Studies on the cytoprotective role of autophagy in necrosis

229 346 0
Studies on the cytoprotective role of autophagy in necrosis

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

STUDIES ON THE CYTOPROTECTIVE ROLE OF AUTOPHAGY IN NECROSIS WU YOUTONG (M. Med, Huazhong Univ Sci & Tech, P. R. CHINA) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF EPIDEMIOLOGY AND PUBLIC HEALTH NATIONAL UNIVERSITY OF SINGAPORE 2010 ACKNOWLEDGEMENTS I would like to dedicate my sincere and deep gratitude to my supervisor, Associate Professor Shen Han-Ming, for his enthusiastic professional guidance. Prof. Shen has led me into the vast world of biological research and has guided me with inspirations during the tough, yet exciting, journey throughout this study. Besides acting as a mentor, Prof. Shen has also treated me like a friend, sharing with me of his invaluable experience in life philosophy, communication skills, and even protocols for delicious cooking. All of these will be appreciated in my whole life. I would also like to express my sincere thanks to Prof. Ong Choon Nam, who offered me the precious opportunity for working in this lab in Singapore, which fueled my dream to pursue this Ph.D degree. It has been a great pleasure for me to work in the big family of Department of Epidemiology and Public Health in the last four years. All people in the lab are always kind and helpful. I would like to list the members of this big family with honor and gratitude: Mr. Ong Her Yam, Dr. Zhang Siyuan, Dr. Huang Qing, Dr. Lu Guo-Dong, Dr. Zhou Jing, Ms. Su Jin, and Mr. Ong Yeong Bing. Finally, I would like to express my deep appreciation to my wife, Ms Tan Hui-Ling. Without her contribution of diligent and elegant works to this study and her dedicated love and understanding, this thesis would not be possible. II TABLE OF CONTENTS ACKNOWLEDGEMENTS II TABLE OF CONTENTS III SUMMARY VI LIST OF TABLES IX LIST OF FIGURES X LIST OF ABBREVIATIONS XII LIST OF PUBLICATIONS XV CHAPTER INTRODUCTION 1.1 PROGRAMMED CELL DEATH: APOPTOSIS VERSUS NECROSIS 1.1.1 APOPTOSIS 1.1.2 NECROSIS 1.1.3 CROSSTALK BETWEEN APOPTOSIS AND NECROSIS 1.2 AUTOPHAGY 1.2.1 GENERAL INTRODUCTION 1.2.2 DYNAMIC PROCESS OF AUTOPHAGY 1.2.3 MACHINERY FOR AUTOPHAGOSOME FORMATION 1.2.4 REGULATORY PATHWAYS 1.2.5 AUTOPHAGY FUNCTIONS AND IMPLICATIONS IN DISEASES 1.3 CROSSTALK BETWEEN AUTOPHAGY AND PCD 1.3.1 AUTOPHAGY IN APOPTOSIS 1.3.2 AUTOPHAGY IN NECROSIS 1.4 OBJECTIVES 14 19 21 21 22 24 31 42 46 47 53 60 CHAPTER AUTOPHAGY HAS A PROTECTIVE ROLE DURING ZVAD-INDUCED NECROTIC CELL DEATH 62 2.1 INTRODUCTION 2.2 MATERIALS AND METHODS 2.2.1 REAGENTS AND ANTIBODIES 2.2.2 CELL CULTURE AND TREATMENTS 2.2.3 DETECTION OF CELL DEATH 2.2.4 TRANSIENT TRANSFECTION AND CONFOCAL MICROSCOPY ANALYSIS 2.2.5 ELECTRON MICROSCOPY ANALYSIS 63 65 65 66 66 66 67 III 2.2.6 SIRNA 67 2.2.7 MEASUREMENT OF CATHEPSIN B ACTIVITY 67 2.2.8 WESTERN BLOTTING 68 2.3 RESULTS 69 2.3.1 ZVAD INDUCES CASPASE-INDEPENDENT NON-APOPTOTIC CELL DEATH WITH THE PRESENCE OF AUTOPHAGY MARKERS IN L929 CELLS 69 2.3.2 OPPOSITE EFFECTS OF RAPAMYCIN AND CHLOROQUINE ON ZVAD-INDUCED CELL DEATH 70 2.3.3 OPPOSITE EFFECT OF SERUM STARVATION AND ATG GENE KNOCKDOWN ON ZVAD-INDUCED CELL DEATH 71 2.3.4 ZVAD SUPPRESSES LYSOSOMAL FUNCTION VIA ITS INHIBITORY EFFECT ON CATHEPSIN ACTIVITY 72 2.3.5 ZVAD INHIBITS AUTOPHAGOSOME MATURATION 73 2.4 DISCUSSION 74 CHAPTER ACTIVATION OF THE PI3K-AKT-MTOR SIGNALING PATHWAY BY INSULIN PROMOTES NECROTIC CELL DEATH VIA SUPPRESSION OF AUTOPHAGY 91 3.1 INTRODUCTION 3.2 MATERIALS AND METHODS 3.2.1 REAGENTS AND ANTIBODIES 3.2.2 CELL CULTURE 3.2.3 DETECTION OF CELL DEATH/CELL VIABILITY 3.2.4 PLASMIDS AND STABLE TRANSFECTION 3.2.5 CONFOCAL MICROSCOPY 3.2.6 SMALL INTERFERING RNA 3.2.7 WESTERN BLOTTING 3.3 RESULTS 3.3.1 INSULIN PROMOTES CELL DEATH IN NECROTIC CELL DEATH MODELS 3.3.2 INSULIN ABOLISHES THE PROTECTIVE EFFECT OF STARVATION ON NECROTIC 92 94 94 95 95 95 96 96 96 97 97 98 99 CELL DEATH 3.3.3 IGF-1, BUT NOT EGF, HAS A SIMILAR PRO-DEATH EFFECT AS INSULIN 3.3.4 INHIBITION OF PI3K-AKT-MTOR SIGNALING PATHWAY BY CHEMICAL INHIBITORS ABOLISHES THE PRO-DEATH EFFECT OF INSULIN 3.3.5 KNOCKDOWN OF MTOR MITIGATES THE PRO-DEATH EFFECT OF INSULIN 3.3.6 INSULIN INHIBITS AUTOPHAGY INDUCED BY STARVATION 3.4 DISCUSSION 100 101 102 103 CHAPTER ZVAD-INDUCED NECROPTOSIS IN L929 CELLS DEPENDS ON AUTOCRINE PRODUCTION OF TNFΑ MEDIATED VIA THE PKC-MAPKS-AP-1 PATHWAY 119 4.1 INTRODUCTION 120 IV 4.2 MATERIALS AND METHOD 122 4.2.1 REAGENTS AND ANTIBODIES 122 4.2.2 CELL CULTURE 123 4.2.3 DETECTION OF CELL DEATH 123 4.2.4 SIRNA 123 4.2.5 TRANSFECTION AND LUCIFERASE REPORTER ASSAY 124 4.2.6 REVERSE TRANSCRIPTION-PCR 124 4.2.7 MEASUREMENT OF AUTOCRINE TNFΑ IN CULTURE MEDIUM BY ELISA 125 4.2.8 WESTERN BLOTTING 125 4.3 RESULTS 125 4.3.1 ZVAD AND BOCD, BUT NOT QVD, INDUCE NECROSIS IN L929 CELLS 125 4.3.2 ZVAD-INDUCED NECROTIC CELL DEATH REQUIRES DE NOVO PROTEIN SYNTHESIS 127 4.3.3 ZVAD-INDUCED CELL DEATH IS RIP1- AND RIP3-DEPENDENT 127 4.3.4 ZVAD PROMOTES AUTOCRINE PRODUCTION OF TNFΑ 128 4.3.5 BLOCKAGE OF TNFΑ SIGNALING SUPPRESSES ZVAD-INDUCED NECROPTOSIS 129 4.3.6 NF-ΚB PATHWAY IS NOT INVOLVED IN ZVAD-INDUCED AUTOCRINE PRODUCTION OF TNFΑ, BUT PLAYS A PROTECTIVE ROLE DURING ZVAD-INDUCED NECROPTOSIS 130 4.3.7 AP-1 ACTIVITY IS REQUIRED FOR ZVAD-INDUCED TNFΑ PRODUCTION AND CELL DEATH 131 4.3.8 ZVAD-INDUCED AP-1 ACTIVATION IS MEDIATED BY JNK AND ERK 132 4.3.9 PKC PLAYS A CRITICAL ROLE IN ZVAD-MEDIATED MAPKS-AP1 ACTIVATION, TNFΑ PRODUCTION, AND CELL DEATH 133 4.3.10 ZVAD SENSITIZES TNFΑ-INDUCED NECROPTOSIS IN L929 CELLS 135 4.3.11 DEFECT IN AUTOPHAGY ENHANCES AP-1 ACTIVITY 135 4.4 DISCUSSION 137 CHAPTER GENERAL DISCUSSIONS AND CONCLUSIONS 163 5.1 AUTOPHAGY PLAYS A PRO-SURVIVAL ROLE IN ZVAD-INDUCED NECROSIS 5.2 ZVAD INHIBITS AUTOPHAGY VIA SUPPRESSION OF CATHEPSIN ACTIVITY 5.3 SUPPRESSION OF AUTOPHAGY BY ACTIVATION OF PI3K-AKT-MTOR AXIS 165 166 5.4 AUTOCRINE TNFΑ IS THE DEATH SIGNAL IN ZVAD-INDUCED NECROSIS 5.5 DUAL ROLE OF ZVAD DURING INDUCTION OF NECROPTOSIS 5.6 THE MECHANISMS FOR AUTOPHAGY TO PROTECT NECROSIS 5.7 CONCLUSIONS 168 170 173 176 178 CHAPTER REFERENCES 181 PROMOTES NECROSIS V SUMMARY Programmed cell death (PCD) is an intrinsically regulated cellular suicide process that can be categorized into apoptosis and necrosis based on their