3,902
Views
60
CrossRef citations to date
0
Altmetric
Research Paper - Basic Science

NUPR1 maintains autolysosomal efflux by activating SNAP25 transcription in cancer cells

, , , , , , , , , , , , , , , , , , ORCID Icon & show all
Pages 654-670 | Received 30 Sep 2016, Accepted 01 Jun 2017, Published online: 31 Dec 2017

References

  • Chen Y, Klionsky DJ. The regulation of autophagy – unanswered questions. J Cell Sci. 2011;124:161–170. doi:10.1242/jcs.064576. PMID:21187343.
  • Klionsky DJ, Abdelmohsen K, Abe A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 2016;12:1–222. doi:10.1080/15548627.2015.1100356. PMID:26799652.
  • Mizushima N, Levine B, Cuervo AM, et al. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075. doi:10.1038/nature06639. PMID:18305538.
  • White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer. 2012;12:401–410. doi:10.1038/nrc3262. PMID:22534666.
  • Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22:124–131. doi:10.1016/j.ceb.2009.11.014. PMID:20034776.
  • Galluzzi L, Pietrocola F, Bravo-San Pedro JM, et al. Autophagy in malignant transformation and cancer progression. Embo J. 2015;34:856–880. doi:10.15252/embj.201490784. PMID:25712477.
  • Cano CE, Hamidi T, Sandi MJ, et al. Nupr1: the Swiss-knife of cancer. J Cell Physiol. 2011;226:1439–1443. doi:10.1002/jcp.22324. PMID:20658514.
  • Goruppi S, Iovanna JL. Stress-inducible protein p8 is involved in several physiological and pathological processes. J Biol Chem. 2010;285:1577–1581. doi:10.1074/jbc.R109.080887. PMID:19926786.
  • Guo X, Wang W, Hu J, et al. Lentivirus-mediated RNAi knockdown of NUPR1 inhibits human nonsmall cell lung cancer growth in vitro and in vivo. Anat Rec (Hoboken). 2012;295:2114–2121. doi:10.1002/ar.22571. PMID:22961798.
  • Grasso D, Garcia MN, Hamidi T, et al. Genetic inactivation of the pancreatitis-inducible gene Nupr1 impairs PanIN formation by modulating Kras(G12D)-induced senescence. Cell Death Differ. 2014;21:1633–1641. doi:10.1038/cdd.2014.74. PMID:24902898.
  • Vincent AJ, Ren S, Harris LG, et al. Cytoplasmic translocation of p21 mediates NUPR1-induced chemoresistance: NUPR1 and p21 in chemoresistance. FEBS Lett. 2012;586:3429–3434. doi:10.1016/j.febslet.2012.07.063. PMID:22858377.
  • Carracedo A, Lorente M, Egia A, et al. The stress-regulated protein p8 mediates cannabinoid-induced apoptosis of tumor cells. Cancer cell. 2006;9:301–312. doi:10.1016/j.ccr.2006.03.005. PMID:16616335.
  • Emma MR, Iovanna JL, Bachvarov D, et al. NUPR1, a new target in liver cancer: implication in controlling cell growth, migration, invasion and sorafenib resistance. Cell Death Dis. 2016;7:e2269. doi:10.1038/cddis.2016.175. PMID:27336713.
  • Sandi MJ, Hamidi T, Malicet C, et al. p8 expression controls pancreatic cancer cell migration, invasion, adhesion, and tumorigenesis. J Cell Physiol. 2011;226:3442–3451. doi:10.1002/jcp.22702. PMID:21344397.
  • Ishida M, Miyamoto M, Naitoh S, et al. The SYT-SSX fusion protein down-regulates the cell proliferation regulator COM1 in t(x;18) synovial sarcoma. Mol Cell Biol. 2007;27:1348–1355. doi:10.1128/MCB.00658-06. PMID:17101797.
  • Jia Q, Zhou W, Yao W, et al. Downregulation of YAP-dependent Nupr1 promotes tumor-repopulating cell growth in soft matrices. Oncogenesis. 2016;5:e220. doi:10.1038/oncsis.2016.29. PMID:27089143.
  • Jiang WG, Davies G, Martin TA, et al. Com-1/p8 acts as a putative tumour suppressor in prostate cancer. Int J Mol Med. 2006;18:981–986. doi:10.3892/ijmm.18.5.981. PMID:17016631.
  • Hamidi T, Algul H, Cano CE, et al. Nuclear protein 1 promotes pancreatic cancer development and protects cells from stress by inhibiting apoptosis. J Clin Invest. 2012;122:2092–2103. doi:10.1172/JCI60144. PMID:22565310.
  • Gironella M, Malicet C, Cano C, et al. p8/nupr1 regulates DNA-repair activity after double-strand gamma irradiation-induced DNA damage. J Cell Physiol. 2009;221:594–602. doi:10.1002/jcp.21889. PMID:19650074.
  • Boya P, Andreau K, Poncet D, et al. Lysosomal membrane permeabilization induces cell death in a mitochondrion-dependent fashion. J Exp Med. 2003;197:1323–1334. doi:10.1084/jem.20021952. PMID:12756268.
  • Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death–apoptosis, autophagy and senescence. Febs J. 2010;277:2–21. doi:10.1111/j.1742-4658.2009.07366.x. PMID:19843174.
  • Hoyer-Hansen M, Bastholm L, Szyniarowski P, et al. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell. 2007;25:193–205. doi:10.1016/j.molcel.2006.12.009. PMID:17244528.
  • Wu LG, Hamid E, Shin W, et al. Exocytosis and endocytosis: modes, functions, and coupling mechanisms. Annu Rev Physiol. 2014;76:301–331. doi:10.1146/annurev-physiol-021113-170305. PMID:24274740.
  • Meng J, Wang J. Role of SNARE proteins in tumourigenesis and their potential as targets for novel anti-cancer therapeutics. Biochim Biophys Acta. 2015;1856:1–12. doi:10.1016/j.bbcan.2015.04.002. PMID:25956199.
  • Jean S, Cox S, Nassari S, et al. Starvation-induced MTMR13 and RAB21 activity regulates VAMP8 to promote autophagosome-lysosome fusion. EMBO Rep. 2015;16:297–311. doi:10.15252/embr.201439464. PMID:25648148.
  • Itakura E, Kishi-Itakura C, Mizushima N, et al. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012;151:1256–1269. doi:10.1016/j.cell.2012.11.001. PMID:23217709.
  • Soderberg O, Gullberg M, Jarvius M, et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods. 2006;3:995–1000. doi:10.1038/nmeth947. PMID:17072308.
  • Behrendorff N, Dolai S, Hong W, et al. Vesicle-associated membrane protein 8 (VAMP8) is a SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) selectively required for sequential granule-to-granule fusion. J Biol Chem. 2011;286:29627–29634. doi:10.1074/jbc.M111.265199. PMID:21733851.
  • Dickerson TJ, Smith GR, Pelletier JC, et al. 8-Hydroxyquinoline and hydroxamic acid inhibitors of botulinum neurotoxin BoNT/A. Curr Top Med Chem. 2014;14:2094–2102. doi:10.2174/1568026614666141022095114. PMID:25335884.
  • Eubanks LM, Hixon MS, Jin W, et al. An in vitro and in vivo disconnect uncovered through high-throughput identification of botulinum neurotoxin A antagonists. Proc Natl Acad Sci U S A. 2007;104:2602–2607. doi:10.1073/pnas.0611213104. PMID:17293454.
  • Xu J, Luo F, Zhang Z, et al. SNARE proteins synaptobrevin, SNAP-25, and syntaxin are involved in rapid and slow endocytosis at synapses. Cell Rep. 2013;3:1414–1421. doi:10.1016/j.celrep.2013.03.010. PMID:23643538.
  • Binz T, Blasi J, Yamasaki S, et al. Proteolysis of SNAP-25 by types E and A botulinal neurotoxins. J Biol Chem 1994;269:1617–20. PMID:8294407.
  • Sarkar S, Davies JE, Huang Z, et al. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J Biol Chem. 2007;282:5641–5652. doi:10.1074/jbc.M609532200. PMID:17182613.
  • He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009;43:67–93. doi:10.1146/annurev-genet-102808-114910. PMID:19653858.
  • Levine B, Kroemer G, et al. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42. doi:10.1016/j.cell.2007.12.018. PMID:18191218.
  • Pietrocola F, Izzo V, Niso-Santano M, et al. Regulation of autophagy by stress-responsive transcription factors. Semin Cancer Biol. 2013;23:310–322. doi:10.1016/j.semcancer.2013.05.008. PMID:23726895.
  • Fullgrabe J, Klionsky DJ, Joseph B. Histone post-translational modifications regulate autophagy flux and outcome. Autophagy. 2013;9:1621–1623. doi:10.4161/auto.25803. PMID:23934085.
  • Fullgrabe J, Klionsky DJ, Joseph B, et al. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol. 2014;15:65–74. doi:10.1038/nrm3716. PMID:24326622.
  • Boya P, Gonzalez-Polo RA, Casares N, et al. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol. 2005;25:1025–1040. doi:10.1128/MCB.25.3.1025-1040.2005. PMID:15657430.
  • Kabeya Y, Mizushima N, Ueno T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. Embo J. 2000;19:5720–5728. doi:10.1093/emboj/19.21.5720. PMID:11060023.
  • Yue Z, Jin S, Yang C, et al. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci U S A. 2003;100:15077–15082. doi:10.1073/pnas.2436255100. PMID:14657337.
  • Settembre C, Di Malta C, Polito VA, et al. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332:1429–1433. doi:10.1126/science.1204592. PMID:21617040.
  • Settembre C, Fraldi A, Medina DL, et al. Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol. 2013;14:283–296. doi:10.1038/nrm3565. PMID:23609508.
  • Chauhan S, Goodwin JG, Chauhan S, et al. ZKSCAN3 is a master transcriptional repressor of autophagy. Mol Cell. 2013;50:16–28. doi:10.1016/j.molcel.2013.01.024. PMID:23434374.
  • Shoji-Kawata S, Sumpter R, Leveno M, et al. Identification of a candidate therapeutic autophagy-inducing peptide. Nature. 2013;494:201–216. doi:10.1038/nature11866. PMID:23364696.
  • Liu Y, Shoji-Kawata S, Sumpter RM, Jr., et al. Autosis is a Na+,K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc Natl Acad Sci U S A. 2013;110:20364–20371. doi:10.1073/pnas.1319661110. PMID:24277826.
  • Perez-Mancera PA, Young AR, Narita M. Inside and out: the activities of senescence in cancer. Nat Rev Cancer. 2014;14:547–558. doi:10.1038/nrc3773. PMID:25030953.
  • Mathon NF, Lloyd AC. Cell senescence and cancer. Nat Rev Cancer. 2001;1:203–13. doi:10.1038/35106045. PMID:11902575.
  • Young AR, Narita M, Ferreira M, et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009;23:798–803. doi:10.1101/gad.519709. PMID:19279323.
  • Shen HM, Mizushima N. At the end of the autophagic road: an emerging understanding of lysosomal functions in autophagy. Trends Biochem Sci. 2014;39:61–71. doi:10.1016/j.tibs.2013.12.001. PMID:24369758.
  • Rong Y, Liu M, Ma L, et al. Clathrin and phosphatidylinositol-4,5-bisphosphate regulate autophagic lysosome reformation. Nat Cell Biol. 2012;14:924–934. doi:10.1038/ncb2557. PMID:22885770.
  • Zhao M, Wu S, Zhou Q, et al. Mechanistic insights into the recycling machine of the SNARE complex. Nature. 2015;518:61–67. doi:10.1038/nature14148. PMID:25581794.
  • Nair U, Jotwani A, Geng J, et al. SNARE proteins are required for macroautophagy. Cell. 2011;146:290–302. doi:10.1016/j.cell.2011.06.022. PMID:21784249.
  • Wu Y, Gu Y, Morphew MK, et al. All three components of the neuronal SNARE complex contribute to secretory vesicle docking. J Cell Biol. 2012;198:323–330. doi:10.1083/jcb.201106158. PMID:22869597.
  • Winkle CC, McClain LM, Valtschanoff JG, et al. A novel Netrin-1-sensitive mechanism promotes local SNARE-mediated exocytosis during axon branching. J Cell Biol. 2014;205:217–232. doi:10.1083/jcb.201311003. PMID:24778312.
  • Amaravadi RK, Yu D, Lum JJ, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117:326–336. doi:10.1172/JCI28833. PMID:17235397.
  • Degenhardt K, Mathew R, Beaudoin B, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10:51–64. doi:10.1016/j.ccr.2006.06.001. PMID:16843265.
  • Cai Q, Lu L, Tian JH, et al. Snapin-regulated late endosomal transport is critical for efficient autophagy-lysosomal function in neurons. Neuron. 2010;68:73–86. doi:10.1016/j.neuron.2010.09.022. PMID:20920792.
  • Diao J, Liu R, Rong Y, et al. ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes. Nature. 2015;520:563–566. doi:10.1038/nature14147. PMID:25686604.
  • Li X, Xu Z, Du W, et al. Aiolos promotes anchorage independence by silencing p66Shc transcription in cancer cells. Cancer cell. 2014;25:575–589. doi:10.1016/j.ccr.2014.03.020. PMID:24823637.
  • Wiederschain D, Wee S, Chen L, et al. Single-vector inducible lentiviral RNAi system for oncology target validation. Cell Cycle. 2009;8:498–504. doi:10.4161/cc.8.3.7701. PMID:19177017.
  • Ma Z, Liu Z, Wu RF, et al. p66(Shc) restrains Ras hyperactivation and suppresses metastatic behavior. Oncogene. 2010;29:5559–5567. doi:10.1038/onc.2010.326. PMID:20676142.
  • Allred DC, Harvey JM, Berardo M, et al. Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 1998;11:155–168. PMID:9504686.
  • Zhang K, Chen Y, Zhang Z, et al. Identification and verification of lysine propionylation and butyrylation in yeast core histones using PTMap software. J Proteome Res. 2009;8:900–906. doi:10.1021/pr8005155. PMID:19113941.
  • Zuniga JE, Schmidt JJ, Fenn T, et al. A potent peptidomimetic inhibitor of botulinum neurotoxin serotype A has a very different conformation than SNAP-25 substrate. Structure. 2008;16:1588–1597. doi:10.1016/j.str.2008.07.011. PMID:18940613.
  • Tsai YC, Maditz R, Kuo CL, et al. Targeting botulinum neurotoxin persistence by the ubiquitin-proteasome system. Proc Natl Acad Sci U S A. 2010;107:16554–16559. doi:10.1073/pnas.1008302107. PMID:20823219.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.