Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-jwnkl Total loading time: 0 Render date: 2024-07-11T11:25:32.812Z Has data issue: false hasContentIssue false

30 - Apoptosis: the extrinsic pathway

from Part 2.2 - Molecular pathways underlying carcinogenesis: apoptosis

Published online by Cambridge University Press:  05 February 2015

Xinchen Teng
Affiliation:
Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
J. Marie Hardwick
Affiliation:
Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Programmed cell death (also known as PCD) is generally defined as a regulated process by which cells contribute to their own demise. Apoptosis is the best-characterized form of programmed cell death, but alternative non-apoptotic cell-death pathways important in human physiology and disease pathology are now actively studied. Regarding apoptosis, there are two general pathways, the extrinsic pathways and the intrinsic pathways, depending on whether the molecular factor that initiates the death pathway is extra-cellular or intra-cellular. Both extrinsic and intrinsic pathways lead to activation of caspases, the proteases that cleave many key protein targets inside cells, resulting in apoptotic cell morphology and cell death within a few minutes to hours. Although there may be alternative molecular pathways that cause apoptosis-like cell morphology, the term apoptosis most often refers to caspase-dependent cell death.

The extrinsic and intrinsic pathways activate different initiator caspases. Each initiator caspase is activated by a unique complex of proteins. The intrinsic death pathway involves mitochondria and is controlled by pro- and anti-apoptotic Bcl-2-family proteins that facilitate or inhibit the release of cytochrome c from the mitochondrial inter-membrane space. In turn, cytosolic cytochrome c and ATP/ADP bind Apaf-1, inducing oligomerization of Apaf-1 into a heptameric ring structure known as the apoptosome. The apoptosome activates caspase-9, which in turn cleaves and activates caspases-3 and -7 to mediate apoptosis during normal development and to prevent cancer. Other intrinsic apoptotic pathways are initiated by assembly of alternative caspase-activating complexes in response to intra-cellular factors, such as the PIDDosome complex, which activates caspase-2, and the inflammasome, which activates caspase-1 (originally known as ICE, IL-1β-converting enzyme).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 353 - 366
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Gruss, HJ, Dower, SK. Tumor necrosis factor ligand superfamily: involvement in the pathology of malignant lymphomas. Blood 1995;85:3378–404.
Bodmer, JL, Schneider, P, Tschopp, J. The molecular architecture of the TNF superfamily. Trends in Biochemical Science 2002;27:19–26.CrossRef
Schulze-Osthoff, K, Ferrari, D, Los, M, Wesselborg, S, Peter, ME.Apoptosis signaling by death receptors. European Journal of Biochemistry 1998;254:439–59.CrossRefGoogle ScholarPubMed
Ashkenazi, A, Dixit, VM. Death receptors: signaling and modulation. Science 1998;281:1305–8.CrossRef
Schneider, P, Holler, N, Bodmer, JL, et al. Conversion of membrane-bound Fas(CD95) ligand to its soluble form is associated with downregulation of its proapoptotic activity and loss of liver toxicity. Journal of Experimental Medicine 1998;187:1205–13.CrossRefGoogle ScholarPubMed
Wajant, H, Moosmayer, D, Wüest, T, et al. Differential activation of TRAIL-R1 and -2 by soluble and membrane TRAIL allows selective surface antigen-directed activation of TRAIL-R2 by a soluble TRAIL derivative. Oncogene 2001;20:4101–6.CrossRef
Grell, M, Wajane, H, Zimmermann, G, Scheurich, P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proceedings of the National Academy of Sciences USA 1998;95:570–5.CrossRef
Smith, CA, Farrah, T, Goodwin, RG. The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell 1994;76:959–62.CrossRef
Banner, DW, D’Arcy, A, Janes, W, et al. Crystal structure of the soluble human 55 kd TNF receptor-human TNF beta complex: implications for TNF receptor activation. Cell 1993;73:431–45.CrossRef
Schneider, P, Bodmer, JL, Thome, M, et al. Characterization of two receptors for TRAIL. FEBS Letters 1997;416:329–34.CrossRef
Schneider, P, Bodmer, JL, Holler, N, et al. Characterization of Fas (Apo-1, CD95)-Fas ligand interaction. Journal of Biological Chemistry, 1997;272:18 827–33.CrossRefGoogle ScholarPubMed
Mongkolsapaya, J, Grimes, JM, Chen, N, et al. Structure of the TRAIL-DR5 complex reveals mechanisms conferring specificity in apoptotic initiation. Nature Structural Biology 1999;6:1048–53.CrossRef
Locksley, RM, Killeen, N, Lenardo, MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001;104:487–501.CrossRef
Chan, FK. Three is better than one: pre-ligand receptor assembly in the regulation of TNF receptor signaling. Cytokine 2007;37:101–7.CrossRef
Jin, Z, El-Deiry, WS. Overview of cell death signaling pathways. Cancer Biology and Therapy 2005;4:139–63.