distinct morphological characteristics. Autophagy refers to an evolutionarily conserved process that sequesters and targets bulk cellular constituents for lysosomal degradation. Autophagy has been found to be implicated in regulation of PCD under various cellular settings. At present, the role of autophagy on PCD is highly controversial. Although autophagy generally serves as a cell survival mechanism under stress conditions such as starvation, there are reports showing that autophagy executes caspase-independent cell death, known as autophagic cell death. However, in many cases the evidence supporting autophagy as a cell death mechanism is frequently circumstantial and appears inadequate. zVAD, a pan-caspase inhibitor, has been shown to induce robust necrosis in L929 cells, and such necrosis has been reported as autophagic cell death. However, the molecular mechanism underlying such cell death has not been fully elucidated. Therefore, the main objective of this study is to investigate the regulatory role of autophagy in necrosis and to elucidate the underlying molecular mechanisms using in vitro mammalian cell models. The following investigations have been conducted: (i) examining the role of autophagy in zVAD-induced necrosis by modulation of autophagy via either pharmacological or genetic approaches; (ii) studying the regulatory role of class I PI3K-Akt-mTOR signaling axis in modulation of autophagy and necrosis; and (iii) elucidating the molecular mechanism underlying zVAD-induced necrosis. VI In this study, we first demonstrated that autophagy played a cytoprotective role during zVAD-induced necrosis. Moreover, zVAD was able to suppress autophagy via suppression of lysosome function via inhibition of cathepsin enzyme activity. One surprising finding of this study was that growth factors such as insulin and IGF-1 and nutrients such as amino acids were able to enhance zVAD-induced necrosis via activation of the PI3K-Akt-mTOR pathway and subsequent suppression of autophagy. Moreover, the pro-death function of insulin/amino acids was also observed in other two necrosis models, including MNNG-induced necrosis in L929 cells and H2O2-induced necrosis in Bax/Bak double knockout cells, where autophagy acted as a pro-survival mechanism. Finally, we identified that zVAD-induced necrosis was RIP1- and RIP3-mediated necroptosis that depended on the autocrine production of TNFα. zVAD promoted the autocrine production of TNFα at the transcription level, which was required for induction of cell death. We also demonstrated that zVAD promoted TNFα production via the PKC-MAPKs-AP-1 pathway. Moreover, we presented evidence showing that defects in autophagy might promote zVAD-induced cell death by enhancing AP-1 activity. In conclusion, data from this study demonstrate that (i) autophagy plays a cell survival strategy in the three necrosis models tested in this study; (ii) growth factors and amino acids promote necrosis in these models via activation of the PI3K-Akt-mTOR pathway and subsequent suppression of autophagy; and (iii) zVAD-induced necroptosis depends on autocrine production of TNFα that is mediated via the PKC-MAPKs-AP-1 signaling pathway. Taken together, results from the VII above-described studies provide novel insights for a better understanding of the role of autophagy in necrosis. VIII LIST OF ABBREVIATIONS 3-MA 4EBP1 ActD AIF AMPK ANT Apaf-1 Atg ATP BAFF Bak Bax Bcl-2 Beclin BH BNIP3 BocD-fmk CARD CHX CNS CQ DAPK DED DEVD-cho DISCs DR DRAM DUB EAA EBSS EGF EGFP eIF2α ER ERK FADD FBS FIP200 fmk FoxO FoxOs GAPDH 3-methyladenine eukaryotic initiation factor 4E binding protein actinomycin D apoptosis-inducing factor AMP-activated protein kinase adenine nucleotide translocase apoptotic peptidase activating factor-1 autophagy-related adenosine triphosphate B-cell activating factor Bcl-2 homologous antagonist Bcl-2 associated X protein B-cell lymphoma-2 coiled-coil, myosin-like BCL2 interacting protein Bcl-2 homology BCL2 and adenovirus E1B 19 kDa-interacting protein Boc-Asp(Ome)-fmk caspase recruitment domain cycloheximide central nervous system chloroquine death-associated protein kinase death effector domain Asp-Glu-Val-Asp-cho death-inducing signaling complexes death receptor damage-regulated autophagy modulator deubiquitinating enzyme essential amino acid earle’s balanced salt solution epidermal growth factor green fluorescent protein eucaryotic translation initiation factor 2α endoplasmic reticulum extracellular signal-regulated kinases Fas-associated death domain fetal bovine serum focal adhesion kinase family interacting protein of 200 Kd fluoromethylketone forkhead box O forkhead box O transcription factors glyceraldehyde-3-phosphate dehydrogenase IX GDP GLUD1 GLUL GTP HIF1 hVps IAP ICAD IETD-fmk IETD-oph IGF-1 IKK IL IκB JNK KO LC3 LPS MAPK Mcl-1 Mdm MEF MNNG MOMP mRFP mTOR mTORC1 mTORC2 MTT NAD NADPH NF-κB NIK Nox1 oph PAR PARP PAS PCD PDK1 PE PERK PH PI PI3K Guanosine diposphate glutamate dehydrogenase glutamate ammoniavligase Guanosine triposphate hypoxia-induced factor human vacuolar protein sorting inhibitor of apoptosis inhibitor of caspase activated DNase or DNA fragmentation factor Z-Ile-Glu(OMe)-Thr-Asp(OMe)-fmk Z-Ile-Glu(OMe)-Thr-Asp(OMe)-oph insulin-like growth factor-1 IκB kinase interleukin Inhibitor of κB c-Jun N-terminal kinase knockout rat microtubule-associated protein light chain lipopolysaccharide mitogen-activated protein kinase Myeloid cell leukemia sequence-1 murine double minute mouse embryonic fibroblast Methylnitronitrosoguanidine mitochondrial outer membrane permeabilization monomeric red fluorescent protein mammalian target of rapamycin mTOR complex mTOR complex 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide β-nicotinamide adenine dinucleotide nicotinamide adenine dinucleotide phosphate hydrogen nuclear factor kappa-light-chain-enhancer of activated B cells NF-κB-inducing kinase NADPH oxidase 2,6-difluorophenoxy methyl Ketone poly(ADP-ribose) poly(ADP-ribose) polymerase pre-autophagosome structure programmed cell death phosphoinositide-dependent kinase phosphatidylethanolamine PKR-like ER kinase pleckstrin homology propidium iodide phosphoinositide-3 kinase X Liang, C., Feng, P., Ku, B., Dotan, I., Canaani, D., Oh, B.H., and Jung, J.U. (2006). Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat Cell Biol 8, 688-699. Liang, C., Lee, J.S., Inn, K.S., Gack, M.U., Li, Q., Roberts, E.A., Vergne, I., Deretic, V., Feng, P., Akazawa, C., et al. (2008). Beclin1-binding UVRAG targets the class C Vps complex to coordinate autophagosome maturation and endocytic trafficking. Nat Cell Biol 10, 776-787. Liang, X.H., Jackson, S., Seaman, M., Brown, K., Kempkes, B., Hibshoosh, H., and Levine, B. (1999). Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402, 672-676. Liang, X.H., Kleeman, L.K., Jiang, H.H., Gordon, G., Goldman, J.E., Berry, G., Herman, B., and Levine, B. (1998). Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol 72, 8586-8596. Lin, Y., Choksi, S., Shen, H.M., Yang, Q.F., Hur, G.M., Kim, Y.S., Tran, J.H., Nedospasov, S.A., and Liu, Z.G. (2004). Tumor necrosis factor-induced nonapoptotic cell death requires receptor-interacting protein-mediated cellular reactive oxygen species accumulation. J Biol Chem 279, 10822-10828. Lin, Y., Devin, A., Rodriguez, Y., and Liu, Z.G. (1999). Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev 13, 2514-2526. Lindsten, T., Golden, J.A., Zong, W.X., Minarcik, J., Harris, M.H., and Thompson, C.B. (2003). The proapoptotic activities of Bax and Bak limit the size of the neural stem cell pool. J Neurosci 23, 11112-11119. Lockshin, R.A., and Williams, C.M. (1965). Programmed Cell Death--I. Cytology of Degeneration in the Intersegmental Muscles of the Pernyi Silkmoth. J Insect Physiol 11, 123-133. Locksley, R.M., Killeen, N., and Lenardo, M.J. (2001). The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104, 487-501. Long, X., Lin, Y., Ortiz-Vega, S., Yonezawa, K., and Avruch, J. (2005a). Rheb binds and regulates the mTOR kinase. Curr Biol 15, 702-713. Long, X., Ortiz-Vega, S., Lin, Y., and Avruch, J. (2005b). Rheb binding to mammalian target of rapamycin (mTOR) is regulated by amino acid sufficiency. J Biol Chem 280, 23433-23436. Lum, J.J., Bauer, D.E., Kong, M., Harris, M.H., Li, C., Lindsten, T., and Thompson, C.B. (2005a). Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 120, 237-248. 198 Lum, J.J., DeBerardinis, R.J., and Thompson, C.B. (2005b). Autophagy in metazoans: cell survival in the land of plenty. Nat Rev Mol Cell Biol 6, 439-448. Luo, X., Budihardjo, I., Zou, H., Slaughter, C., and Wang, X. (1998). Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 94, 481-490. Ma, L., Chen, Z., Erdjument-Bromage, H., Tempst, P., and Pandolfi, P.P. (2005). Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis. Cell 121, 179-193. Mackay, H.J., and Twelves, C.J. (2007). Targeting the protein kinase C family: are we there yet? Nat Rev Cancer 7, 554-562. Madden, D.T., Egger, L., and Bredesen, D.E. (2007). A calpain-like protease inhibits autophagic cell death. Autophagy 3, 519-522. Maiuri, M.C., Criollo, A., Tasdemir, E., Vicencio, J.M., Tajeddine, N., Hickman, J.A., Geneste, O., and Kroemer, G. (2007a). BH3-only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin and Bcl-2/Bcl-X(L). Autophagy 3, 374-376. Maiuri, M.C., Le Toumelin, G., Criollo, A., Rain, J.C., Gautier, F., Juin, P., Tasdemir, E., Pierron, G., Troulinaki, K., Tavernarakis, N., et al. (2007b). Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J 26, 2527-2539. Maiuri, M.C., Zalckvar, E., Kimchi, A., and Kroemer, G. (2007c). Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol 8, 741-752. Mammucari, C., Milan, G., Romanello, V., Masiero, E., Rudolf, R., Del Piccolo, P., Burden, S.J., Di Lisi, R., Sandri, C., Zhao, J., et al. (2007). FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6, 458-471. Manning, B.D., and Cantley, L.C. (2007). AKT/PKB signaling: navigating downstream. Cell 129, 1261-1274. Marino, G., Uria, J.A., Puente, X.S., Quesada, V., Bordallo, J., and Lopez-Otin, C. (2003). Human autophagins, a family of cysteine proteinases potentially implicated in cell degradation by autophagy. J Biol Chem 278, 3671-3678. Mathew, R., Karp, C.M., Beaudoin, B., Vuong, N., Chen, G., Chen, H.Y., Bray, K., Reddy, A., Bhanot, G., Gelinas, C., et al. (2009). Autophagy suppresses tumorigenesis through elimination of p62. Cell 137, 1062-1075. 199 Mathew, R., Kongara, S., Beaudoin, B., Karp, C.M., Bray, K., Degenhardt, K., Chen, G., Jin, S., and White, E. (2007). Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev 21, 1367-1381. Mathew, R., and White, E. (2007). Why sick cells produce tumors: the protective role of autophagy. Autophagy 3, 502-505. Matsumura, H., Shimizu, Y., Ohsawa, Y., Kawahara, A., Uchiyama, Y., and Nagata, S. (2000). Necrotic death pathway in Fas receptor signaling. J Cell Biol 151, 1247-1256. Matsunaga, K., Saitoh, T., Tabata, K., Omori, H., Satoh, T., Maejima, I., Shirahama-Noda, K., Ichimura, T., Isobe, T., Akira, S., et al. (2009). Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. Matsuura, A., Tsukada, M., Wada, Y., and Ohsumi, Y. (1997). Apg1p, a novel protein kinase required for the autophagic process in Saccharomyces cerevisiae. Gene 192, 245-250. Mayo, L.D., and Donner, D.B. (2001). A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci U S A 98, 11598-11603. McCubrey, J.A., Steelman, L.S., Chappell, W.H., Abrams, S.L., Wong, E.W., Chang, F., Lehmann, B., Terrian, D.M., Milella, M., Tafuri, A., et al. (2007). Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 1773, 1263-1284. Meijer, A.J., and Codogno, P. (2006). Signalling and autophagy regulation in health, aging and disease. Mol Aspects Med 27, 411-425. Meijer, A.J., and Codogno, P. (2009). Autophagy: regulation and role in disease. Crit Rev Clin Lab Sci 46, 210-240. Meijer, W.H., van der Klei, I.J., Veenhuis, M., and Kiel, J.A. (2007). ATG genes involved in non-selective autophagy are conserved from yeast to man, but the selective Cvt and pexophagy pathways also require organism-specific genes. Autophagy 3, 106-116. Michalak, E.M., Villunger, A., Adams, J.M., and Strasser, A. (2008). In several cell types tumour suppressor p53 induces apoptosis largely via Puma but Noxa can contribute. Cell Death Differ 15, 1019-1029. Micheau, O., and Tschopp, J. (2003). Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114, 181-190. Mizushima, N., Kuma, A., Kobayashi, Y., Yamamoto, A., Matsubae, M., Takao, 200 T., Natsume, T., Ohsumi, Y., and Yoshimori, T. (2003). Mouse Apg16L, a novel WD-repeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5 conjugate. J Cell Sci 116, 1679-1688. Mizushima, N., Levine, B., Cuervo, A.M., and Klionsky, D.J. (2008). Autophagy fights disease through cellular self-digestion. Nature 451, 1069-1075. Mizushima, N., Noda, T., Yoshimori, T., Tanaka, Y., Ishii, T., George, M.D., Klionsky, D.J., Ohsumi, M., and Ohsumi, Y. (1998a). A protein conjugation system essential for autophagy. Nature 395, 395-398. Mizushima, N., Sugita, H., Yoshimori, T., and Ohsumi, Y. (1998b). A new protein conjugation system in human. The counterpart of the yeast Apg12p conjugation system essential for autophagy. J Biol Chem 273, 33889-33892. Mizushima, N., Yamamoto, A., Hatano, M., Kobayashi, Y., Kabeya, Y., Suzuki, K., Tokuhisa, T., Ohsumi, Y., and Yoshimori, T. (2001). Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol 152, 657-668. Mizushima, N., and Yoshimori, T. (2007). How to Interpret LC3 Immunoblotting. Autophagy 3, 542-545. Mizushima, N., Yoshimori, T., and Levine, B. (2010). Methods in mammalian autophagy research. Cell 140, 313-326. Mizushima, N., Yoshimori, T., and Ohsumi, Y. (2002). Mouse Apg10 as an Apg12-conjugating enzyme: analysis by the conjugation-mediated yeast two-hybrid method. FEBS Lett 532, 450-454. Moretti, L., Kim, K.W., Jung, D.K., Willey, C.D., and Lu, B. (2009). Radiosensitization of solid tumors by Z-VAD, a pan-caspase inhibitor. Mol Cancer Ther. Nakano, K., and Vousden, K.H. (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7, 683-694. Newton, A.C. (1995). Protein kinase C: structure, function, and regulation. J Biol Chem 270, 28495-28498. Nicklin, P., Bergman, P., Zhang, B., Triantafellow, E., Wang, H., Nyfeler, B., Yang, H., Hild, M., Kung, C., Wilson, C., et al. (2009). Bidirectional Transport of Amino Acids Regulates mTOR and Autophagy. Cell 136, 521-534. Nobukuni, T., Joaquin, M., Roccio, M., Dann, S.G., Kim, S.Y., Gulati, P., Byfield, M.P., Backer, J.M., Natt, F., Bos, J.L., et al. (2005). Amino acids mediate mTOR/raptor signaling through activation of class 201 phosphatidylinositol 3OH-kinase. Proc Natl Acad Sci U S A 102, 14238-14243. Nussbaum, A.K., and Whitton, J.L. (2004). The contraction phase of virus-specific CD8+ T cells is unaffected by a pan-caspase inhibitor. J Immunol 173, 6611-6618. O'Donnell, M.A., Legarda-Addison, D., Skountzos, P., Yeh, W.C., and Ting, A.T. (2007). Ubiquitination of RIP1 regulates an NF-kappaB-independent cell-death switch in TNF signaling. Curr Biol 17, 418-424. Obara, K., Noda, T., Niimi, K., and Ohsumi, Y. (2008a). Transport of phosphatidylinositol 3-phosphate into the vacuole via autophagic membranes in Saccharomyces cerevisiae. Genes Cells 13, 537-547. Obara, K., Sekito, T., Niimi, K., and Ohsumi, Y. (2008b). The Atg18-Atg2 complex is recruited to autophagic membranes via phosphatidylinositol 3-phosphate and exerts an essential function. J Biol Chem 283, 23972-23980. Oda, E., Ohki, R., Murasawa, H., Nemoto, J., Shibue, T., Yamashita, T., Tokino, T., Taniguchi, T., and Tanaka, N. (2000a). Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053-1058. Oda, K., Arakawa, H., Tanaka, T., Matsuda, K., Tanikawa, C., Mori, T., Nishimori, H., Tamai, K., Tokino, T., Nakamura, Y., et al. (2000b). p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell 102, 849-862. Ogata, M., Hino, S., Saito, A., Morikawa, K., Kondo, S., Kanemoto, S., Murakami, T., Taniguchi, M., Tanii, I., Yoshinaga, K., et al. (2006). Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 26, 9220-9231. Ollinger, K., and Roberg, K. (1997). Nutrient deprivation of cultured rat hepatocytes increases the desferrioxamine-available iron pool and augments the sensitivity to hydrogen peroxide. J Biol Chem 272, 23707-23711. Oltersdorf, T., Elmore, S.W., Shoemaker, A.R., Armstrong, R.C., Augeri, D.J., Belli, B.A., Bruncko, M., Deckwerth, T.L., Dinges, J., Hajduk, P.J., et al. (2005). An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435, 677-681. Paglin, S., Hollister, T., Delohery, T., Hackett, N., McMahill, M., Sphicas, E., Domingo, D., and Yahalom, J. (2001). A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res 61, 439-444. 202 Pankiv, S., Clausen, T.H., Lamark, T., Brech, A., Bruun, J.A., Outzen, H., Overvatn, A., Bjorkoy, G., and Johansen, T. (2007). p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282, 24131-24145. Pattingre, S., Bauvy, C., Carpentier, S., Levade, T., Levine, B., and Codogno, P. (2009). Role of JNK1-dependent Bcl-2 Phosphorylation in Ceramide-induced Macroautophagy. J Biol Chem 284, 2719-2728. Pattingre, S., Tassa, A., Qu, X., Garuti, R., Liang, X.H., Mizushima, N., Packer, M., Schneider, M.D., and Levine, B. (2005). Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 122, 927-939. Petersen, S.L., Wang, L., Yalcin-Chin, A., Li, L., Peyton, M., Minna, J., Harran, P., and Wang, X. (2007). Autocrine TNFalpha signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis. Cancer Cell 12, 445-456. Petiot, A., Ogier-Denis, E., Blommaart, E.F., Meijer, A.J., and Codogno, P. (2000). Distinct classes of phosphatidylinositol 3'-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J Biol Chem 275, 992-998. Pickart, C.M. (2004). Back to the future with ubiquitin. Cell 116, 181-190. Pieper, A.A., Verma, A., Zhang, J., and Snyder, S.H. (1999). Poly (ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol Sci 20, 171-181. Pietsch, E.C., Sykes, S.M., McMahon, S.B., and Murphy, M.E. (2008). The p53 family and programmed cell death. Oncogene 27, 6507-6521. Proikas-Cezanne, T., Waddell, S., Gaugel, A., Frickey, T., Lupas, A., and Nordheim, A. (2004). 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 23, 9314-9325. Qu, X., Yu, J., Bhagat, G., Furuya, N., Hibshoosh, H., Troxel, A., Rosen, J., Eskelinen, E.L., Mizushima, N., Ohsumi, Y., et al. (2003). Promotion of tumorigenesis by heterozygous disruption of the beclin autophagy gene. J Clin Invest 112, 1809-1820. Qu, X., Zou, Z., Sun, Q., Luby-Phelps, K., Cheng, P., Hogan, R.N., Gilpin, C., and Levine, B. (2007). Autophagy gene-dependent clearance of apoptotic cells during embryonic development. Cell 128, 931-946. Ravi, R., Bedi, G.C., Engstrom, L.W., Zeng, Q., Mookerjee, B., Gelinas, C., Fuchs, E.J., and Bedi, A. (2001). Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-kappaB. Nat Cell Biol 3, 409-416. 