusterin as a target for therapeutic antibodies. Oncogene. 2010;29(6):831–844. doi:10.1038/onc.2009.39919935703
  • Trougakos IP, Lourda M, Antonelou MH, et al. Intracellular Clusterin inhibits mitochondrial apoptosis by suppressing p53-activating stress signals and stabilizing the cytosolic Ku70-Bax protein complex. Clin Cancer Res. 2009;15(1):48–59. doi:10.1158/1078-0432.CCR-08-180519118032
  • Criswell T, Klokov D, Beman M, et al. Repression of IR-inducible Clusterin expression by the p53 tumor suppressor protein. Cancer Biol Ther. 2003;2(4):372–380. 14508108
  • Klokov D, Leskov K, Araki S, et al. Low dose IR-induced IGF-1 sClusterin expression: a p53-repressed expression cascade that interferes with TGFβ1 signaling to confer a pro-survival bystander effect. Oncogene. 2013;32(4):479–490. doi:10.1038/onc.2012.6422391565
  • Goetz EM, Shankar B, Zou Y, et al. ATM-dependent IGF-1 induction regulates secretory Clusterin expression after DNA damage and in genetic instability. Oncogene. 2011;30(35):3745–3754. doi:10.1038/onc.2011.9221460853
  • Wang Y, Wang X, Zhao H, et al. Clusterin confers resistance to TNF-alpha-induced apoptosis in breast cancer cells through NF-kappaB activation and Bcl-2 overexpression. J Chemother. 2012;24(6):348–357. doi:10.1179/1973947812Y.000000004923174100
  • Santilli G, Aronow BJ, Sala A. Essential requirement of apolipoprotein J (Clusterin) signaling for Ilapak expression and regulation of NF-kappaB activity. J Biol Chem. 2003;278(40):38214–38219. doi:10.1074/jbc.C30025220012882985
  • Cervellera M, Raschella G, Santilli G, et al. Direct transactivation of the anti-apoptotic gene apolipoprotein J (Clusterin) by B-MYB. J Biol Chem. 2000;275(28):21055–21060. doi:10.1074/jbc.M00205520010770937
  • Li J, Jia L, Zhao P, et al. Stable knockdown of Clusterin by vectorbased RNA interference in a human breast cancer cell line inhibits tumour cell invasion and metastasis. J Int Med Res. 2012;40(2):545–555. doi:10.1177/14732300120400021622613415
  • Huang MB, Gonzalez RR, Lillard J, et al. Secretion modification region-derived peptide blocks exosome release and mediates cell cycle arrest in breast cancer cells. Oncotarget. 2017;8(7):11302–11315. doi:10.18632/oncotarget.1451328076321
  • Wang C, Jiang K, Gao D, et al. Clusterin protects hepatocellular carcinoma cells from endoplasmic reticulum stress induced apoptosis through GRP78. PLoS One. 2013;8(2):e55981. doi:10.1371/journal.pone.005598123457489
  • Mansoori B, Mohammadi A, Davudian S, et al. The different mechanisms of cancer drug resistance: a brief review. Adv Pharm Bull. 2017;7(3):339–348. doi:10.15171/apb.2017.04129071215
  • Tang L, Wei F, Wu Y, et al. Role of metabolism in cancer cell radioresistance and radiosensitization methods. J Exp Clin Cancer Res. 2018;37(1):87. doi:10.1186/s13046-018-0758-729688867
  • July LV, Akbari M, Zellweger T, et al. Clusterin expression is significantly enhanced in prostate cancer cells following androgen withdrawal therapy. Prostate. 2002;50(3):179–188. 11813210
  • Wang D, Liang H, Mao X, et al. Changes of transthyretin and Clusterin after androgen ablation therapy and correlation with prostate cancer malignancy. Transl Oncol. 2012;5(2):124–132. 22496929
  • Niu ZH, Wang Y, Chun B, et al. Secretory Clusterin (sClusterin) overexpression is associated with resistance to preoperative neoadjuvant chemotherapy in primary breast cancer. Eur Rev Med Pharmacol Sci. 2013;17(10):1337–1344. 23740447
  • Marchegiani G, Paulo JA, Sahora K, et al. The proteome of postsurgical pancreatic juice. Pancreas. 2015;44(4):574–582. doi:10.1097/MPA.000000000000030425875796
  • Xu M, Chen X, Han Y, et al. Clusterin silencing sensitizes pancreatic cancer MIA-PaCa-2 cells to gemcitabine via regulation of NF-kB/Bcl-2 signaling. Int J Clin Exp Med. 2015;8(8):12476–12486. 26550158
  • Wang X, Xie J, Lu X, et al. Melittin inhibits tumor growth and decreases resistance to gemcitabine by downregulating cholesterol pathway gene CLU in pancreatic ductal adenocarcinoma. Cancer Lett. 2017;399:1–9. doi:10.1016/j.canlet.2017.04.01228428074
  • Ma G, Cai H, Gao L, et al. sClusterin regulates cisplatin chemosensitivity of lung cancer cells in vivo. World J Surg Oncol. 2015;13:80. doi:10.1186/s12957-015-0501-125884382
  • Zhang B, Zhang K, Liu Z, et al. Secreted Clusterin gene silencing enhances chemosensitivity of a549 cells to cisplatin through AKT and ERK1/2 pathways in vitro. Cell Physiol Biochem. 2014;33(4):1162–1175. doi:10.1159/00035868524751980
  • Wang X, Zou F, Zhong J, et al. Secretory Clusterin mediates oxaliplatin resistance via the Gadd45a/PI3K/Akt signaling pathway in hepatocellular carcinoma. J Cancer. 2018;9(8):1403–1413. doi:10.7150/jca.2384929721050
  • Zhong J, Yu X, Dong X, et al. Downregulation of secreted Clusterin potentiates the lethality of sorafenib in hepatocellular carcinoma in association with the inhibition of ERK1/2 signals. Int J Mol Med. 2018;41(5):2893–2900. doi:10.3892/ijmm.2018.346329436591
  • Lamoureux F, Baud’huin M, Ory B, et al. Clusterin inhibition using OGX-011 synergistically enhances zoledronic acid activity in osteosarcoma. Oncotarget. 2014;5(17):7805–7819. doi:10.18632/oncotarget.230825138053
  • Matsumoto H, Yamamoto Y, Shiota M, et al. Cotargeting androgen receptor and Clusterin delays castrate-resistant prostate cancer progression by inhibiting adaptive stress response and AR stability. Cancer Res. 2013;73(16):5206–5217. doi:10.1158/0008-5472.CAN-13-035923786771
  • Kususda Y, Miyake H, Gleave ME, et al. Clusterin inhibition using OGX-011 synergistically enhances antitumour activity of sorafenib in a human renal cell carcinoma model. Br J Cancer. 2012;106(12):1945–1952. doi:10.1038/bjc.2012.20922588555
  • Laskin JJ, Nicholas G, Lee C, et al. Phase I/II trial of custirsen (OGX-011), an inhibitor of Clusterin, in combination with a gemcitabine and platinum regimen in patients with previously untreated advanced non-small cell lung cancer. J Thorac Oncol. 2012;7(3):579–586. doi:10.1097/JTO.0b013e31823f459c22198426
  • Oncogenex Pharmaceuticals. TA Multinational, Randomized, Open-Label Study of Custirsen In Patients With Advanced or Metastatic (Stage IV) Non-Small Cell Lung Cancer. Available from: https://clinicaltrials.gov/ct2/show/NCT01630733. NLM identifier: NCT01630733. Accessed December 1, 2018.
  • Chi KN, Hotte SJ, Yu EY, et al. Randomized phase II study of docetaxel and prednisone with or without OGX-011 in patients with metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28(27):4247–4254. doi:10.1200/JCO.2009.26.877120733135
  • Chi KN, Higano CS, Blumenstein B, et al. Custirsen in combination with docetaxel and prednisone for patients with metastatic castration-resistant prostate cancer (SYNERGY trial): a phase 3, multicentre, open-label, randomised trial. Lancet Oncol. 2017;18(4):473–485. doi:10.1016/S1470-2045(17)30168-728283282
  • Peng M, Darko KO, Tao T, et al. Combination of metformin with chemotherapeutic drugs via different molecular mechanisms. Cancer Treat Rev. 2017;54:24–33. doi:10.1016/j.ctrv.2017.01.00528161619
  • Deng J, Peng M, Wang Z, et al. Novel application of metformin combined with targeted drugs on anticancer treatment. Cancer Sci. 2018. doi:10.1111/cas.13849
  • Gupta GP, Massagué J. Cancer metastasis: building a framework. Cell. 2006;127:679–695. doi:10.1016/j.cell.2006.11.00117110329
  • Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nat Med. 2006;12:895–904. doi:10.1038/nm146916892035