Chinnaiyan, AM, O’Rourke, K, Tweari M, Dixit VM. FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 1995;81:505–12.CrossRef
Hsu, H, Xiong, J, Goeddel, DV. The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell 1995;81:495–504.CrossRef
Yeh, WC, Pompa, JL, McCurrach, ME, et al. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science 1998;279:1954–8.CrossRef
Newton, K, Harris, AW, Bath, ML, Smith, KG, Strasser, A. A dominant interfering mutant of FADD/MORT1 enhances deletion of autoreactive thymocytes and inhibits proliferation of mature T lymphocytes. EMBO Journal, 1998;17:706–18.CrossRef
Zornig, M, Hueber, AO, Evan, G. p53-dependent impairment of T-cell proliferation in FADD dominant-negative transgenic mice. Current Biology, 1998;8:467–70.CrossRef
Zhang, J, Cado, D, Chen, A, Kabra, NH, Winoto, A. Fas-mediated apoptosis and activation-induced T-cell proliferation are defective in mice lacking FADD/Mort1. Nature 1998;392:296–300.CrossRef
Oberst, A, Dillon, CP, Weinlich, R, et al. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 2011;471:363–7.CrossRef
Kaiser, WJ, Upton, JW, Long, AB, et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 2011;471:368–72.CrossRef
Zhang, H, Zhou, X, McQuade, T, et al. Functional complementation between FADD and RIP1 in embryos and lymphocytes. Nature 2011;471:373–6.CrossRef
Chan, KF, Siegel, MR, Lenardo, JM. Signaling by the TNF receptor superfamily and T cell homeostasis. Immunity 2000;13:419–22.CrossRef
O’Reilly, LA, Tai, L, Lee, L, et al. Membrane-bound Fas ligand only is essential for Fas-induced apoptosis. Nature 2009;461:659–63.CrossRef
Muppidi, JR, Siegel, RM. Ligand-independent redistribution of Fas (CD95) into lipid rafts mediates clonotypic T cell death. Nature Immunology 2004;5:182–9.CrossRef
Holler, N, Tardivel, A, Kovacsovics-Banowski, M, et al. Two adjacent trimeric Fas ligands are required for Fas signaling and formation of a death-inducing signaling complex. Molecular and Cellular Biology, 2003;23:1428–40.CrossRef
Scott, FL, Stec, B, Pop, C, et al. The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature 2009;457:1019–22.CrossRef
Mace, PD, Riedl, SJ. Molecular cell death platforms and assemblies. Current Opinion in Cell Biology 2010;22:828–36.CrossRef
Esposito, D, Sankar, A, Morgner, N, et al. Solution NMR investigation of the CD95/FADD homotypic death domain complex suggests lack of engagement of the CD95 C terminus. Structure 2010;18:1378–90.CrossRef
Wang, L, Yang, JK, Kabaleeswaran, V, et al. The Fas-FADD death domain complex structure reveals the basis of DISC assembly and disease mutations. Nature Structural and Molecular Biology 2010;17:1324–9.CrossRef
Park, HH, Logette, E, Raunser, S, et al. Death domain assembly mechanism revealed by crystal structure of the oligomeric PIDDosome core complex. Cell 2007;128:533–46.CrossRef
Park, HH. Structural analyses of death domains and their interactions. Apoptosis 2011;16:209–20.CrossRef
Kersse, K, Verspurten, J, Vanden Berghe, T, Vandenabeele, P. The death-fold superfamily of homotypic interaction motifs. Trends in Biochemical Science, 2011;36:541–52.CrossRef
Yan, N, Shi, Y. Mechanisms of apoptosis through structural biology. Annual Review of Cell and Developmental Biology 2005;21:35–56.CrossRef
Schleich, K, Warnken, U, Fricker, N, et al. Stoichiometry of the CD95 death-inducing signaling complex: experimental and modeling evidence for a death effector domain chain model. Molecular Cell 2012;47:306–19.CrossRef
Dickens, LS, Boyd, RS, Jukes-Jones, R, et al. A death effector domain chain DISC model reveals a crucial role for caspase-8 chain assembly in mediating apoptotic cell death. Molecular Cell 2012;47:291–305.CrossRef
Muzio, M, Stockwell, BR, Stennicke, HR, Salvesen, GS, Dixit, VM.An induced proximity model for caspase-8 activation. Journal of Biological Chemistry 1998;273:2926–30.CrossRefGoogle ScholarPubMed
Salvesen, GS, Dixit, VM. Caspase activation: the induced-proximity model. Proceedings of the National Academy of Sciences USA 1999;96:10 964–7.
Boatright, KM, Renatus, M, Scott, FL, et al. A unified model for apical caspase activation. Molecular Cell 2003;11:529–41.CrossRef
Keller, N, Mares, J, Zerbe, O, Grütter, MG. Structural and biochemical studies on procaspase-8: new insights on initiator caspase activation. Structure 2009;17:438–48.CrossRef
Keller, N, Grütter, MG, Zerbe, O. Studies of the molecular mechanism of caspase-8 activation by solution NMR. Cell Death and Differentiation 2010;17:710–18.CrossRef
Chang, DW, Xing, Z, Capacio, VL, Peter, ME, Yang, X. Interdimer processing mechanism of procaspase-8 activation. EMBO Journal 2003;22:4132–42.CrossRef
Pop, C, Fitsgerald, P, Green, DR, Salvesen, GS. Role of proteolysis in caspase-8 activation and stabilization. Biochemistry 2007;46:4398–407.CrossRef
Jin, Z, Li, Y, Pitti, R, et al. Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell 2009;137:721–35.CrossRef
Gonzalvez, F, Lawrence, D, Yang, B, et al. TRAF2 sets a threshold for extrinsic apoptosis by tagging caspase-8 with a ubiquitin shutoff timer. Molecular Cell 2012;48:888–99.CrossRef
Pan, G, O’Rourke, K, Chinnaiyan, AM, et al. The receptor for the cytotoxic ligand TRAIL. Science 1997;276:111–3.CrossRef
Sheridan, JP, Marsters, SA, Pitti, RM, et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science 1997;277:818–21.CrossRef
Marsters, SA, Sheridan, JP, Pitti, RM, et al. A novel receptor for Apo2L/ TRAIL contains a truncated death domain. Current Biology 1997;7:1003–6.CrossRef
Pitti, RM, Marsters, SA, Lawrence, D, et al. Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature 1998;396:699–703.CrossRef
Emery, JG, McDonnell, P, Burke, MB, et al. Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. Journal of Biological Chemistry 1998;273:14 363–7.CrossRefGoogle ScholarPubMed
LeBlanc, HN, Ashkenazi, A. Apo2L/TRAIL and its death and decoy receptors. Cell Death and Differentiation, 2003;10:66–75.CrossRef
Ashkenazi, A, Dixit, VM. Apoptosis control by death and decoy receptors. Current Opinion in Cell Biology 1999;11:255–60.CrossRef
Wagner, KW, Punnoose, EA, Januario, T, et al. Death-receptor O-glycosylation controls tumor-cell sensitivity to the proapoptotic ligand Apo2L/TRAIL. Nature Medicine 2007;13:1070–7.CrossRef
Shatnyeva, OM, Kubarenko, AV, Weber, CE, et al. Modulation of the CD95-induced apoptosis: the role of CD95 N-glycosylation. PLoS One 2011;6:e19927.
Peter, ME, Hellbardt, S, Schwartz-Albiez, R, et al. Cell surface sialylation plays a role in modulating sensitivity towards APO-1-mediated apoptotic cell death. Cell Death and Differentiation 1995;2:163–71.
Keppler, OT, Peter, ME, Hinderlich, S, et al. Differential sialylation of cell surface glycoconjugates in a human B lymphoma cell line regulates susceptibility for CD95 (APO-1/Fas)-mediated apoptosis and for infection by a lymphotropic virus. Glycobiology 1999;9:557–69.CrossRef
Swindall, AF, Bellis, SL.Sialylation of the Fas death receptor by ST6Gal-I provides protection against Fas-mediated apoptosis in colon carcinoma cells. Journal of Biological Chemistry 2011;286:22 982–90.CrossRefGoogle ScholarPubMed
Feig, C, Tchikov, V, Schütze, S, Peter, ME. Palmitoylation of CD95 facilitates formation of SDS-stable receptor aggregates that initiate apoptosis signaling. EMBO Journal 2007;26:221–31.CrossRef
Chakrabandhu, K, Hérincs, Z, Huault, S, et al. Palmitoylation is required for efficient Fas cell death signaling. EMBO Journal 2007;26:209–20.CrossRef
Rossin, A, Derouet, M, Abdel-Sater, F, Hueber, AO. Palmitoylation of the TRAIL receptor DR4 confers an efficient TRAIL-induced cell death signalling. Biochemical Journal 2009;419:185–92.CrossRef
Irmler, M, Thome, M, Hahne, M, et al. Inhibition of death receptor signals by cellular FLIP. Nature 1997;388:190–5.CrossRef
Scaffidi, C, Schmitz, I, Krammer, PH, Peter, ME.The role of c-FLIP in modulation of CD95-induced apoptosis. Journal of Biological Chemistry 1999;274:1541–8.CrossRefGoogle ScholarPubMed
Micheau, O. Cellular FLICE-inhibitory protein: an attractive therapeutic target? Expert Opinion on Therapeutic Targets 2003;7:559–73.
Golks, A, Brenner, D, Fritsch, C, Krammer, PH, Lavrik, IN.c-FLIPR, a new regulator of death receptor-induced apoptosis. Journal of Biological Chemistry 2005;280:14507–13.CrossRefGoogle ScholarPubMed
Chang, DW, Xing, Z, Pan, Y, et al. c-FLIP(L) is a dual function regulator for caspase-8 activation and CD95-mediated apoptosis. EMBO Journal 2002;21:3704–14.CrossRef
Micheau, O, Thome, M, Schneider, P, et al. The long form of FLIP is an activator of caspase-8 at the Fas death-inducing signaling complex. Journal of Biological Chemistry 2002;277:45 162–71.CrossRefGoogle Scholar
Yu, JW, Jeffrey, PD, Shi, Y. Mechanism of procaspase-8 activation by c-FLIPL. Proceedings of the National Academy of Sciences USA 2009;106:8169–74.CrossRef
Yu, JW, Shi, Y. FLIP and the death effector domain family. Oncogene, 2008;27:6216–27.CrossRef
Kataoka, T, Tschopp, J. N-terminal fragment of c-FLIP(L) processed by caspase 8 specifically interacts with TRAF2 and induces activation of the NF-kappaB signaling pathway. Molecular Cell Biology 2004;24:2627–36.CrossRef
Bagnoli, M, Canevari, SMezzanzanica, D.Cellular FLICE-inhibitory protein (c-FLIP) signalling: a key regulator of receptor-mediated apoptosis in physiologic context and in cancer. International Journal of Biochemistry and Cell Biology 2010;42:210–13.CrossRefGoogle Scholar
Kreuzaler, P, Watson, CJ. Killing a cancer: what are the alternatives? Nature Reviews Cancer 2012;12:411–24.
Barnhart, BC, Alappat, EC, Peter, ME. The CD95 type I/type II model. Seminars on Immunology 2003;15:185–93.CrossRef
Scaffidi, C, Fulda, S, Srinivasan, A, et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO Journal 1998;17:1675–87.CrossRef
Ozoren, N, El-Deiry, WS. Defining characteristics of Types I and II apoptotic cells in response to TRAIL. Neoplasia, 2002;4:551–7.CrossRef
Clem, RJ, Cheng, EH, Karp, CL, et al. Modulation of cell death by Bcl-XL through caspase interaction. Proceedings of the National Academy of Sciences USA 1998;95:554–9.CrossRef
Cheng, EH, Kirsch, DG, Clem, RJ, et al. Conversion of Bcl-2 to a Bax-like death effector by caspases. Science 1997;278:1966–8.CrossRef
Li, H, Zhu, H, Xu, CJ, Yuan, J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1998;94:491–501.CrossRef
Luo, X, Budihardjo, I, Zou, H, Slaughter, C, Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 1998;94:481–90.CrossRef
Kroemer, G, Galluzzi, L, Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiolological Reviews 2007;87:99–163.CrossRef
Li, P, Nijhawan, D, Budihardjo, I, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997;91:479–89.CrossRef
Guicciardi, ME, Gores, GJ. Life and death by death receptors. FASEB Journal 2009;23:1625–37.CrossRef
Aouacheria, A, Rech de Laval, V, Combet, C, Hardwick, JM. Evolution of Bcl-2 homology motifs: homology versus homoplasy. Trends in Cell Biology 2012;23:103–11.CrossRef
Scorrano, L.Caspase-8 goes cardiolipin: a new platform to provide mitochondria with microdomains of apoptotic signals?Journal of Cell Biology 2008;183:579–81.CrossRefGoogle ScholarPubMed
Gonzalvez, F, Schug, ZT, Houtkooper, RH, et al. Cardiolipin provides an essential activating platform for caspase-8 on mitochondria. Journal of Cell Biology 2008;183:681–96.CrossRefGoogle ScholarPubMed
Le Roy, C, Wrana, JL. Clathrin- and non-clathrin-mediated endocytic regulation of cell signalling. Nature Reviews Molecular Cell Biology 2005;6:112–26.CrossRef
Teis, D, Huber, LA. The odd couple: signal transduction and endocytosis. Cellular and Molecular Life Sciences 2003;60:2020–33.CrossRef
Miaczynska, M, Pelkmans, L, Zerial, M. Not just a sink: endosomes in control of signal transduction. Current Opinion in Cell Biology 2004;16:400–6.CrossRef
Algeciras-Schimnich, A, Shen, L, Barnhart, BC, et al. Molecular ordering of the initial signaling events of CD95. Molecular and Cellular Biology 2002;22:207–20.CrossRef
Parlato, S, Giammarioli, AM, Logozzi, M, et al. CD95 (APO-1/Fas) linkage to the actin cytoskeleton through ezrin in human T lymphocytes: a novel regulatory mechanism of the CD95 apoptotic pathway. EMBO Journal 2000;19:5123–34.CrossRef
Siegel, RM, Muppidi, JR, Sarker, M, et al. SPOTS: signaling protein oligomeric transduction structures are early mediators of death receptor-induced apoptosis at the plasma membrane. Journal of Cell Biology 2004;167:735–44.CrossRefGoogle ScholarPubMed
Eramo, A, Sargiacomo, M, Ricci-Vitiani, L, et al. CD95 death-inducing signaling complex formation and internalization occur in lipid rafts of type I and type II cells. European Journal of Immunology 2004;34:1930–40.CrossRefGoogle ScholarPubMed
Legembre, P, Daburon, S, Moreau, P, Moreau, JF, Taupin, JL.Modulation of Fas-mediated apoptosis by lipid rafts in T lymphocytes. Journal of Immunology 2006;176:716–20.CrossRefGoogle ScholarPubMed
Lee, KH, Feig, C, Tchikov, V, et al. The role of receptor internalization in CD95 signaling. EMBO Journal 2006;25:1009–23.CrossRef
Schutze, S, Schneider-Brachert, W. Impact of TNF-R1 and CD95 internalization on apoptotic and antiapoptotic signaling. Results and Problems in Cell Differentiation 2009;49:63–85.CrossRef
Schneider-Brachert, W, Tchikov, V, Merkel, O, et al. Inhibition of TNF receptor 1 internalization by adenovirus 14.7K as a novel immune escape mechanism. Journal of Clinical Investigation 2006;116:2901–13.CrossRefGoogle ScholarPubMed
Yuan, J, Kroemer, G. Alternative cell death mechanisms in development and beyond. Genes and Development 2010;24:2592–602.CrossRef
Micheau, O, Tschopp, J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 2003;114:181–90.CrossRef
Schneider-Brachert, W, Tchikov, V, Neumeyer, J, et al. Compartmentalizat-ion of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles. Immunity 2004;21:415–28.
Zhao, X, Liu, Y, Ma, Q, et al. Caveolin-1 negatively regulates TRAIL-induced apoptosis in human hepatocarcinoma cells. Biochemical and Biophysical Research Communications 2009;378:21–6.CrossRef
Austin, CD, Lawrence, DA, Peden, AA, et al. Death-receptor activation halts clathrin-dependent endocytosis. Proceedings of the National Academy of Sciences USA 2006;103:10 283–8.
Kohlhaas, SL, Craxton, A, Sun, XM, Pinkoski, MJ, Cohen, GM.Receptor-mediated endocytosis is not required for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. Journal of Biological Chemistry 2007;282:12 831–41.CrossRefGoogle Scholar
Schutze, S, Tchikov, V, Schneider-Brachert, W. Regulation of TNFR1 and CD95 signalling by receptor compartmentalization. Nature Reviews Molecular Cell Biology 2008;9:655–62.CrossRef
Schütze, S, Machleidt, T, Adam, D, et al. Inhibition of receptor internalization by monodansylcadaverine selectively blocks p55 tumor necrosis factor receptor death domain signaling. Journal of Biological Chemistry, 1999;274():10203–12.
Woo, CH, Machleidt, T, Adam, D, et al. Inhibition of receptor internalization attenuates the TNFalpha-induced ROS generation in non-phagocytic cells. Biochemical and Biophysical Research Communications 2006;351:972–8.CrossRef
Declercq, W, Vanden Berghe, T, Vandenabeele, P. RIP kinases at the crossroads of cell death and survival. Cell 2009;138:229–32.CrossRef
Meylan, E, Tschopp, J. The RIP kinases: crucial integrators of cellular stress. Trends in Biochemical Sciences 2005;30:151–9.CrossRef
Wilson, NS, Dixit, V, Ashkenazi, A. Death receptor signal transducers: nodes of coordination in immune signaling networks. Nature Immunology 2009;10:348–55.CrossRef
Ea, CK, Deng, L, Xia, ZP, Pineda, G, Chen, ZJ. Activation of IKK by TNFalpha requires site-specific ubiquitination of RIP1 and polyubiquitin binding by NEMO. Molecular Cell 2006;22:245–57.CrossRef
Mahoney, DJ, Cheung, HH, Mrad, RL, et al. Both cIAP1 and cIAP2 regulate TNFalpha-mediated NF-kappaB activation. Proceedings of the National Academy of Sciences USA 2008;105:11 778–83.
Varfolomeev, E, Goncharov, T, Fedorova, AV, et al. c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. Journal of Biological Chemistry 2008;283:24 295–9.CrossRefGoogle ScholarPubMed
Ikeda, F, Deribe, YL, Skånland, SS, et al. SHARPIN forms a linear ubiquitin ligase complex regulating NF-kappaB activity and apoptosis. Nature 2011;471:637–41.CrossRef
Tokunaga, F, Nakagawa, T, Nakahara, M, et al. SHARPIN is a component of the NF-kappaB-activating linear ubiquitin chain assembly complex. Nature 2011;471:633–6.CrossRef
Gerlach, B, Cordier, SM, Schmukle, AC, et al. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 2011;471:591–6.CrossRef
O’Donnell, MA, Legarda-Addison, D, Skountzos, P, Yeh, WC, Ting, AT. Ubiquitination of RIP1 regulates an NF-kappaB-independent cell-death switch in TNF signaling. Current Biology 2007;17:418–24.CrossRef
Wang, L, Du, F, Wang, X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell 2008;133:693–703.CrossRef
Clem, RJ, Sheu, TT, Richter, BW, et al. c-IAP1 is cleaved by caspases to produce a proapoptotic C-terminal fragment. Journal of Biological Chemistry 2001;276:7602–8.CrossRefGoogle ScholarPubMed
Hitomi, J, Christofferson, DE, Ng, A, et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 2008;135:1311–23.CrossRef
Galluzzi, L, Kroemer, G. Necroptosis: a specialized pathway of programmed necrosis. Cell 2008;135:1161–3.CrossRef
Festjens, N, Vanden Berghe, T, Cornelis, S, Vandenabeele, P. RIP1, a kinase on the crossroads of a cell's decision to live or die. Cell Death and Differentiation 2007;14:400–10.CrossRef
Degterev, A, Hitomi, J, Germsheid, M, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nature Chemical Biology 2008;4:313–21.CrossRef
Tenev, T, Bianchi, K, Darding, M, et al. The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Molecular Cell 2011;43:432–48.CrossRef
Feoktistova, M, Geserick, P, Kellert, B, et al. cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Molecular Cell 2011;43:449–63.CrossRef
Hahne, M, Rimoldi, D, Schröter, M, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science 1996;274:1363–6.CrossRef
Strand, S, Hofmann, WJ, Hug, H, et al. Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cells–a mechanism of immune evasion? Nature Medicine 1996;2:1361–6.
Leithauser, F, Dhein, J, Mechtersheimer, G, et al. Constitutive and induced expression of APO-1, a new member of the nerve growth factor/tumor necrosis factor receptor superfamily, in normal and neoplastic cells. Laboratory Investigations 1993;69:415–29.
Moller, P, Koretz, K, Leithäuser, F, et al. Expression of APO-1 (CD95), a member of the NGF/TNF receptor superfamily, in normal and neoplastic colon epithelium. International Journal of Cancer 1994;57:371–7.CrossRefGoogle Scholar
Ozoren, N, Fisher, MJ, Kim, K, et al. Homozygous deletion of the death receptor DR4 gene in a nasopharyngeal cancer cell line is associated with TRAIL resistance. International Journal of Oncology, 2000;16:917–25.Google Scholar
Horak, P, Pils, D, Haller, G, et al. Contribution of epigenetic silencing of tumor necrosis factor-related apoptosis inducing ligand receptor 1 (DR4) to TRAIL resistance and ovarian cancer. Molecular Cancer Research, 2005;3:335–43.CrossRef
Elias, A, Seigelin, MD, Steinmüller, A, et al. Epigenetic silencing of death receptor 4 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in gliomas. Clinical Cancer Research 2009;15:5457–65.CrossRef
Bae, SI, Cheriyath, V, Jacobs, BS, Reu, FJ, Borden, EC. Reversal of methylation silencing of Apo2L/TRAIL receptor 1 (DR4) expression overcomes resistance of SK-MEL-3 and SK-MEL-28 melanoma cells to interferons (IFNs) or Apo2L/TRAIL. Oncogene 2008;27:490–8.CrossRef
Jin, Z, McDonald, ER, 3rd, Dicker DT, El-Deiry WS. Deficient tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor transport to the cell surface in human colon cancer cells selected for resistance to TRAIL-induced apoptosis. Journal of Biological Chemistry 2004;279:35 829–39.CrossRefGoogle Scholar
Shin, MS, Park, WS, Kim, SY, et al. Alterations of Fas (Apo-1/CD95) gene in cutaneous malignant melanoma. American Journal of Pathology 1999;154:1785–91.CrossRefGoogle ScholarPubMed
Shin, MS, Kim, HS, Lee, SH, et al. Alterations of Fas-pathway genes associated with nodal metastasis in non-small cell lung cancer. Oncogene 2002;21:4129–36.CrossRef
Lee, SH, Shin, MS, Kim, HS, et al. Alterations of the DR5/TRAIL receptor 2 gene in non-small cell lung cancers. Cancer Research 1999;59:5683–6.
Pai, SI, Wu, GS, Ozören, N, et al. Rare loss-of-function mutation of a death receptor gene in head and neck cancer. Cancer Research 1998;58:3513–18.
Shin, MS, Kim, HS, Lee, SH, et al. Mutations of tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1) and receptor 2 (TRAIL-R2) genes in metastatic breast cancers. Cancer Research 2001;61:4942–6.
Fisher, MJ, Virmani, AK, Wu, L, et al. Nucleotide substitution in the ectodomain of trail receptor DR4 is associated with lung cancer and head and neck cancer. Clinical Cancer Research 2001;7:1688–97.
Bin, L, Thorburn, J, Thomas, LR, et al. Tumor-derived mutations in the TRAIL receptor DR5 inhibit TRAIL signaling through the DR4 receptor by competing for ligand binding. Journal of Biological Chemistry 2007;282:28 189–94.CrossRefGoogle ScholarPubMed
Shen, HW, Wu, YL, Peng, SY. [Overexpression and genomic amplification of decoy receptor 3 in hepatocellular carcinoma and significance thereof]. Zhonghua Yi Xue Za Zhi, 2003;83:744–7.
Bai, C, Connolly, B, Matzker, ML, et al. Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplification and its location in a four-gene cluster. Proceedings of the National Academy of Sciences USA 2000;97:1230–5.CrossRef
Ohshima, K, Haraoka, S, Sughara, M, et al. Amplification and expression of a decoy receptor for fas ligand (DcR3) in virus (EBV or HTLV-I) associated lymphomas. Cancer Letters 2000;160:89–97.CrossRef
Roth, W, Isenmann, S, Nakamura, M, et al. Soluble decoy receptor 3 is expressed by malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer Research 2001;61:2759–65.
Wu, Y, Guo, E, Yu, J, Xie, Q.High DcR3 expression predicts stage pN2–3 in gastric cancer. American Journal of Clinical Oncology 2008;31:79–83.CrossRefGoogle ScholarPubMed
Tourneur, L, Delluc, S, Lévy, V, et al. Absence or low expression of fas-associated protein with death domain in acute myeloid leukemia cells predicts resistance to chemperiodicalapy and poor outcome. Cancer Research 2004;64:8101–8.CrossRef
Soung, YH, Lee, JW, Kim, SY, et al. Mutation of FADD gene is rare in human colon and stomach cancers. APMIS, 2004;112:595–7.CrossRef
Stupack, DG. Caspase-8 as a therapeutic target in cancer. Cancer Letters 2013;322:133–40.CrossRef
Kim, HS, Lee, JW, Soung, YH, et al. Inactivating mutations of caspase-8 gene in colorectal carcinomas. Gastroenterology 2003;125:708–15.CrossRef
Mandruzzato, S, Brasseur, F, Andry, G, Boon, T, van der Bruggen, P.A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. Journal of Experimental Medicine 1997;186:785–93.CrossRefGoogle ScholarPubMed
Soung, YH, Lee, JW, Kim, SY, et al. CASPASE-8 gene is inactivated by somatic mutations in gastric carcinomas. Cancer Research 2005;65:815–21.
Barbero, S, Barilà, D, Mielgo, A, et al. Identification of a critical tyrosine residue in caspase 8 that promotes cell migration. Journal of Biological Chemistry 2008;283:13 031–4.CrossRefGoogle ScholarPubMed
Senft, J, Helfer, B, Frisch, SM. Caspase-8 interacts with the p85 subunit of phosphatidylinositol 3-kinase to regulate cell adhesion and motility. Cancer Research 2007;67:11 505–9.
Torres, VA, Mielgo, A, Barbero, S, et al. Rab5 mediates caspase-8-promoted cell motility and metastasis. Molecular Biology of the Cell 2010;21:369–76.CrossRef
Safa, AR, Day, TW, Wu, CH. Cellular FLICE-like inhibitory protein (C-FLIP): a novel target for cancer therapy. Current Cancer Drug Targets 2008;8:37–46.CrossRef
Bagnoli, M, Balladore, E, Luison, E, et al. Sensitization of p53-mutated epithelial ovarian cancer to CD95-mediated apoptosis is synergistically induced by cisplatin pretreatment. Molecular Cancer Therapeutics 2007;6:762–72.CrossRef
Geserick, P, Drewniok, C, Hupe, M, et al. Suppression of cFLIP is sufficient to sensitize human melanoma cells to TRAIL- and CD95L-mediated apoptosis. Oncogene 2008;27:3211–20.CrossRef
Rogers, KM, Thomas, M, Galligan, L, et al. Cellular FLICE-inhibitory protein regulates chemperiodicalapy-induced apoptosis in breast cancer cells. Molecular Cancer Therapeutics 2007;6:1544–51.CrossRef
Ozoren, N, El-Deiry WS. Cell surface Death receptor signaling in normal and cancer cells. Seminars on Cancer Biology 2003;13:135–47.CrossRef
Schulze-Bergkamen, H, Krammer, PH, Apoptosis in cancer–implications for therapy. Seminars on Oncology 2004;31:90–119.
Mellier, G, Huang, S, Shenoy, K, Pervaiz, S. TRAILing death in cancer. Molecular Aspects of Medicine, 2010;31():93–112.
Koschny, R, Walczak, H, Ganten, TM.The promise of TRAIL–potential and risks of a novel anticancer therapy. Journal of Molecular Medicine (Berlin) 2007;85:923–35.CrossRefGoogle ScholarPubMed
Kurokawa, M, Zhao, C, Reya, T, Kornbluth, S. Inhibition of apoptosome formation by suppression of Hsp90beta phosphorylation in tyrosine kinase-induced leukemias. Molecular and Cellular Biology 2008;28:5494–506.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Apoptosis: the extrinsic pathway
    • By Xinchen Teng, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA, J. Marie Hardwick, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.031
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Apoptosis: the extrinsic pathway
    • By Xinchen Teng, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA, J. Marie Hardwick, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.031
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Apoptosis: the extrinsic pathway
    • By Xinchen Teng, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA, J. Marie Hardwick, Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.031
Available formats
×