203 Ravikumar, B., Berger, Z., Vacher, C., O'Kane, C.J., and Rubinsztein, D.C. (2006). Rapamycin pre-treatment protects against apoptosis. Hum Mol Genet 15, 1209-1216. Ray, C.A., Black, R.A., Kronheim, S.R., Greenstreet, T.A., Sleath, P.R., Salvesen, G.S., and Pickup, D.J. (1992). Viral inhibition of inflammation: cowpox virus encodes an inhibitor of the interleukin-1 beta converting enzyme. Cell 69, 597-604. Rayet, B., and Gelinas, C. (1999). Aberrant rel/nfkb genes and activity in human cancer. Oncogene 18, 6938-6947. Reggiori, F., Shintani, T., Nair, U., and Klionsky, D.J. (2005). Atg9 cycles between mitochondria and the pre-autophagosomal structure in yeasts. Autophagy 1, 101-109. Reggiori, F., Tucker, K.A., Stromhaug, P.E., and Klionsky, D.J. (2004). The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev Cell 6, 79-90. Riley, T., Sontag, E., Chen, P., and Levine, A. (2008). Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9, 402-412. Roisin-Bouffay, C., Luciani, M.F., Klein, G., Levraud, J.P., Adam, M., and Golstein, P. (2004). Developmental cell death in dictyostelium does not require paracaspase. J Biol Chem 279, 11489-11494. Rouschop, K.M., Ramaekers, C.H., Schaaf, M.B., Keulers, T.G., Savelkouls, K.G., Lambin, P., Koritzinsky, M., and Wouters, B.G. (2009). Autophagy is required during cycling hypoxia to lower production of reactive oxygen species. Radiother Oncol 92, 411-416. Saftig, P., Hetman, M., Schmahl, W., Weber, K., Heine, L., Mossmann, H., Koster, A., Hess, B., Evers, M., von Figura, K., et al. (1995). Mice deficient for the lysosomal proteinase cathepsin D exhibit progressive atrophy of the intestinal mucosa and profound destruction of lymphoid cells. Embo J 14, 3599-3608. Saitoh, T., Fujita, N., Jang, M.H., Uematsu, S., Yang, B.G., Satoh, T., Omori, H., Noda, T., Yamamoto, N., Komatsu, M., et al. (2008). Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 456, 264-268. Salazar, M., Carracedo, A., Salanueva, I.J., Hernandez-Tiedra, S., Lorente, M., Egia, A., Vazquez, P., Blazquez, C., Torres, S., Garcia, S., et al. (2009a). Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest. 204 Salazar, M., Carracedo, A., Salanueva, I.J., Hernandez-Tiedra, S., Lorente, M., Egia, A., Vazquez, P., Blazquez, C., Torres, S., Garcia, S., et al. (2009b). Cannabinoid action induces autophagymediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest 119, 1359-1372. Salomon, A.R., Voehringer, D.W., Herzenberg, L.A., and Khosla, C. (2000). Understanding and exploiting the mechanistic basis for selectivity of polyketide inhibitors of F(0)F(1)-ATPase. Proc Natl Acad Sci U S A 97, 14766-14771. Samara, C., Syntichaki, P., and Tavernarakis, N. (2007). Autophagy is required for necrotic cell death in Caenorhabditis elegans. Cell Death Differ. Sancak, Y., Peterson, T.R., Shaul, Y.D., Lindquist, R.A., Thoreen, C.C., Bar-Peled, L., and Sabatini, D.M. (2008). The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320, 1496-1501. Sarbassov, D.D., Ali, S.M., Kim, D.H., Guertin, D.A., Latek, R.R., Erdjument-Bromage, H., Tempst, P., and Sabatini, D.M. (2004). Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 14, 1296-1302. Sarbassov, D.D., Guertin, D.A., Ali, S.M., and Sabatini, D.M. (2005). Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098-1101. Satoo, K., Noda, N.N., Kumeta, H., Fujioka, Y., Mizushima, N., Ohsumi, Y., and Inagaki, F. (2009). The structure of Atg4B-LC3 complex reveals the mechanism of LC3 processing and delipidation during autophagy. EMBO J. Saucedo, L.J., Gao, X., Chiarelli, D.A., Li, L., Pan, D., and Edgar, B.A. (2003). Rheb promotes cell growth as a component of the insulin/TOR signalling network. Nat Cell Biol 5, 566-571. Scaringi, L., Cornacchione, P., Ayroldi, E., Corazzi, L., Capodicasa, E., Rossi, R., and Marconi, P. (2004). Omeprazole induces apoptosis in jurkat cells. Int J Immunopathol Pharmacol 17, 331-342. Scarlatti, F., Granata, R., Meijer, A.J., and Codogno, P. (2009). Does autophagy have a license to kill mammalian cells? Cell Death Differ 16, 12-20. Scherz-Shouval, R., Sagiv, Y., Shorer, H., and Elazar, Z. (2003). The COOH terminus of GATE-16, an intra-Golgi transport modulator, is cleaved by the human cysteine protease HsApg4A. J Biol Chem 278, 14053-14058. Schmelzle, T., and Hall, M.N. (2000). TOR, a central controller of cell growth. 205 Cell 103, 253-262. Schotte, P., Declercq, W., Van Huffel, S., Vandenabeele, P., and Beyaert, R. (1999). Non-specific effects of methyl ketone peptide inhibitors of caspases. FEBS Lett 442, 117-121. Schulze-Osthoff, K., Bakker, A.C., Vanhaesebroeck, B., Beyaert, R., Jacob, W.A., and Fiers, W. (1992). Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation. J Biol Chem 267, 5317-5323. Schweichel, J.U., and Merker, H.J. (1973). The morphology of various types of cell death in prenatal tissues. Teratology 7, 253-266. Scott, R.C., Juhasz, G., and Neufeld, T.P. (2007). Direct induction of autophagy by Atg1 inhibits cell growth and induces apoptotic cell death. Curr Biol 17, 1-11. Shaw, R.J., Bardeesy, N., Manning, B.D., Lopez, L., Kosmatka, M., DePinho, R.A., and Cantley, L.C. (2004). The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell 6, 91-99. Shaw, R.J., and Cantley, L.C. (2006). Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441, 424-430. Shen, H.M., Lin, Y., Choksi, S., Tran, J., Jin, T., Chang, L., Karin, M., Zhang, J., and Liu, Z.G. (2004). Essential roles of receptor-interacting protein and TRAF2 in oxidative stress-induced cell death. Mol Cell Biol 24, 5914-5922. Shen, H.M., and Liu, Z.G. (2006). JNK signaling pathway is a key modulator in cell death mediated by reactive oxygen and nitrogen species. Free Radic Biol Med 40, 928-939. Shetty, S., Graham, B.A., Brown, J.G., Hu, X., Vegh-Yarema, N., Harding, G., Paul, J.T., and Gibson, S.B. (2005). Transcription factor NF-kappaB differentially regulates death receptor expression involving histone deacetylase 1. Mol Cell Biol 25, 5404-5416. Shimizu, S., Kanaseki, T., Mizushima, N., Mizuta, T., Arakawa-Kobayashi, S., Thompson, C.B., and Tsujimoto, Y. (2004). Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol 6, 1221-1228. Song, G., Ouyang, G., and Bao, S. (2005). The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med 9, 59-71. Stack, J.H., DeWald, D.B., Takegawa, K., and Emr, S.D. (1995). 