2,802
Views
87
CrossRef citations to date
0
Altmetric
Basic Research Paper

Autophagy is differentially induced in prostate cancer LNCaP, DU145 and PC-3 cells via distinct splicing profiles of ATG5

, , , , , & show all
Pages 20-32 | Received 28 Apr 2012, Accepted 28 Sep 2012, Published online: 17 Oct 2012

References

  • Abbas A, Gupta S. The role of histone deacetylases in prostate cancer. Epigenetics 2008; 3:300 - 9; http://dx.doi.org/10.4161/epi.3.6.7273; PMID: 19029799
  • Zhao X, Yang W, Shi C, Ma W, Liu J, Wang Y, et al. The G1 phase arrest and apoptosis by intrinsic pathway induced by valproic acid inhibit proliferation of BGC-823 gastric carcinoma cells. Tumour Biol 2011; 32:335 - 46; http://dx.doi.org/10.1007/s13277-010-0126-5; PMID: 21113745
  • Kuendgen A, Schmid M, Schlenk R, Knipp S, Hildebrandt B, Steidl C, et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer 2006; 106:112 - 9; http://dx.doi.org/10.1002/cncr.21552; PMID: 16323176
  • Weichert W, Röske A, Gekeler V, Beckers T, Stephan C, Jung K, et al. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 2008; 98:604 - 10; http://dx.doi.org/10.1038/sj.bjc.6604199; PMID: 18212746
  • Xia Q, Sung J, Chowdhury W, Chen CL, Höti N, Shabbeer S, et al. Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Res 2006; 66:7237 - 44; http://dx.doi.org/10.1158/0008-5472.CAN-05-0487; PMID: 16849572
  • Shabbeer S, Kortenhorst MS, Kachhap S, Galloway N, Rodriguez R, Carducci MA. Multiple Molecular pathways explain the anti-proliferative effect of valproic acid on prostate cancer cells in vitro and in vivo. Prostate 2007; 67:1099 - 110; http://dx.doi.org/10.1002/pros.20587; PMID: 17477369
  • Valentini A, Biancolella M, Amati F, Gravina P, Miano R, Chillemi G, et al. Valproic acid induces neuroendocrine differentiation and UGT2B7 up-regulation in human prostate carcinoma cell line. Drug Metab Dispos 2007; 35:968 - 72; http://dx.doi.org/10.1124/dmd.107.014662; PMID: 17371798
  • Sharma S, Symanowski J, Wong B, Dino P, Manno P, Vogelzang N. A phase II clinical trial of oral valproic acid in patients with castration-resistant prostate cancers using an intensive biomarker sampling strategy. Transl Oncol 2008; 1:141 - 7; PMID: 18795124
  • Long J, Zhao J, Yan Z, Liu Z, Wang N. Antitumor effects of a novel sulfur-containing hydroxamate histone deacetylase inhibitor H40. Int J Cancer 2009; 124:1235 - 44; http://dx.doi.org/10.1002/ijc.24074; PMID: 19058176
  • Iacopino F, Urbano R, Graziani G, Muzi A, Navarra P, Sica G. Valproic acid activity in androgen-sensitive and -insensitive human prostate cancer cells. Int J Oncol 2008; 32:1293 - 303; PMID: 18497991
  • Tanida I, Ueno T, Kominami E. LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 2004; 36:2503 - 18; http://dx.doi.org/10.1016/j.biocel.2004.05.009; PMID: 15325588
  • Mizushima N, Yamamoto A, Hatano M, Kobayashi Y, Kabeya Y, Suzuki K, et al. Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol 2001; 152:657 - 68; http://dx.doi.org/10.1083/jcb.152.4.657; PMID: 11266458
  • Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN, et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer 2009; 125:717 - 22; http://dx.doi.org/10.1002/ijc.24402; PMID: 19431142
  • Turner LS, Cheng JC, Beckham TH, Keane TE, Norris JS, Liu X. Autophagy is increased in prostate cancer cells overexpressing acid ceramidase and enhances resistance to C6 ceramide. Prostate Cancer Prostatic Dis 2011; 14:30 - 7; http://dx.doi.org/10.1038/pcan.2010.47; PMID: 21116286
  • Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy 2010; 6:322 - 9; http://dx.doi.org/10.4161/auto.6.3.11625; PMID: 20224296
  • Carew JS, Nawrocki ST, Kahue CN, Zhang H, Yang C, Chung L, et al. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007; 110:313 - 22; http://dx.doi.org/10.1182/blood-2006-10-050260; PMID: 17363733
  • Ko H, Kim YJ, Park JS, Park JH, Yang HO. Autophagy inhibition enhances apoptosis induced by ginsenoside Rk1 in hepatocellular carcinoma cells. Biosci Biotechnol Biochem 2009; 73:2183 - 9; http://dx.doi.org/10.1271/bbb.90250; PMID: 19809182
  • Wu Z, Chang PC, Yang JC, Chu CY, Wang LY, Chen NT, et al. Autophagy blockade sensitizes prostate cancer cells towards Src family kinase inhibitors. Genes Cancer 2010; 1:40 - 9; http://dx.doi.org/10.1177/1947601909358324; PMID: 20811583
  • Michaelis M, Suhan T, Michaelis UR, Beek K, Rothweiler F, Tausch L, et al. Valproic acid induces extracellular signal-regulated kinase 1/2 activation and inhibits apoptosis in endothelial cells. Cell Death Differ 2006; 13:446 - 53; http://dx.doi.org/10.1038/sj.cdd.4401759; PMID: 16167071
  • Fu J, Shao CJ, Chen FR, Ng HK, Chen ZP. Autophagy induced by valproic acid is associated with oxidative stress in glioma cell lines. Neuro Oncol 2010; 12:328 - 40; http://dx.doi.org/10.1093/neuonc/nop005; PMID: 20308311
  • Xiong N, Jia M, Chen C, Xiong J, Zhang Z, Huang J, et al. Potential autophagy enhancers attenuate rotenone-induced toxicity in SH-SY5Y. Neuroscience 2011; 199:292 - 302; http://dx.doi.org/10.1016/j.neuroscience.2011.10.031; PMID: 22056603
  • Ouyang D, Zhang Y, Xu L, Li J, Zha Q, He X. Histone deacetylase inhibitor valproic acid sensitizes B16F10 melanoma cells to cucurbitacin B treatment. Acta Biochim Biophys Sin (Shanghai) 2011; 43:487 - 95; http://dx.doi.org/10.1093/abbs/gmr032; PMID: 21628505
  • Cao C, Subhawong T, Albert JM, Kim KW, Geng L, Sekhar KR, et al. Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Cancer Res 2006; 66:10040 - 7; http://dx.doi.org/10.1158/0008-5472.CAN-06-0802; PMID: 17047067
  • Hanada T, Noda NN, Satomi Y, Ichimura Y, Fujioka Y, Takao T, et al. The Atg12-Atg5 conjugate has a novel E3-like activity for protein lipidation in autophagy. J Biol Chem 2007; 282:37298 - 302; http://dx.doi.org/10.1074/jbc.C700195200; PMID: 17986448
  • Korolchuk VI, Menzies FM, Rubinsztein DC. A novel link between autophagy and the ubiquitin-proteasome system. Autophagy 2009; 5:862 - 3; PMID: 19458478
  • Larsen KB, Lamark T, Øvervatn A, Harneshaug I, Johansen T, Bjørkøy G. A reporter cell system to monitor autophagy based on p62/SQSTM1. Autophagy 2010; 6:784 - 93; http://dx.doi.org/10.4161/auto.6.6.12510; PMID: 20574168
  • Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 2007; 282:24131 - 45; http://dx.doi.org/10.1074/jbc.M702824200; PMID: 17580304
  • Johansen T, Lamark T. Selective autophagy mediated by autophagic adapter proteins. Autophagy 2011; 7:279 - 96; http://dx.doi.org/10.4161/auto.7.3.14487; PMID: 21189453
  • Fujita K, Maeda D, Xiao Q, Srinivasula SM. Nrf2-mediated induction of p62 controls Toll-like receptor-4-driven aggresome-like induced structure formation and autophagic degradation. Proc Natl Acad Sci U S A 2011; 108:1427 - 32; http://dx.doi.org/10.1073/pnas.1014156108; PMID: 21220332
  • van Brussel JP, van Steenbrugge GJ, Romijn JC, Schröder FH, Mickisch GH. Chemosensitivity of prostate cancer cell lines and expression of multidrug resistance-related proteins. Eur J Cancer 1999; 35:664 - 71; http://dx.doi.org/10.1016/S0959-8049(98)00435-3; PMID: 10492644
  • Fujita N, Itoh T, Omori H, Fukuda M, Noda T, Yoshimori T. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell 2008; 19:2092 - 100; http://dx.doi.org/10.1091/mbc.E07-12-1257; PMID: 18321988
  • Sou YS, Tanida I, Komatsu M, Ueno T, Kominami E. Phosphatidylserine in addition to phosphatidylethanolamine is an in vitro target of the mammalian Atg8 modifiers, LC3, GABARAP, and GATE-16. J Biol Chem 2006; 281:3017 - 24; http://dx.doi.org/10.1074/jbc.M505888200; PMID: 16303767
  • Nishida Y, Arakawa S, Fujitani K, Yamaguchi H, Mizuta T, Kanaseki T, et al. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 2009; 461:654 - 8; http://dx.doi.org/10.1038/nature08455; PMID: 19794493
  • Barth S, Glick D, Macleod KF. Autophagy: assays and artifacts. J Pathol 2010; 221:117 - 24; http://dx.doi.org/10.1002/path.2694; PMID: 20225337
  • Vadlamudi RK, Joung I, Strominger JL, Shin J. p62, a phosphotyrosine-independent ligand of the SH2 domain of p56lck, belongs to a new class of ubiquitin-binding proteins. J Biol Chem 1996; 271:20235 - 7; http://dx.doi.org/10.1074/jbc.271.34.20235; PMID: 8702753
  • Watanabe Y, Tanaka M. p62/SQSTM1 in autophagic clearance of a non-ubiquitylated substrate. J Cell Sci 2011; 124:2692 - 701; http://dx.doi.org/10.1242/jcs.081232; PMID: 21771882
  • Seibenhener ML, Babu JR, Geetha T, Wong HC, Krishna NR, Wooten MW. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol Cell Biol 2004; 24:8055 - 68; http://dx.doi.org/10.1128/MCB.24.18.8055-8068.2004; PMID: 15340068
  • Shvets E, Elazar Z. Autophagy-independent incorporation of GFP-LC3 into protein aggregates is dependent on its interaction with p62/SQSTM1. Autophagy 2008; 4:1054 - 6; PMID: 18776740
  • Nelson WG, Kastan MB. DNA strand breaks: the DNA template alterations that trigger p53-dependent DNA damage response pathways. Mol Cell Biol 1994; 14:1815 - 23; PMID: 8114714
  • Schneider E, Montenarh M, Wagner P. Regulation of CAK kinase activity by p53. Oncogene 1998; 17:2733 - 41; http://dx.doi.org/10.1038/sj.onc.1202504; PMID: 9840937
  • van Bokhoven A, Varella-Garcia M, Korch C, Johannes WU, Smith EE, Miller HL, et al. Molecular characterization of human prostate carcinoma cell lines. Prostate 2003; 57:205 - 25; http://dx.doi.org/10.1002/pros.10290; PMID: 14518029
  • Chen X, Wong JY, Wong P, Radany EH. Low-dose valproic acid enhances radiosensitivity of prostate cancer through acetylated p53-dependent modulation of mitochondrial membrane potential and apoptosis. Mol Cancer Res 2011; 9:448 - 61; http://dx.doi.org/10.1158/1541-7786.MCR-10-0471; PMID: 21303901
  • Fortson WS, Kayarthodi S, Fujimura Y, Xu H, Matthews R, Grizzle WE, et al. Histone deacetylase inhibitors, valproic acid and trichostatin-A induce apoptosis and affect acetylation status of p53 in ERG-positive prostate cancer cells. Int J Oncol 2011; 39:111 - 9; PMID: 21519790
  • Roy S, Packman K, Jeffrey R, Tenniswood M. Histone deacetylase inhibitors differentially stabilize acetylated p53 and induce cell cycle arrest or apoptosis in prostate cancer cells. Cell Death Differ 2005; 12:482 - 91; http://dx.doi.org/10.1038/sj.cdd.4401581; PMID: 15746940
  • Catalano MG, Fortunati N, Pugliese M, Poli R, Bosco O, Mastrocola R, et al. Valproic acid, a histone deacetylase inhibitor, enhances sensitivity to doxorubicin in anaplastic thyroid cancer cells. J Endocrinol 2006; 191:465 - 72; http://dx.doi.org/10.1677/joe.1.06970; PMID: 17088416
  • Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN. Valproic acid alters chromatin structure by regulation of chromatin modulation proteins. Cancer Res 2005; 65:3815 - 22; http://dx.doi.org/10.1158/0008-5472.CAN-04-2478; PMID: 15867379
  • Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett A, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 2007; 39:596 - 604; http://dx.doi.org/10.1038/ng2032; PMID: 17435756
  • Naser SA, Arce M, Khaja A, Fernandez M, Naser N, Elwasila S, et al. Role of ATG16L, NOD2 and IL23R in Crohn’s disease pathogenesis. World J Gastroenterol 2012; 18:412 - 24; http://dx.doi.org/10.3748/wjg.v18.i5.412; PMID: 22346247
  • Plantinga TS, Crisan TO, Oosting M, van de Veerdonk FL, de Jong DJ, Philpott DJ, et al. Crohn’s disease-associated ATG16L1 polymorphism modulates pro-inflammatory cytokine responses selectively upon activation of NOD2. Gut 2011; 60:1229 - 35; http://dx.doi.org/10.1136/gut.2010.228908; PMID: 21406388
  • Plantinga TS, Joosten LA, Netea MG. ATG16L1 polymorphisms are associated with NOD2-induced hyperinflammation. Autophagy 2011; 7:1074 - 5; http://dx.doi.org/10.4161/auto.7.9.15867; PMID: 21673517
  • Ouyang D, He X, Xu L, Wang X, Gao Q, Guo H. Differential cell surface expression of rhesus macaque’s major histocompatibility complex class I alleles Mamu-B*1703 and Mamu-B*0101. Acta Biochim Biophys Sin (Shanghai) 2010; 42:281 - 7; http://dx.doi.org/10.1093/abbs/gmq019; PMID: 20383467