全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Cells  2012 

14-3-3 Proteins are Regulators of Autophagy

DOI: 10.3390/cells1040754

Keywords: 14-3-3 proteins, autophagy, cell signaling

Full-Text   Cite this paper   Add to My Lib

Abstract:

14-3-3 proteins are implicated in the regulation of proteins involved in a variety of signaling pathways. 14-3-3-dependent protein regulation occurs through phosphorylation-dependent binding that results, in many cases, in the release of survival signals in cells. Autophagy is a cell digestion process that contributes to overcoming nutrient deprivation and is initiated under stress conditions. However, whether autophagy is a cell survival or cell death mechanism remains under discussion and may depend on context. Nevertheless, autophagy is a cellular process that determines cell fate and is tightly regulated by different signaling pathways, some of which, for example MAPK, PI3K and mTOR, are tightly regulated by 14-3-3 proteins. It is therefore important to understand the role of 14-3-3 protein in modulating the autophagic process. Within this context, direct binding of 14-3-3 to mTOR regulatory proteins, such as TSC2 and PRAS40, connects 14-3-3 with autophagy regulatory processes. In addition, 14-3-3 binding to human vacuolar protein sorting 34 (hVps34), a class III phosphatidylinositol-3-kinase (PI3KC3), indicates the involvement of 14-3-3 proteins in regulating autophagosome formation. hVps34 is involved in vesicle trafficking processes such as autophagy, and its activation is needed for initiation of autophagy. Chromatography and overlay techniques suggest that hVps34 directly interacts with 14-3-3 proteins under physiological conditions, thereby maintaining hVps34 in an inactive state. In contrast, nutrient starvation promotes dissociation of the 14-3-3–hVps34 complex, thereby enhancing hVps34 lipid kinase activity. Thus, 14-3-3 proteins are regulators of autophagy through regulating key components of the autophagic machinery. This review summarizes the role of 14-3-3 protein in the control of target proteins involved in regulating the master switches of autophagy.

References

[1]  Chaudhri, M.; Scarabel, M.; Aitken, A. Mammalian and yeast 14-3-3 isoforms form distinct patterns of dimers in vivo. Biochem. Biophys. Res. Commun. 2003, 300, 679–685, doi:10.1016/S0006-291X(02)02902-9.
[2]  Jones, D.H.; Ley, S.; Aitken, A. Isoforms of 14-3-3 protein can form homo- and heterodimers in vivo and in vitro: implications for function as adapter proteins. FEBS Lett. 1995, 368, 55–58, doi:10.1016/0014-5793(95)00598-4.
[3]  Mackintosh, C. Dynamic interactions between 14-3-3 proteins and phosphoproteins regulate diverse cellular processes. Biochem J. 2004, 381, 329–342, doi:10.1042/BJ20031332.
[4]  Muslin, A.J.; Tanner, J.W.; Allen, P.M.; Shaw, A.S. Interaction of 14-3-3 with signaling proteins is mediated by the recognition of phosphoserine. Cell 1996, 84, 889–897, doi:10.1016/S0092-8674(00)81067-3.
[5]  Yaffe, M.B.; Rittinger, K.; Volinia, S.; Caron, P.R.; Aitken, A.; Leffers, H.; Gamblin, S.J.; Smerdon, S.J.; Cantley, L.C. The structural basis for 14-3-3: phosphopeptide binding specificity. Cell 1997, 91, 961–971, doi:10.1016/S0092-8674(00)80487-0.
[6]  Johnson, C.; Crowther, S.; Stafford, M.J.; Campbell, D.G.; Toth, R.; MacKintosh, C. Bioinformatic and experimental survey of 14-3-3-binding sites. Biochem J. 2010, 427, 69–78, doi:10.1042/BJ20091834.
[7]  Aitken, A.; Howell, S.; Jones, D.; Madrazo, J.; Patel, Y. 14-3-3 alpha and delta are the phosphorylated forms of raf-activating 14-3-3 beta and zeta. In vivo stoichiometric phosphorylation in brain at a Ser-Pro-Glu-Lys MOTIF. In J. Biol. Chem.; 1995; Volume 270, pp. 5706–5709.
[8]  Moreira, J.M.; Shen, T.; Ohlsson, G.; Gromov, P.; Gromova, I.; Celis, J.E. A combined proteome and ultrastructural localization analysis of 14-3-3 proteins in transformed human amnion (AMA) cells: definition of a framework to study isoform-specific differences. Mol. Cell. Proteomics 2008, 7, 1225–1240, doi:10.1074/mcp.M700439-MCP200.
[9]  Kilani, R.T.; Medina, A.; Aitken, A.; Jalili, R.B.; Carr, M.; Ghahary, A. Identification of different isoforms of 14-3-3 protein family in human dermal and epidermal layers. Mol. Cell. Biochem. 2008, 314, 161–169, doi:10.1007/s11010-008-9777-6.
[10]  Yaffe, M.B. How do 14-3-3 proteins work? Gatekeeper phosphorylation and the molecular anvil hypothesis. FEBS Lett. 2002, 513, 53–57, doi:10.1016/S0014-5793(01)03288-4.
[11]  Wilker, E.; Yaffe, M.B. 14-3-3 Proteins—a focus on cancer and human disease. J. Mol. Cell. Cardiol. 2004, 37, 633–642, doi:10.1016/j.yjmcc.2004.04.015.
[12]  Meek, S.E.; Lane, W.S.; Piwnica-Worms, H. Comprehensive proteomic analysis of interphase and mitotic 14-3-3-binding proteins. J. Biol. Chem. 2004, 279, 32046–32054, doi:10.1074/jbc.M403044200.
[13]  Pozuelo Rubio, M.; Geraghty, K.M.; Wong, B.H.; Wood, N.T.; Campbell, D.G.; Morrice, N.; Mackintosh, C. 14-3-3-affinity purification of over 200 human phosphoproteins reveals new links to regulation of cellular metabolism, proliferation and trafficking. Biochem. J. 2004, 379, 395–408, doi:10.1042/BJ20031797.
[14]  Jin, J.; Smith, F.D.; Stark, C.; Wells, C.D.; Fawcett, J.P.; Kulkarni, S.; Metalnikov, P.; O’Donnell, P.; Taylor, P.; Taylor, L.; Zougman, A.; Woodgett, J.R.; Langeberg, L.K.; Scott, J.D.; Pawson, T. Proteomic, functional, and domain-based analysis of in vivo 14-3-3 binding proteins involved in cytoskeletal regulation and cellular organization. Curr. Biol. 2004, 14, 1436–1450, doi:10.1016/j.cub.2004.07.051.
[15]  Kjarland, E.; Keen, T.J.; Kleppe, R. Does isoform diversity explain functional differences in the 14-3-3 protein family’? Curr. Pharm. Biotechnol. 2006, 7, 217–223, doi:10.2174/138920106777549777.
[16]  Pozuelo-Rubio, M. Proteomic and biochemical analysis of 14-3-3-binding proteins during C2- ceramide-induced apoptosis. FEBS J. 2010, 277, 3321–3342, doi:10.1111/j.1742-4658.2010.07730.x.
[17]  Chang, I.F.; Curran, A.; Woolsey, R.; Quilici, D.; Cushman, J.C.; Mittler, R.; Harmon, A.; Harper, J.F. Proteomic profiling of tandem affinity purified 14-3-3 protein complexes in Arabidopsis thaliana. Proteomics 2009, 9, 2967–2985, doi:10.1002/pmic.200800445.
[18]  Puri, P.; Myers, K.; Kline, D.; Vijayaraghavan, S. Proteomic analysis of bovine sperm YWHA binding partners identify proteins involved in signaling and metabolism. Biol. Reprod. 2008, 79, 1183–1191, doi:10.1095/biolreprod.108.068734.
[19]  Benzinger, A.; Muster, N.; Koch, H.B.; Yates, J.R., 3rd; Hermeking, H. Targeted proteomic analysis of 14-3-3 sigma, a p53 effector commonly silenced in cancer. Mol. Cell. Proteomics 2005, 4, 785–795, doi:10.1074/mcp.M500021-MCP200.
[20]  Masters, S.C.; Subramanian, R.R.; Truong, A.; Yang, H.; Fujii, K.; Zhang, H.; Fu, H. Survival-promoting functions of 14-3-3 proteins. Biochem. Soc. Trans. 2002, 30, 360–365.
[21]  Zha, J.; Harada, H.; Yang, E.; Jockel, J.; Korsmeyer, S.J. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 1996, 87, 619–628, doi:10.1016/S0092-8674(00)81382-3.
[22]  Nomura, M.; Shimizu, S.; Sugiyama, T.; Narita, M.; Ito, T.; Matsuda, H.; Tsujimoto, Y. 14-3-3 Interacts directly with and negatively regulates pro-apoptotic Bax. J. Biol. Chem. 2003, 278, 2058–2065.
[23]  Zhang, L.; Chen, J.; Fu, H. Suppression of apoptosis signal-regulating kinase 1-induced cell death by 14-3-3 proteins. Proc. Natl. Acad. Sci. USA 1999, 96, 8511–8515, doi:10.1073/pnas.96.15.8511.
[24]  Brunet, A.; Bonni, A.; Zigmond, M.J.; Lin, M.Z.; Juo, P.; Hu, L.S.; Anderson, M.J.; Arden, K.C.; Blenis, J.; Greenberg, M.E. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 1999, 96, 857–868, doi:10.1016/S0092-8674(00)80595-4.
[25]  Masters, S.C.; Fu, H. 14-3-3 proteins mediate an essential anti-apoptotic signal. J. Biol. Chem. 2001, 276, 45193–45200, doi:10.1074/jbc.M105971200.
[26]  Cao, W.; Yang, X.; Zhou, J.; Teng, Z.; Cao, L.; Zhang, X.; Fei, Z. Targeting 14-3-3 protein, difopein induces apoptosis of human glioma cells and suppresses tumour growth in mice. Apoptosis 2010, 15, 230–241, doi:10.1007/s10495-009-0437-4.
[27]  Qi, W.; Martinez, J.D. Reduction of 14-3-3 proteins correlates with increased sensitivity to killing of human lung cancer cells by ionizing radiation. Radiat. Res. 2003, 160, 217–223, doi:10.1667/RR3038.
[28]  Niemantsverdriet, M.; Wagner, K.; Visser, M.; Backendorf, C. Cellular functions of 14-3-3 zeta in apoptosis and cell adhesion emphasize its oncogenic character. Oncogene 2008, 27, 1315–1319, doi:10.1038/sj.onc.1210742.
[29]  Neal, C.L.; Yao, J.; Yang, W.; Zhou, X.; Nguyen, N.T.; Lu, J.; Danes, C.G.; Guo, H.; Lan, K.H.; Ensor, J.; Hittelman, W.; Hung, M.C.; Yu, D. 14-3-3zeta overexpression defines high risk for breast cancer recurrence and promotes cancer cell survival. Cancer Res. 2009, 69, 3425–3432, doi:10.1158/0008-5472.CAN-08-2765.
[30]  Kumar, A.P.; Garcia, G.E.; Orsborn, J.; Levin, V.A.; Slaga, T.J. 2-Methoxyestradiol interferes with NF kappa B transcriptional activity in primitive neuroectodermal brain tumors: implications for management. Carcinogenesis 2003, 24, 209–216, doi:10.1093/carcin/24.2.209.
[31]  Levine, B.; Klionsky, D.J. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev. Cell 2004, 6, 463–477, doi:10.1016/S1534-5807(04)00099-1.
[32]  Kroemer, G.; Marino, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293, doi:10.1016/j.molcel.2010.09.023.
[33]  Shintani, T.; Klionsky, D.J. Autophagy in health and disease: a double-edged sword. Science 2004, 306, 990–995, doi:10.1126/science.1099993.
[34]  Rubinsztein, D.C.; Gestwicki, J.E.; Murphy, L.O.; Klionsky, D.J. Potential therapeutic applications of autophagy. Nat. Rev. Drug Discov. 2007, 6, 304–312, doi:10.1038/nrd2272.
[35]  Galluzzi, L.; Aaronson, S.A.; Abrams, J.; Alnemri, E.S.; Andrews, D.W.; Baehrecke, E.H.; Bazan, N.G.; Blagosklonny, M.V.; Blomgren, K.; Borner, C.; Bredesen, D.E.; Brenner, C.; Castedo, M.; Cidlowski, J.A.; Ciechanover, A.; Cohen, G.M.; De Laurenzi, V.; De Maria, R.; Deshmukh, M.; Dynlacht, B.D.; El-Deiry, W.S.; Flavell, R.A.; Fulda, S.; Garrido, C.; Golstein, P.; Gougeon, M.L.; Green, D.R.; Gronemeyer, H.; Hajnoczky, G.; Hardwick, J.M.; Hengartner, M.O.; Ichijo, H.; Jaattela, M.; Kepp, O.; Kimchi, A.; Klionsky, D.J.; Knight, R.A.; Kornbluth, S.; Kumar, S.; Levine, B.; Lipton, S.A.; Lugli, E.; Madeo, F.; Malomi, W.; Marine, J.C.; Martin, S.J.; Medema, J.P.; Mehlen, P.; Melino, G.; Moll, U.M.; Morselli, E.; Nagata, S.; Nicholson, D.W.; Nicotera, P.; Nunez, G.; Oren, M.; Penninger, J.; Pervaiz, S.; Peter, M.E.; Piacentini, M.; Prehn, J.H.; Puthalakath, H.; Rabinovich, G.A.; Rizzuto, R.; Rodrigues, C.M.; Rubinsztein, D.C.; Rudel, T.; Scorrano, L.; Simon, H.U.; Steller, H.; Tschopp, J.; Tsujimoto, Y.; Vandenabeele, P.; Vitale, I.; Vousden, K.H.; Youle, R.J.; Yuan, J.; Zhivotovsky, B.; Kroemer, G. Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes. Cell Death Differ. 2009, 16, 1093–1107.
[36]  Kroemer, G.; Jaattela, M. Lysosomes and autophagy in cell death control. Nat. Rev. Cancer 2005, 5, 886–897, doi:10.1038/nrc1738.
[37]  Maiuri, M.C.; Zalckvar, E.; Kimchi, A.; Kroemer, G. Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol 2007, 8, 741–752, doi:10.1038/nrm2239.
[38]  Mizushima, N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr. Opin. Cell Biol. 2010, 22, 132–139, doi:10.1016/j.ceb.2009.12.004.
[39]  Jung, C.H.; Jun, C.B.; Ro, S.H.; Kim, Y.M.; Otto, N.M.; Cao, J.; Kundu, M.; Kim, D.H. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 2009, 20, 1992–2003, doi:10.1091/mbc.E08-12-1249.
[40]  Chang, Y.Y.; Juhasz, G.; Goraksha-Hicks, P.; Arsham, A.M.; Mallin, D.R.; Muller, L.K.; Neufeld, T.P. Nutrient-dependent regulation of autophagy through the target of rapamycin pathway. Biochem. Soc. Trans. 2009, 37, 232–236, doi:10.1042/BST0370232.
[41]  Bjorkoy, G.; Lamark, T.; Brech, A.; Outzen, H.; Perander, M.; Overvatn, A.; Stenmark, H.; Johansen, T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 2005, 171, 603–614, doi:10.1083/jcb.200507002.
[42]  Pankiv, S.; Clausen, T.H.; Lamark, T.; Brech, A.; Bruun, J.A.; Outzen, H.; Overvatn, A.; Bjorkoy, G.; Johansen, T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282, 24131–24145.
[43]  Levine, B.; Yuan, J. Autophagy in cell death: an innocent convict? J. Clin. Invest. 2005, 115, 2679–2688, doi:10.1172/JCI26390.
[44]  Baehrecke, E.H. Autophagy: dual roles in life and death? Nat. Rev. Mol. Cell Biol. 2005, 6, 505–510, doi:10.1038/nrm1666.
[45]  Kondo, Y.; Kanzawa, T.; Sawaya, R.; Kondo, S. The role of autophagy in cancer development and response to therapy. Nat. Rev. Cancer 2005, 5, 726–734, doi:10.1038/nrc1692.
[46]  Gozuacik, D.; Kimchi, A. Autophagy and cell death. Curr. Top. Dev. Biol. 2007, 78, 217–245, doi:10.1016/S0070-2153(06)78006-1.
[47]  Lockshin, R.A.; Zakeri, Z. Programmed cell death and apoptosis: origins of the theory. Nat Rev. Mol. Cell Biol. 2001, 2, 545–550, doi:10.1038/35080097.
[48]  Daido, S.; Kanzawa, T.; Yamamoto, A.; Takeuchi, H.; Kondo, Y.; Kondo, S. Pivotal role of the cell death factor BNIP3 in ceramide-induced autophagic cell death in malignant glioma cells. Cancer Res. 2004, 64, 4286–4293, doi:10.1158/0008-5472.CAN-03-3084.
[49]  Steinberg, G.R.; Kemp, B.E. AMPK in Health and Disease. Physiol. Rev. 2009, 89, 1025–1078, doi:10.1152/physrev.00011.2008.
[50]  Wullschleger, S.; Loewith, R.; Hall, M.N. TOR signaling in growth and metabolism. Cell 2006, 124, 471–484, doi:10.1016/j.cell.2006.01.016.
[51]  Inoki, K.; Zhu, T.; Guan, K.L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003, 115, 577–590, doi:10.1016/S0092-8674(03)00929-2.
[52]  Corradetti, M.N.; Inoki, K.; Bardeesy, N.; DePinho, R.A.; Guan, K.L. Regulation of the TSC pathway by LKB1: evidence of a molecular link between tuberous sclerosis complex and Peutz-Jeghers syndrome. Genes Dev. 2004, 18, 1533–1538, doi:10.1101/gad.1199104.
[53]  Shaw, R.J.; Bardeesy, N.; Manning, B.D.; Lopez, L.; Kosmatka, M.; DePinho, R.A.; Cantley, L.C. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell 2004, 6, 91–99, doi:10.1016/j.ccr.2004.06.007.
[54]  Liu, L.; Cash, T.P.; Jones, R.G.; Keith, B.; Thompson, C.B.; Simon, M.C. Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol. Cell 2006, 21, 521–531, doi:10.1016/j.molcel.2006.01.010.
[55]  Shaw, R.J. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol (Oxf) 2009, 196, 65–80, doi:10.1111/j.1748-1716.2009.01972.x.
[56]  Miyazaki, M.; McCarthy, J.J.; Esser, K.A. Insulin like growth factor-1-induced phosphorylation and altered distribution of tuberous sclerosis complex (TSC)1/TSC2 in C2C12 myotubes. FEBS J. 2010, 277, 2180–2191, doi:10.1111/j.1742-4658.2010.07635.x.
[57]  Cai, S.L.; Tee, A.R.; Short, J.D.; Bergeron, J.M.; Kim, J.; Shen, J.; Guo, R.; Johnson, C.L.; Kiguchi, K.; Walker, C.L. Activity of TSC2 is inhibited by AKT-mediated phosphorylation and membrane partitioning. J. Cell Biol. 2006, 173, 279–289, doi:10.1083/jcb.200507119.
[58]  Huang, J.; Manning, B.D. A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem. Soc. Trans. 2009, 37, 217–222, doi:10.1042/BST0370217.
[59]  Sancak, Y.; Thoreen, C.C.; Peterson, T.R.; Lindquist, R.A.; Kang, S.A.; Spooner, E.; Carr, S.A.; Sabatini, D.M. PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. Mol. Cell 2007, 25, 903–915, doi:10.1016/j.molcel.2007.03.003.
[60]  Vander Haar, E.; Lee, S.I.; Bandhakavi, S.; Griffin, T.J.; Kim, D.H. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat. Cell Biol. 2007, 9, 316–323, doi:10.1038/ncb1547.
[61]  Wang, L.; Harris, T.E.; Lawrence, J.C., Jr. Regulation of proline-rich Akt substrate of 40 kDa (PRAS40) function by mammalian target of rapamycin complex 1 (mTORC1)-mediated phosphorylation. J. Biol. Chem. 2008, 283, 15619–15627.
[62]  Nascimento, E.B.; Snel, M.; Guigas, B.; van der Zon, G.C.; Kriek, J.; Maassen, J.A.; Jazet, I.M.; Diamant, M.; Ouwens, D.M. Phosphorylation of PRAS40 on Thr246 by PKB/AKT facilitates efficient phosphorylation of Ser183 by mTORC1. Cell Signal. 2010, 22, 961–967, doi:10.1016/j.cellsig.2010.02.002.
[63]  Fonseca, B.D.; Smith, E.M.; Lee, V.H.; MacKintosh, C.; Proud, C.G. PRAS40 is a target for mammalian target of rapamycin complex 1 and is required for signaling downstream of this complex. J. Biol. Chem. 2007, 282, 24514–24524.
[64]  Gwinn, D.M.; Shackelford, D.B.; Egan, D.F.; Mihaylova, M.M.; Mery, A.; Vasquez, D.S.; Turk, B.E.; Shaw, R.J. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 2008, 30, 214–226, doi:10.1016/j.molcel.2008.03.003.
[65]  Brugarolas, J.; Lei, K.; Hurley, R.L.; Manning, B.D.; Reiling, J.H.; Hafen, E.; Witters, L.A.; Ellisen, L.W.; Kaelin, W.G., Jr. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 2004, 18, 2893–2904, doi:10.1101/gad.1256804.
[66]  Corradetti, M.N.; Inoki, K.; Guan, K.L. The stress-induced proteins RTP801 and RTP801L are negative regulators of the mammalian target of rapamycin pathway. J. Biol. Chem. 2005, 280, 9769–9772.
[67]  Sofer, A.; Lei, K.; Johannessen, C.M.; Ellisen, L.W. Regulation of mTOR and cell growth in response to energy stress by REDD1. Mol. Cell Biol. 2005, 25, 5834–5845, doi:10.1128/MCB.25.14.5834-5845.2005.
[68]  DeYoung, M.P.; Horak, P.; Sofer, A.; Sgroi, D.; Ellisen, L.W. Hypoxia regulates TSC1/2- mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling. Genes Dev. 2008, 22, 239–251, doi:10.1101/gad.1617608.
[69]  Xiang, X.; Yuan, M.; Song, Y.; Ruderman, N.; Wen, R.; Luo, Z. 14-3-3 facilitates insulin-stimulated intracellular trafficking of insulin receptor substrate 1. Mol. Endocrinol. 2002, 16, 552–562, doi:10.1210/me.16.3.552.
[70]  McKay, M.M.; Morrison, D.K. Integrating signals from RTKs to ERK/MAPK. Oncogene 2007, 26, 3113–3121, doi:10.1038/sj.onc.1210394.
[71]  Murphy, L.O.; Blenis, J. MAPK signal specificity: the right place at the right time. Trends Biochem. Sci. 2006, 31, 268–275, doi:10.1016/j.tibs.2006.03.009.
[72]  Cagnol, S.; Chambard, J.C. 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.
[73]  Ellington, A.A.; Berhow, M.A.; Singletary, K.W. Inhibition of Akt signaling and enhanced ERK1/2 activity are involved in induction of macroautophagy by triterpenoid B-group soyasaponins in colon cancer cells. Carcinogenesis 2006, 27, 298–306, doi:10.1093/carcin/bgi214.
[74]  Choi, C.H.; Jung, Y.K.; Oh, S.H. Autophagy induction by capsaicin in malignant human breast cells is modulated by p38 and extracellular signal-regulated mitogen-activated protein kinases and retards cell death by suppressing endoplasmic reticulum stress-mediated apoptosis. Mol. Pharmacol. 2010, 78, 114–125, doi:10.1124/mol.110.063495.
[75]  Wang, S.H.; Shih, Y.L.; Lee, C.C.; Chen, W.L.; Lin, C.J.; Lin, Y.S.; Wu, K.H.; Shih, C.M. The role of endoplasmic reticulum in cadmium-induced mesangial cell apoptosis. Chem. Biol. Interact. 2009, 181, 45–51, doi:10.1016/j.cbi.2009.05.004.
[76]  Yang, L.Y.; Wu, K.H.; Chiu, W.T.; Wang, S.H.; Shih, C.M. The cadmium-induced death of mesangial cells results in nephrotoxicity. Autophagy 2009, 5, 571–572, doi:10.4161/auto.5.4.8311.
[77]  Cheng, Y.; Qiu, F.; Tashiro, S.; Onodera, S.; Ikejima, T. ERK and JNK mediate TNFalpha- induced p53 activation in apoptotic and autophagic L929 cell death. Biochem. Biophys. Res.Commun. 2008, 376, 483–488, doi:10.1016/j.bbrc.2008.09.018.
[78]  Wang, J.; Whiteman, M.W.; Lian, H.; Wang, G.; Singh, A.; Huang, D.; Denmark, T. A non-canonical MEK/ERK signaling pathway regulates autophagy via regulating Beclin 1. J. Biol.Chem. 2009, 284, 21412–21424, doi:10.1074/jbc.M109.026013.
[79]  Corcelle, E.; Nebout, M.; Bekri, S.; Gauthier, N.; Hofman, P.; Poujeol, P.; Fenichel, P.; Mograbi, B. Disruption of autophagy at the maturation step by the carcinogen lindane is associated with the sustained mitogen-activated protein kinase/extracellular signal-regulated kinase activity. Cancer Res. 2006, 66, 6861–6870.
[80]  Tzivion, G.; Luo, Z.; Avruch, J. A dimeric 14-3-3 protein is an essential cofactor for Raf kinase activity. Nature 1998, 394, 88–92, doi:10.1038/27938.
[81]  Dhillon, A.S.; Meikle, S.; Peyssonnaux, C.; Grindlay, J.; Kaiser, C.; Steen, H.; Shaw, P.E.; Mischak, H.; Eychene, A.; Kolch, W. A Raf-1 mutant that dissociates MEK/extracellular signal-regulated kinase activation from malignant transformation and differentiation but not proliferation. Mol. Cell Biol. 2003, 23, 1983–1993.
[82]  Dumaz, N.; Marais, R. Protein kinase A blocks Raf-1 activity by stimulating 14-3-3 binding and blocking Raf-1 interaction with Ras. J. Biol. Chem. 2003, 278, 29819–29823, doi:10.1074/jbc.C300182200.
[83]  Wagner, E.F.; Nebreda, A.R. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat. Rev. Cancer 2009, 9, 537–549, doi:10.1038/nrc2694.
[84]  Webber, J.L.; Tooze, S.A. New insights into the function of Atg9. FEBS Lett. 2010, 584, 1319–1326, doi:10.1016/j.febslet.2010.01.020.
[85]  Cui, Q.; Tashiro, S.; Onodera, S.; Minami, M.; Ikejima, T. Oridonin induced autophagy in human cervical carcinoma HeLa cells through Ras, JNK, and P38 regulatio. J. Pharmacol. Sci. 2007, 105, 317–325, doi:10.1254/jphs.FP0070336.
[86]  Liao, P.C.; Ng, L.T.; Lin, L.T.; Richardson, C.D.; Wang, G.H.; Lin, C.C. Resveratrolarrests cell cycle and induces apoptosis in human hepatocellular carcinoma Huh-7 cells. J. Med. Food 2010, 13, 1415–1423, doi:10.1089/jmf.2010.1126.
[87]  Kim, D.S.; Kim, J.H.; Lee, G.H.; Kim, H.T.; Lim, J.M.; Chae, S.W.; Chae, H.J.; Kim, H.R. p38 Mitogen-activated protein kinase is involved in endoplasmic reticulum stress-induced cell death and autophagy in human gingival fibroblasts. Biol. Pharm. Bull. 2010, 33, 545–549, doi:10.1248/bpb.33.545.
[88]  Lim, S.C.; Hahm, K.S.; Lee, S.H.; Oh, S.H. Autophagy involvement in cadmium resistance through induction of multidrug resistance-associated protein and counterbalance of endoplasmic reticulum stress WI38 lung epithelial fibroblast cells. Toxicology 2010, 276, 18–26, doi:10.1016/j.tox.2010.06.010.
[89]  Thyagarajan, A.; Jedinak, A.; Nguyen, H.; Terry, C.; Baldridge, L.A.; Jiang, J.; Sliva, D. Triterpenes from Ganoderma Lucidum induce autophagy in colon cancer through the inhibition of p38 mitogen-activated kinase (p38 MAPK). Nutr. Cancer 2010, 62, 630–640, doi:10.1080/01635580903532390.
[90]  Colosetti, P.; Puissant, A.; Robert, G.; Luciano, F.; Jacquel, A.; Gounon, P.; Cassuto, J.P.; Auberger, P. Autophagy is an important event for megakaryocytic differentiation of the chronic myelogenous leukemia K562 cell line. Autophagy 2009, 5, 1092–1098, doi:10.4161/auto.5.8.9889.
[91]  Webber, J.L.; Tooze, S.A. Coordinated regulation of autophagy by p38alpha MAPK through mAtg9 and p38IP. Embo J. 2010, 29, 27–40, doi:10.1038/emboj.2009.321.
[92]  Fritz, A.; Brayer, K.J.; McCormick, N.; Adams, D.G.; Wadzinski, B.E.; Vaillancourt, R.R. Phosphorylation of serine 526 is required for MEKK3 activity, and association with 14-3-3 blocks dephosphorylation. J. Biol. Chem. 2006, 281, 6236–6245.
[93]  Xing, H.; Zhang, S.; Weinheimer, C.; Kovacs, A.; Muslin, A.J. 14-3-3 proteins block apoptosis and differentially regulate MAPK cascades. Embo J. 2000, 19, 349–358, doi:10.1093/emboj/19.3.349.
[94]  Basu, A. The potential of protein kinase C as a target for anticancer treatment. Pharmacol.Ther. 1993, 59, 257–280, doi:10.1016/0163-7258(93)90070-T.
[95]  Ozpolat, B.; Akar, U.; Mehta, K.; Lopez-Berestein, G. PKC delta and tissue transglutaminase are novel inhibitors of autophagy in pancreatic cancer cells. Autophagy 2007, 3, 480–483.
[96]  Lorand, L.; Graham, R.M. Transglutaminases: crosslinking enzymes with pleiotropic functions. Nat. Rev. Mol. Cell Biol. 2003, 4, 140–156, doi:10.1038/nrm1014.
[97]  Chen, J.L.; Lin, H.H.; Kim, K.J.; Lin, A.; Forman, H.J.; Ann, D.K. Novel roles for protein kinase Cdelta-dependent signaling pathways in acute hypoxic stress-induced autophagy. J. Biol. Chem. 2008, 283, 34432–34444.
[98]  Sakaki, K.; Wu, J.; Kaufman, R.J. Protein kinase Ctheta is required for autophagy in response to stress in the endoplasmic reticulum. J. Biol. Chem. 2008, 283, 15370–15380.
[99]  Jiang, H.; Cheng, D.; Liu, W.; Peng, J.; Feng, J. Protein kinase C inhibits autophagy andphosphorylates LC3. Biochem. Biophys. Res. Commun. 2010, 395, 471–476, doi:10.1016/j.bbrc.2010.04.030.
[100]  Acs, P.; Szallasi, Z.; Kazanietz, M.G.; Blumberg, P.M. Differential activation of PKC isozymes by 14-3-3 zeta protein. Biochem. Biophys. Res. Commun. 1995, 216, 103–109, doi:10.1006/bbrc.1995.2597.
[101]  Oriente, F.; Andreozzi, F.; Romano, C.; Perruolo, G.; Perfetti, A.; Fiory, F.; Miele, C.; Beguinot, F.; Formisano, P. Protein kinase C-alpha regulates insulin action and degradation by interacting with insulin receptor substrate-1 and 14-3-3 epsilon. J. Biol. Chem. 2005, 280, 40642–40649.
[102]  Kim, Y.H.; Kim, Y.S.; Kang, S.S.; Noh, H.S.; Kim, H.J.; Cho, G.J.; Choi, W.S. Expression of 14-3-3 zeta and interaction with protein kinase C in the rat retina in early diabetes. Diabetologia 2005, 48, 1411–1415, doi:10.1007/s00125-005-1774-7.
[103]  Dai, J.G.; Murakami, K. Constitutively and autonomously active protein kinase C associated with 14-3-3 zeta in the rodent brain. J. Neurochem. 2003, 84, 23–34.
[104]  Hausser, A.; Storz, P.; Link, G.; Stoll, H.; Liu, Y.C.; Altman, A.; Pfizenmaier, K.; Johannes, F.J. Protein kinase C mu is negatively regulated by 14-3-3 signal transduction proteins. J. Biol. Chem. 1999, 274, 9258–9264.
[105]  Meller, N.; Liu, Y.C.; Collins, T.L.; Bonnefoy-Berard, N.; Baier, G.; Isakov, N.; Altman, A. Direct interaction between protein kinase C theta (PKC theta) and 14-3-3 tau in T cells: 14-3-3 overexpression results in inhibition of PKC theta translocation and function. Mol. Cell Biol. 1996, 16, 5782–5791.
[106]  Robinson, K.; Jones, D.; Patel, Y.; Martin, H.; Madrazo, J.; Martin, S.; Howell, S.; Elmore, M.; Finnen, M.J.; Aitken, A. Mechanism of inhibition of protein kinase C by 14-3-3 isoforms. 14- 3–3 isoforms do not have phospholipase A2 activity. Biochem. J. 1994, 29, 853–861.
[107]  Van Der Hoeven, P.C.; Van Der Wal, J.C.; Ruurs, P.; Van Blitterswijk, W.J. Protein kinase C activation by acidic proteins including 14-3-3. Biochem. J. 2000, 347, 781–785, doi:10.1042/0264-6021:3470781.
[108]  Tanji, M.; Horwitz, R.; Rosenfeld, G.; Waymire, J.C. Activation of protein kinase C by purified bovine brain 14-3-3: comparison with tyrosine hydroxylase activation. J. Neurochem. 1994, 63, 1908–1916.
[109]  Hoyer-Hansen, M.; Jaattela, M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ. 2007, 14, 1576–1582, doi:10.1038/sj.cdd.4402200.
[110]  Davare, M.A.; Saneyoshi, T.; Guire, E.S.; Nygaard, S.C.; Soderling, T.R. Inhibition of calcium/calmodulin-dependent protein kinase kinase by protein 14-3-3. J. Biol. Chem. 2004, 279, 52191–52199.
[111]  Tsukada, M.; Ohsumi, Y. Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS Lett. 1993, 333, 169–174, doi:10.1016/0014-5793(93)80398-E.
[112]  Kamada, Y.; Funakoshi, T.; Shintani, T.; Nagano, K.; Ohsumi, M.; Ohsumi, Y. Tor-mediated induction of autophagy via an Apg1 protein kinase complex. J. Cell Biol. 2000, 150, 1507–1513, doi:10.1083/jcb.150.6.1507.
[113]  Kabeya, Y.; Kamada, Y.; Baba, M.; Takikawa, H.; Sasaki, M.; Ohsumi, Y. Atg17 functions in cooperation with Atg1 and Atg13 in yeast autophagy. Mol. Biol. Cell 2005, 16, 2544–2553, doi:10.1091/mbc.E04-08-0669.
[114]  Kamada, Y.; Yoshino, K.; Kondo, C.; Kawamata, T.; Oshiro, N.; Yonezawa, K.; Ohsumi, Y. Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol. Cell Biol. 2010, 30, 1049–1058, doi:10.1128/MCB.01344-09.
[115]  Kuroyanagi, H.; Yan, J.; Seki, N.; Yamanouchi, Y.; Suzuki, Y.; Takano, T.; Muramatsu, M.; Shirasawa, T. Human ULK1, a novel serine/threonine kinase related to UNC-51 kinase of Caenorhabditis elegans: cDNA cloning, expression, and chromosomal assignmet. Genomics 1998, 51, 76–85, doi:10.1006/geno.1998.5340.
[116]  Chan, E.Y.; Kir, S.; Tooze, S.A. siRNA screening of the kinome identifies ULK1 as a multidomain modulator of autophagy. J. Biol. Chem. 2007, 282, 25464–25474, doi:10.1074/jbc.M703663200.
[117]  Hara, T.; Takamura, A.; Kishi, C.; Iemura, S.; Natsume, T.; Guan, J.L.; Mizushima, N. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J. Cell Biol. 2008, 181, 497–510, doi:10.1083/jcb.200712064.
[118]  Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141, doi:10.1038/ncb2152.
[119]  Lee, J.W.; Park, S.; Takahashi, Y.; Wang, H.G. The association of AMPK with ULK1 regulates autophagy. PLoS One 2010, e15394, 1–9.
[120]  Cantley, L.C. The phosphoinositide 3-kinase pathway. Science 2002, 296, 1655–1657, doi:10.1126/science.296.5573.1655.
[121]  Lawlor, M.A.; Alessi, D.R. PKB/Akt: A key mediator of cell proliferation, survival and insulin responses? J. Cell Sci. 2001, 114, 2903–2910.
[122]  Wang, Y.; Yoshioka, K.; Azam, M.A.; Takuwa, N.; Sakurada, S.; Kayaba, Y.; Sugimoto, N.; Inoki, I.; Kimura, T.; Kuwaki, T.; Takuwa, Y. Class II phosphoinositide 3-kinase alpha-isoform regulates Rho, myosin phosphatase and contraction in vascular smooth muscle. Biochem. J. 2006, 394, 581–592, doi:10.1042/BJ20051471.
[123]  Domin, J.; Harper, L.; Aubyn, D.; Wheeler, M.; Florey, O.; Haskard, D.; Yuan, M.; Zicha, D. The class II phosphoinositide 3-kinase PI3K-C2beta regulates cell migration by a PtdIns3P dependent mechanism. J. Cell Physiol. 2005, 205, 452–462, doi:10.1002/jcp.20478.
[124]  Backer, J.M. The regulation and function of Class III PI3Ks: novel roles for Vps34. Biochem. J. 2008, 410, 1–17, doi:10.1042/BJ20071427.
[125]  Proikas-Cezanne, T.; Waddell, S.; Gaugel, A.; Frickey, T.; Lupas, A.; Nordheim, A. WIPI- 1alpha (WIPI49), a member of the novel 7-bladed WIPI protein family, is aberrantly expressed in human cancer and is linked to starvation-induced autophagy. Oncogene 2004, 23, 9314–9325, doi:10.1038/sj.onc.1208331.
[126]  Polson, H.E.; de Lartigue, J.; Rigden, D.J.; Reedijk, M.; Urbe, S.; Clague, M.J.; Tooze, S.A. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy 2010, 6.
[127]  Krick, R.; Tolstrup, J.; Appelles, A.; Henke, S.; Thumm, M. The relevance of the phosphatidylinositolphosphat-binding motif FRRGT of Atg18 and Atg21 for the Cvt pathway and autophagy. FEBS Lett. 2006, 580, 4632–4638, doi:10.1016/j.febslet.2006.07.041.
[128]  Ghosh, P.; Wu, M.; Zhang, H.; Sun, H. mTORC1 signaling requires proteasomal function and the involvement of CUL4-DDB1 ubiquitin E3 ligase. Cell Cycle 2008, 7, 373–381, doi:10.4161/cc.7.3.5267.
[129]  Zhong, Y.; Wang, Q.J.; Li, X.; Yan, Y.; Backer, J.M.; Chait, B.T.; Heintz, N.; Yue, Z. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1- phosphatidylinositol-3-kinase complex. Nat. Cell Biol. 2009, 11, 468–476, doi:10.1038/ncb1854.
[130]  Pozuelo-Rubio, M. Regulation of autophagic activity by 14-3-3zeta proteins associated with class III phosphatidylinositol-3-kinase. Cell Death Differ. 2011, 18, 479–492, doi:10.1038/cdd.2010.118.
[131]  Wang, B.; Lin, S.; Lin, W.-C. 14-3-3tau regulates Beclin 1 and is required for autophagy. 2010, 5, e10409, 1–10.
[132]  Pattingre, S.; Tassa, A.; Qu, X.; Garuti, R.; Liang, X.H.; Mizushima, N.; Packer, M.; Schneider, M.D.; Levine, B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122, 927–939, doi:10.1016/j.cell.2005.07.002.
[133]  Lee, J.S.; Li, Q.; Lee, J.Y.; Lee, S.H.; Jeong, J.H.; Lee, H.R.; Chang, H.; Zhou, F.C.; Gao, S.J.; Liang, C.; Jung, J.U. FLIP-mediated autophagy regulation in cell death control. Nat. Cell. Biol. 2009, 11, 1355–1362, doi:10.1038/ncb1980.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413