206 Vesicle-mediated protein transport: regulatory interactions between the Vps15 protein kinase and the Vps34 PtdIns 3-kinase essential for protein sorting to the vacuole in yeast. J Cell Biol 129, 321-334. Steed, P.M., Tansey, M.G., Zalevsky, J., Zhukovsky, E.A., Desjarlais, J.R., Szymkowski, D.E., Abbott, C., Carmichael, D., Chan, C., Cherry, L., et al. (2003). Inactivation of TNF signaling by rationally designed dominant-negative TNF variants. Science 301, 1895-1898. Sun, Q., Fan, W., Chen, K., Ding, X., Chen, S., and Zhong, Q. (2008). Identification of Barkor as a mammalian autophagy-specific factor for Beclin and class III phosphatidylinositol 3-kinase. Proc Natl Acad Sci U S A 105, 19211-19216. Suzuki, N.N., Yoshimoto, K., Fujioka, Y., Ohsumi, Y., and Inagaki, F. (2005). The crystal structure of plant ATG12 and its biological implication in autophagy. Autophagy 1, 119-126. Taguchi-Atarashi, N., Hamasaki, M., Matsunaga, K., Omori, H., Ktistakis, N.T., Yoshimori, T., and Noda, T. (2010). Modulation of local PtdIns3P levels by the PI phosphatase MTMR3 regulates constitutive autophagy. Traffic. Takahashi, Y., Coppola, D., Matsushita, N., Cualing, H.D., Sun, M., Sato, Y., Liang, C., Jung, J.U., Cheng, J.Q., Mule, J.J., et al. (2007). Bif-1 interacts with Beclin through UVRAG and regulates autophagy and tumorigenesis. Nat Cell Biol 9, 1142-1151. Takeuchi, H., Kondo, Y., Fujiwara, K., Kanzawa, T., Aoki, H., Mills, G.B., and Kondo, S. (2005). Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res 65, 3336-3346. Tal, M.C., Sasai, M., Lee, H.K., Yordy, B., Shadel, G.S., and Iwasaki, A. (2009). Absence of autophagy results in reactive oxygen species-dependent amplification of RLR signaling. Proc Natl Acad Sci U S A 106, 2770-2775. Tanida, I., Sou, Y.S., Ezaki, J., Minematsu-Ikeguchi, N., Ueno, T., and Kominami, E. (2004). HsAtg4B/HsApg4B/autophagin-1 cleaves the carboxyl termini of three human Atg8 homologues and delipidates microtubule-associated protein light chain 3- and GABAA receptor-associated protein-phospholipid conjugates. J Biol Chem 279, 36268-36276. Tanida, I., Tanida-Miyake, E., Komatsu, M., Ueno, T., and Kominami, E. (2002). Human Apg3p/Aut1p homologue is an authentic E2 enzyme for multiple substrates, GATE-16, GABARAP, and MAP-LC3, and facilitates the conjugation of hApg12p to hApg5p. J Biol Chem 277, 13739-13744. 207 Tanida, I., Tanida-Miyake, E., Ueno, T., and Kominami, E. (2001). The human homolog of Saccharomyces cerevisiae Apg7p is a Protein-activating enzyme for multiple substrates including human Apg12p, GATE-16, GABARAP, and MAP-LC3. J Biol Chem 276, 1701-1706. Tasdemir, E., Chiara Maiuri, M., Morselli, E., Criollo, A., D'Amelio, M., Djavaheri-Mergny, M., Cecconi, F., Tavernarakis, N., and Kroemer, G. (2008a). A dual role of p53 in the control of autophagy. Autophagy 4, 810-814. Tasdemir, E., Maiuri, M.C., Galluzzi, L., Vitale, I., Djavaheri-Mergny, M., D'Amelio, M., Criollo, A., Morselli, E., Zhu, C., Harper, F., et al. (2008b). Regulation of autophagy by cytoplasmic p53. Nat Cell Biol 10, 676-687. Tassa, A., Roux, M.P., Attaix, D., and Bechet, D.M. (2003). Class III phosphoinositide 3-kinase--Beclin1 complex mediates the amino acid-dependent regulation of autophagy in C2C12 myotubes. Biochem J 376, 577-586. Tee, A.R., Fingar, D.C., Manning, B.D., Kwiatkowski, D.J., Cantley, L.C., and Blenis, J. (2002). Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc Natl Acad Sci U S A 99, 13571-13576. Tee, A.R., Manning, B.D., Roux, P.P., Cantley, L.C., and Blenis, J. (2003). Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr Biol 13, 1259-1268. Temkin, V., Huang, Q., Liu, H., Osada, H., and Pope, R.M. (2006). Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol Cell Biol 26, 2215-2225. Tresse, E., Kosta, A., Luciani, M.F., and Golstein, P. (2007). From autophagic to necrotic cell death in Dictyostelium. Semin Cancer Biol 17, 94-100. Tsytsykova, A.V., Falvo, J.V., Schmidt-Supprian, M., Courtois, G., Thanos, D., and Goldfeld, A.E. (2007). Post-induction, stimulus-specific regulation of tumor necrosis factor mRNA expression. J Biol Chem 282, 11629-11638. Ullman, E., Fan, Y., Stawowczyk, M., Chen, H.M., Yue, Z., and Zong, W.X. (2007). Autophagy promotes necrosis in apoptosis-deficient cells in response to ER stress. Cell Death Differ. Van Noorden, C.J. (2001). The history of Z-VAD-FMK, a tool for understanding the significance of caspase inhibition. Acta Histochem 103, 241-251. Vandenabeele, P., Vanden Berghe, T., and Festjens, N. (2006). Caspase 208 inhibitors promote alternative cell death pathways. Sci STKE 2006, pe44. Vander Haar, E., Lee, S.I., Bandhakavi, S., Griffin, T.J., and Kim, D.H. (2007). Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol 9, 316-323. Vanhaesebroeck, B., and Alessi, D.R. (2000). The PI3K-PDK1 connection: more than just a road to PKB. Biochem J 346 Pt 3, 561-576. Varfolomeev, E., Blankenship, J.W., Wayson, S.M., Fedorova, A.V., Kayagaki, N., Garg, P., Zobel, K., Dynek, J.N., Elliott, L.O., Wallweber, H.J., et al. (2007). IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 131, 669-681. Vaux, D.L., Cory, S., and Adams, J.M. (1988). Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 335, 440-442. Vercammen, D., Beyaert, R., Denecker, G., Goossens, V., Van Loo, G., Declercq, W., Grooten, J., Fiers, W., and Vandenabeele, P. (1998a). Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med 187, 1477-1485. Vercammen, D., Brouckaert, G., Denecker, G., Van de Craen, M., Declercq, W., Fiers, W., and Vandenabeele, P. (1998b). Dual signaling of the Fas receptor: initiation of both apoptotic and necrotic cell death pathways. J Exp Med 188, 919-930. Vercammen, D., Vandenabeele, P., Beyaert, R., Declercq, W., and Fiers, W. (1997). Tumour necrosis factor-induced necrosis versus anti-Fas-induced apoptosis in L929 cells. Cytokine 9, 801-808. Vercammen, D., Vandenabeele, P., Declercq, W., Van de Craen, M., Grooten, J., and Fiers, W. (1995). Cytotoxicity in L929 murine fibrosarcoma cells after triggering of transfected human p75 tumour necrosis factor (TNF) receptor is mediated by endogenous murine TNF. Cytokine 7, 463-470. Vilcek, J., and Lee, T.H. (1991). Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem 266, 7313-7316. Vince, J.E., Wong, W.W., Khan, N., Feltham, R., Chau, D., Ahmed, A.U., Benetatos, C.A., Chunduru, S.K., Condon, S.M., McKinlay, M., et al. (2007). IAP antagonists target cIAP1 to induce TNFalpha-dependent apoptosis. Cell 131, 682-693. Virgin, H.W., and Levine, B. (2009). Autophagy genes in immunity. Nat Immunol 10, 461-470. 209 Wahl, G.M. (2006). Mouse bites dogma: how mouse models are changing our views of how P53 is regulated in vivo. Cell Death Differ 13, 973-983. Webber, J.L., and Tooze, S.A. (2009). Coordinated regulation of autophagy by p38alpha MAPK through mAtg9 and p38IP. EMBO J. Wei, Y., Pattingre, S., Sinha, S., Bassik, M., and Levine, B. (2008). JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell 30, 678-688. White, E. (2008). Autophagic cell death unraveled: Pharmacological inhibition of apoptosis and autophagy enables necrosis. Autophagy 4, 399-401. Wu, H., Yan, Y., and Backer, J.M. (2007). Regulation of class IA PI3Ks. Biochem Soc Trans 35, 242-244. Wu, Y.T., Tan, H.L., Huang, Q., Kim, Y.S., Pan, N., Ong, W.Y., Liu, Z.G., Ong, C.N., and Shen, H.M. (2008a). Autophagy plays a protective role during zVAD-induced necrotic cell death. Autophagy 4, 457-466. Wu, Y.T., Tan, H.L., Huang, Q., Ong, C.N., and Shen, H.M. (2009a). Activation of the PI3K-Akt-mTOR signaling pathway promotes necrotic cell death via suppression of autophagy. Autophagy 5. Wu, Y.T., Tan, H.L., Huang, Q., Ong, C.N., and Shen, H.M. (2009b). Activation of the PI3K-Akt-mTOR signaling pathway promotes necrotic cell death via suppression of autophagy. Autophagy 5, 824-834. Wu, Y.T., Zhang, S., Kim, Y.S., Tan, H.L., Whiteman, M., Ong, C.N., Liu, Z.G., Ichijo, H., and Shen, H.M. (2008b). Signaling pathways from membrane lipid rafts to JNK1 activation in reactive nitrogen species-induced non-apoptotic cell death. Cell Death Differ 15, 386-397. Wullschleger, S., Loewith, R., and Hall, M.N. (2006). TOR signaling in growth and metabolism. Cell 124, 471-484. Xie, Z., and Klionsky, D.J. (2007). Autophagosome formation: core machinery and adaptations. Nat Cell Biol 9, 1102-1109. Xie, Z., Nair, U., and Klionsky, D.J. (2008). Dissecting autophagosome formation: the missing pieces. Autophagy 4, 920-922. Xu, Y., Huang, S., Liu, Z.G., and Han, J. (2006a). Poly(ADP-ribose) polymerase-1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2-mediated JNK1 activation. J Biol Chem 281, 8788-8795. Xu, Y., Kim, S.O., Li, Y., and Han, J. (2006b). Autophagy contributes to caspase-independent macrophage cell death. J Biol Chem 281, 19179-19187. 210 Yamada, T., Carson, A.R., Caniggia, I., Umebayashi, K., Yoshimori, T., Nakabayashi, K., and Scherer, S.W. (2005). Endothelial nitric-oxide synthase antisense (NOS3AS) gene encodes an autophagy-related protein (APG9-like2) highly expressed in trophoblast. J Biol Chem 280, 18283-18290. Yan, Y., and Backer, J.M. (2007). Regulation of class III (Vps34) PI3Ks. Biochem Soc Trans 35, 239-241. Yan, Y., Flinn, R.J., Wu, H., Schnur, R.S., and Backer, J.M. (2009). hVps15, but not Ca2+/CaM, is required for the activity and regulation of hVps34 in mammalian cells. Biochem J 417, 747-755. Yang, C.F., Shen, H.M., and Ong, C.N. (1999). Protective effect of ebselen against hydrogen peroxide-induced cytotoxicity and DNA damage in HepG2 cells. Biochem Pharmacol 57, 273-279. Yang, Q., and Guan, K.L. (2007). Expanding mTOR signaling. Cell Res 17, 666-681. Yee, K.S., Wilkinson, S., James, J., Ryan, K.M., and Vousden, K.H. (2009). PUMA- and Bax-induced autophagy contributes to apoptosis. Cell Death Differ. Yonish-Rouach, E., Resnitzky, D., Lotem, J., Sachs, L., Kimchi, A., and Oren, M. (1991). Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6. Nature 352, 345-347. Yorimitsu, T., and Klionsky, D.J. (2005). Autophagy: molecular machinery for self-eating. Cell Death Differ 12 Suppl 2, 1542-1552. Yoshimori, T. (2007). Autophagy: paying Charon's toll. Cell 128, 833-836. Young, A.R., Chan, E.Y., Hu, X.W., Kochl, R., Crawshaw, S.G., High, S., Hailey, D.W., Lippincott-Schwartz, J., and Tooze, S.A. (2006). Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J Cell Sci 119, 3888-3900. Yousefi, S., Perozzo, R., Schmid, I., Ziemiecki, A., Schaffner, T., Scapozza, L., Brunner, T., and Simon, H.U. (2006). Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol 8, 1124-1132. Yu, L., Alva, A., Su, H., Dutt, P., Freundt, E., Welsh, S., Baehrecke, E.H., and Lenardo, M.J. (2004). Regulation of an ATG7-beclin program of autophagic cell death by caspase-8. Science 304, 1500-1502. Yu, L., Wan, F., Dutta, S., Welsh, S., Liu, Z., Freundt, E., Baehrecke, E.H., and Lenardo, M. (2006a). Autophagic programmed cell death by selective catalase degradation. Proc Natl Acad Sci U S A 103, 4952-4957. 211 Yu, S.W., Andrabi, S.A., Wang, H., Kim, N.S., Poirier, G.G., Dawson, T.M., and Dawson, V.L. (2006b). Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103, 18314-18319. Yuan, J., Shaham, S., Ledoux, S., Ellis, H.M., and Horvitz, H.R. (1993). The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell 75, 641-652. Yue, Z., Jin, S., Yang, C., Levine, A.J., and Heintz, N. (2003). Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci U S A 100, 15077-15082. Zakeri, Z., and Lockshin, R.A. (2008). Cell death: history and future. Adv Exp Med Biol 615, 1-11. Zalckvar, E., Berissi, H., Mizrachy, L., Idelchuk, Y., Koren, I., Eisenstein, M., Sabanay, H., Pinkas-Kramarski, R., and Kimchi, A. (2009). DAP-kinase-mediated phosphorylation on the BH3 domain of beclin promotes dissociation of beclin from Bcl-X(L) and induction of autophagy. EMBO Rep. Zalevsky, J., Secher, T., Ezhevsky, S.A., Janot, L., Steed, P.M., O'Brien, C., Eivazi, A., Kung, J., Nguyen, D.H., Doberstein, S.K., et al. (2007). Dominant-negative inhibitors of soluble TNF attenuate experimental arthritis without suppressing innate immunity to infection. J Immunol 179, 1872-1883. Zha, J., Harada, H., Yang, E., Jockel, J., and Korsmeyer, S.J. (1996). Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 87, 619-628. Zhang, D.W., Shao, J., Lin, J., Zhang, N., Lu, B.J., Lin, S.C., Dong, M.Q., and Han, J. (2009). RIP3, an Energy Metabolism Regulator that Switches TNF-Induced Cell Death from Apoptosis to Necrosis. Science. Zhang, S., Lin, Y., Kim, Y.S., Hande, M.P., Liu, Z.G., and Shen, H.M. (2007). c-Jun N-terminal kinase mediates hydrogen peroxide-induced cell death via sustained poly(ADP-ribose) polymerase-1 activation. Cell Death Differ 14, 1001-1010. Zhang, S., Lin, Z.N., Yang, C.F., Shi, X., Ong, C.N., and Shen, H.M. (2004). Suppressed NF-kappaB and sustained JNK activation contribute to the sensitization effect of parthenolide to TNF-alpha-induced apoptosis in human cancer cells. Carcinogenesis 25, 2191-2199. Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker, 212 S.H., and Goldberg, A.L. (2007). FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 6, 472-483. Zheng, L., Bidere, N., Staudt, D., Cubre, A., Orenstein, J., Chan, F.K., and Lenardo, M. (2006). Competitive control of independent programs of tumor necrosis factor receptor-induced cell death by TRADD and RIP1. Mol Cell Biol 26, 3505-3513. Zhong, Y., Wang, Q.J., Li, X., Yan, Y., Backer, J.M., Chait, B.T., Heintz, N., and Yue, Z. (2009). Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nat Cell Biol. Zhou, X., Ikenoue, T., Chen, X., Li, L., Inoki, K., and Guan, K.L. (2009). Rheb controls misfolded protein metabolism by inhibiting aggresome formation and autophagy. Proc Natl Acad Sci U S A. Zong, W.X., Ditsworth, D., Bauer, D.E., Wang, Z.Q., and Thompson, C.B. (2004). Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev 18, 1272-1282. Zong, W.X., Li, C., Hatzivassiliou, G., Lindsten, T., Yu, Q.C., Yuan, J., and Thompson, C.B. (2003). Bax and Bak can localize to the endoplasmic reticulum to initiate apoptosis. J Cell Biol 162, 59-69. Zong, W.X., and Thompson, C.B. (2006). Necrotic death as a cell fate. Genes Dev 20, 1-15. 213 [...]... effect of starvation on zVAD-induced necrotic cell death in L929 cells Figure 3.3 Other growth factors and amino acids have a similar pro-death effect as insulin Figure 3.4 Inhibition of the PI3K activity abolishes the pro-death effect of insulin on zVAD-induced necrosis in L929 cells Figure 3.5 Inhibition of mTOR activity by rapamycin abolishes the pro-death effect XIII of insulin Figure 3.6 Knockdown of. .. protective role in zVAD-induced cell death Figure 4.6 Knockdown of c-Jun blocks zVAD-induced autocrine TNFα production and necroptosis Figure 4.7 zVAD-induced TNFα transcription is depending on the MAPKs-AP-1 signaling pathway Figure 4.8 Critical role of PKC in zVAD-induced MAPKs-AP-1 activation, autocrine of TNFα and necroptosis Figure 4.9 Promotion of autocrine of TNFα combining with caspase-8 inhibition induces... bind to and antagonize functions of the anti-apoptosis Bcl-2 proteins to promote apoptosis (Danial, 2007) The Bcl-2 family members are predominantly involved in regulation of the intrinsic apoptotic pathway The fundamental underlying mechanism is that the Bax and Bak are able to induce the mitochondrial outer membrane permeabilization (MOMP) and the release of cytochrome c for initiating the intrinsic... 2008) According to the length of their prodomains and the positions in the apoptotic signaling cascade, caspases can be classified into two groups, the initiator caspases (caspase-1, -2, -4, -5, -8, -9, -10, -11, -12) and the effector caspases (caspase-3, -6, -7) Initiator caspases harbor long prodomains containing a protein-protein interaction motif, the death effector domain (DED) or the caspase... such machineries have been established, the extrinsic and the intrinsic pathways The extrinsic apoptotic pathway is classically initiated by the cell death receptors, such as TNF receptor 1 (TNFR1), Fas, and death receptor (DR) 4/5 The engagement of the cell death ligands with their respective receptors induces the formation of intracellular death-inducing signaling complexes (DISCs) consisting of multiple... mitigates the pro-death effect of insulin on necrosis Figure 3.7 Insulin suppresses autophagy induced by starvation Figure 4.1 Caspase inhibition is not sufficient for zVAD to induce necrosis Figure 4.2 zVAD-induced necrosis requires de novo protein synthesis and depends on RIP1 and RIP3 Figure 4.3 zVAD promotes autocrine of TNFα Figure 4.4 Blockage of TNF signaling pathway prevents zVAD-induced cell... implying that these two types of necrosis may utilize distinct routes for JNK activation and the consequent cell demise Interestingly, JNK has also been found to act upstream of PARP-1 and contributes to sustained PARP-1 activation, leading to necrosis in response to oxidative stress (Zhang et al., 2007) Therefore, the relationship and potential crosstalk between these two types of necrosis remain to... susceptible to necroptosis when engineered with RIP3 (Zhang et al., 2009) However, whether RIP3 also performs similar functions in PARP-1-mediated necrosis is not known These findings may provide novel insights and directions for us to further investigate the key role of caspase in necrotic cell death Interestingly, in addition to being capable of switching apoptosis into necrosis, 20 ... N-(2-Quinolyl)valyl-aspartyl-oph regulatory associated protein of mTOR V-rel reticuloendotheliosis viral oncogene homolog Ras homolog enriched in brain rapamycin-insensitive companion of mTOR receptor interacting protein reactive oxygen species p70 S6 kinase short interfering RNA second mitochondria-derived activator of caspase transactivation domain truncated Bid tandem fluorescent-tagged LC3 construct... its functions (Wahl, 2006) Besides, activation of p53 is governed by a variety of post-translational modifications (PTMs) including acetylation, phosphorylation, methylation, poly(ADP-ribosyl)ation et al For example, aceylation of lysine 373 of p53 by p300 and/or CBP markedly increases its transactivities toward the lower affinity-binding target genes (Knights et al., 2006) Phosphorylation of p53 at . elucidate the underlying molecular mechanisms using in vitro mammalian cell models. The following investigations have been conducted: (i) examining the role of autophagy in zVAD-induced necrosis. SUPPRESSION OF AUTOPHAGY BY ACTIVATION OF PI3K-AKT-MTOR AXIS PROMOTES NECROSIS 168 5.4 AUTOCRINE TNFΑ IS THE DEATH SIGNAL IN ZVAD-INDUCED NECROSIS 170 5.5 DUAL ROLE OF ZVAD DURING INDUCTION OF NECROPTOSIS. to suppress autophagy via suppression of lysosome function via inhibition of cathepsin enzyme activity. One surprising finding of this study was that growth factors such as insulin and IGF-1

Ngày đăng: 11/09/2015, 10:17

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan