Skip to main content Accessibility help
×
Hostname: page-component-5c6d5d7d68-7tdvq Total loading time: 0 Render date: 2024-08-07T05:07:53.709Z Has data issue: false hasContentIssue false

31 - Apoptosis: the intrinsic pathway

from Part 2.2 - Molecular pathways underlying carcinogenesis: apoptosis

Published online by Cambridge University Press:  05 February 2015

Jody White
Affiliation:
Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 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

Mammalian cells have developed the means for efficient and ordered self-removal from tissues, termed programmed cell death. Of the pathways encompassed in this class, apoptosis is the most highly characterized mode of cell death (1). Apoptosis is a physiological, energy-dependent process whereby a cell systematically breaks down its intra-cellular components and signals patrolling immune cells to engulf the dying cell (1–3). This process prevents the release of the cell's enzymatic or infectious contents and allows for recycling of valuable materials. This process is in contrast to the accidental mode of cell death, termed necrosis, where cells with ruptured cell membranes leach their cellular contents into surrounding healthy tissue, causing damage. In contrast, programmed cell death describes a class of well-conserved cellular mechanisms that provides a way for degradation of diseased, damaged, or unnecessary cells in a non-inflammatory manner (4,5).

Apoptosis has been broadly described as either intrinsically (mitochondrial initiation) or extrinsically (plasma membrane receptor initiation) mediated during the initiation phase of the cell-death process (6,7). Specific proteases and multi-meric complexes are attributed to each of these pathways, although both routes end in the activation of common downstream mediators (8). For the extrinsic pathway, the binding of ligands to death receptors on the plasma membrane leads to the formation of death-inducing signaling complexes (DISCs) that then go on to signal other apoptotic events. Specifics of the extrinsic mode of apoptosis are covered in Chapter 30. In contrast, the intrinsic pathway is initiated by an intra-cellular event, such as mitochondrial and/or DNA damage (9,10). Other cell-death pathways have been suggested that do not fit into either the extrinsic or intrinsic paradigm (11). These events, however, are still classified as apoptosis rather than necrosis because of certain characteristic apoptotic features being present.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 367 - 377
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

Kerr, JF, Wyllie, AH, Currie, AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer 1972;26:239–57.CrossRefGoogle ScholarPubMed
Wyllie, AH, Kerr, JF, Currie, AR. Cell death: the significance of apoptosis. International Review of Cytology 1980;68:251–306.CrossRef
Taylor, RC, Cullen, SP, Martin, SJ. Apoptosis: controlled demolition at the cellular level. Nature Reviews Molecular and Cellular Biology 2008;9:231–41.CrossRef
Raff, M. Cell suicide for beginners. Nature 1998;396:119–22.CrossRef
Ameisen, JC. On the origin, evolution, and nature of programmed cell death: a timeline of four billion years. Cell Death and Differentiation 2002;9:367–93.CrossRef
Movassagh, M, Foo, RS. Simplified apoptotic cascades. Heart Failure Reviews 2008;13:111–19.CrossRef
Budihardjo, I, Oliver, H, Lutter, M, Luo, X, Wang, X. Biochemical pathways of caspase activation during apoptosis. Annual Review of Cell and Developmental Biology 1999;15:269–90.CrossRef
Debatin, KM. Apoptosis pathways in cancer and cancer therapy. Cancer Immunology and Immunotherapy 2004;53:153–9.CrossRef
Opferman, JT. Apoptosis in the development of the immune system. Cell Death and Differentiation 2008;15:234–42.CrossRef
Letai, AG. Diagnosing and exploiting cancer's addiction to blocks in apoptosis. Nature Reviews Cancer 2008;8:121–32.CrossRef
Sperandio, S, de Belle, I, Bredesen, DE. An alternative, nonapoptotic form of programmed cell death. Proceedings of the National Academy of Sciences USA 2000;97:14 376–81.
Du, C, Fang, M, Li, Y, Li, L, Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33–42.CrossRef
Verhagen, AM, Ekert, PG, Pakusch, M, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43–53.CrossRef
Wang, X. The expanding role of mitochondria in apoptosis. Genes and Development 2001;15:2922–33.
Susin, SA, Lorenzo, HK, Zamzami, N, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441–6.CrossRef
Li, LY, Luo, X, Wang, X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 2001;412:95–9.CrossRef
Saelens, X, Festjens, N, Vande Walle, L, et al. Toxic proteins released from mitochondria in cell death. Oncogene 2004;23:2861–74.CrossRef
Bortner, CD, Hughes, FM., Cidlowski, JA. A primary role for K+ and Na+ efflux in the activation of apoptosis. Journal of Biological Chemistry 1997;272:32 436–42.CrossRefGoogle Scholar
Salvesen, GS, Duckett, CS. IAP proteins: blocking the road to death's door. Nature Reviews Molecular and Cellular Biology 2002;3:401–10.CrossRef
Bratton, SB, Cohen, GM. Apoptotic death sensor: an organelle's alter ego? Trends in Pharmacological Science 2001;22:306–15.
Bratton, SB, Walker, G, Srinivasula, SM, et al. Recruitment, activation and retention of caspases-9 and -3 by Apaf-1 apoptosome and associated XIAP complexes. EMBO Journal 2001;20:998–1009.CrossRef
Youle, RJ, Strasser, A. The BCL-2 protein family: opposing activities that mediate cell death. Nature Reviews Molecular and Cellular Biology 2008;9:47–59.CrossRef
Kuwana, T, Bouchier-Hayes, L, Chipuk, JE, et al. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Molecular Cell 2005;17:525–35.CrossRef
Ito, T, Deng, X, Carr, B, May, WS. Bcl-2 phosphorylation required for anti-apoptosis function. Journal of Biological Chemistry 1997;272:11 671–3.CrossRefGoogle ScholarPubMed
Yin, XM, Oltvai, ZN, Korsmeyer, SJ. BH1 and BH2 domains of Bcl-2 are required for inhibition of apoptosis and heterodimerization with Bax. Nature 1994;369:321–3.CrossRef
Huang, ST, Cidlowski, JA. Phosphorylation status modulates Bcl-2 function during glucocorticoid-induced apoptosis in T lymphocytes. FASEB Journal 2002;16:825–32.CrossRef
Reed, JC. Proapoptotic multidomain Bcl-2/Bax-family proteins: mechanisms, physiological roles, and therapeutic opportunities. Cell Death and Differentiation 2006;13:1378–86.CrossRef
Martinou, JC, Green, DR. Breaking the mitochondrial barrier. Nature Reviews Molecular and Cellular Biology 2001;2:63–7.CrossRef
Martinou, JC, Desagher, S, Antonsson, B. Cytochrome c release from mitochondria: all or nothing. Nature Cell Biology 2000;2:E41–3.
Liu, X, Kim, CN, Yang, J, Jemmerson, R, Wang, X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell 1996;86:147–57.CrossRef
Zou, H, Henzel, WJ, Liu, X, Lutschg, A, Wang, X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 1997;90:405–13.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
Yu, J, Zhang, L. No PUMA, no death: implications for p53-dependent apoptosis. Cancer Cell 2003;4:248–9.CrossRef
Yu, J, Wang, P, Ming, L, Wood, MA, Zhang, L. SMAC/Diablo mediates the proapoptotic function of PUMA by regulating PUMA-induced mitochondrial events. Oncogene 2007;26:4189–98.CrossRef
Yoshida, H. The role of Apaf-1 in programmed cell death: from worm to tumor. Cell Structure and Function 2003;28:3–9.CrossRef
Zermati, Y, Mouhamad, S, Stergiou, L, et al. Nonapoptotic role for Apaf-1 in the DNA damage checkpoint. Molecular Cell 2007;28:624–37.CrossRef
Zou, H, Li, Y, Liu, X, Wang, X. An APAF-1.cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. Journal of Biological Chemistry 1999;274:11 549–56.CrossRefGoogle ScholarPubMed
Wang, HL, Bai, H, Li, Y, Sun, J, Wang, XQ. Rationales for expression and altered expression of apoptotic protease activating factor-1 gene in gastric cancer. World Journal of Gastroenterology 2007;13:5060–4.CrossRefGoogle ScholarPubMed
Fadeel, B, Ottosson, A, Pervaiz, S. Big wheel keeps on turning: apoptosome regulation and its role in chemoresistance. Cell Death and Differentiation 2008;15:443–52.CrossRef
Christoph, F, Hinz, S, Kempkensteffen, C, et al. mRNA expression profiles of methylated APAF-1 and DAPK-1 tumor suppressor genes uncover clear cell renal cell carcinomas with aggressive phenotype. Journal of Urology 2007;178:2655–9.CrossRefGoogle ScholarPubMed
Hinz, S, Kempkensteffen, C, Weikert, S, et al. EZH2 polycomb transcriptional repressor expression correlates with methylation of the APAF-1 gene in superficial transitional cell carcinoma of the bladder. Tumour Biology 2007;28:151–7.CrossRef
Paik, SS, Jang, KS, Song, YS, et al. Reduced expression of Apaf-1 in colorectal adenocarcinoma correlates with tumor progression and aggressive phenotype. Annals of Surgical Oncology 2007;14:3453–9.CrossRef
Johnson, CE, Huang, YY, Parrish, AB, et al. Differential Apaf-1 levels allow cytochrome c to induce apoptosis in brain tumors but not in normal neural tissues. Proceedings of the National Academy of Sciences USA 2007;104:20 820–5.
Mills, K, Daish, T, Harvey, KF, et al. The Drosophila melanogaster Apaf-1 homologue ARK is required for most, but not all, programmed cell death. Journal of Cell Biology 2006;172:809–15.CrossRefGoogle Scholar
Ho, AT, Li, QH, Okada, H, Mak, TW, Zacksenhaus, E. XIAP activity dictates Apaf-1 dependency for caspase 9 activation. Molecular and Cellular Biology 2007;27:5673–85.CrossRef
Cain, K, Bratton, SB, Langlais, C, et al. Apaf-1 oligomerizes into biologically active approximately 700-kDa and inactive approximately 1.4-MDa apoptosome complexes. Journal of Biological Chemistry 2000;275:6067–70.CrossRefGoogle ScholarPubMed
Riedl, SJ, Salvesen, GS. The apoptosome: signalling platform of cell death. Nature Reviews Molecular and Cellular Biology 2007;8:405–13.CrossRef
Pandey, P, Saleh, A, Nakazawa, A, et al. Negative regulation of cytochrome c-mediated oligomerization of Apaf-1 and activation of procaspase-9 by heat shock protein 90. EMBO Journal 2000;19:4310–22.CrossRef
Beere, HM, Wolf, BB, Cain, K, et al. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nature Cell Biology 2000;2:469–75.CrossRef
Schafer, ZT, Kornbluth, S. The apoptosome: physiological, developmental, and pathological modes of regulation. Developmental Cell 2006;10:549–61.CrossRef
Cain, K, Langlais, C, Sun, XM, Brown, DG, Cohen, GM. Physiological concentrations of K+ inhibit cytochrome c-dependent formation of the apoptosome. Journal of Biological Chemistry 2001;276:41 985–90.CrossRefGoogle ScholarPubMed
Karki, P, Seong, C, Kim, JE, et al. Intracellular K(+) inhibits apoptosis by suppressing the Apaf-1 apoptosome formation and subsequent downstream pathways but not cytochrome c release. Cell Death and Differentiation 2007;14:2068–75.CrossRef
Thornberry, NA, Lazebnik, Y. Caspases: enemies within. Science 1998;281:1312–16.CrossRef
Earnshaw, WC, Martins, LM, Kaufmann, SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annual Review of Biochemistry 1999;68:383–424.CrossRef
Degterev, A, Boyce, M, Yuan, J. A decade of caspases. Oncogene 2003;22:8543–67.CrossRef
Slee, EA, Adrain, C, Martin, SJ. Serial killers: ordering caspase activation events in apoptosis. Cell Death and Differentiation 1999;6:1067–74.CrossRef
Rathmell, JC, Thompson, CB. The central effectors of cell death in the immune system. Annual Review of Immunology 1999;17:781–828.CrossRef
Hughes, FM, Bortner, CD, Purdy, GD, Cidlowski, JA. Intracellular K+ suppresses the activation of apoptosis in lymphocytes. Journal of Biological Chemistry 1997;272:30 567–76.CrossRefGoogle ScholarPubMed
Hughes, FM, Cidlowski, JA. Potassium is a critical regulator of apoptotic enzymes in vitro and in vivo. Advances in Enzyme Regulation 1999;39:157–71.CrossRef
Johnson, DE. Programmed cell death regulation: basic mechanisms and therapeutic opportunities. Leukemia 2000;14:1340–4.CrossRef
Igney, FH, Krammer, PH. Death and anti-death: tumour resistance to apoptosis. Nature Reviews Cancer 2002;2:277–88.CrossRef
Deveraux, QL, Roy, N, Stennicke, HR, et al. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO Journal 1998;17:2215–23.CrossRef
Pickart, CM. Mechanisms underlying ubiquitination. Annual Review of Biochemistry 2001;70:503–33.CrossRef
Deveraux, QL, Takahashi, R, Salvesen, GS, Reed, JC. X-linked IAP is a direct inhibitor of cell-death proteases. Nature 1997;388:300–4.CrossRef
Holcik, M, Korneluk, RG. XIAP, the guardian angel. Nature Reviews Molecular and Cellular Biology 2001;2:550–6.CrossRef
Liston, P, Fong, WG, Korneluk, RG. The inhibitors of apoptosis: there is more to life than Bcl2. Oncogene 2003;22:8568–80.CrossRef
Schimmer, AD, Dalili, S, Batey, RA, Riedl, SJ. Targeting XIAP for the treatment of malignancy. Cell Death and Differentiation 2006;13:179–88.CrossRef
Wu, G, Chai, J, Suber, TL, et al. Structural basis of IAP recognition by Smac/DIABLO. Nature 2000;408:1008–12.CrossRef
Huang, Y, Park, YC, Rich, RL, et al. Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell 2001;104:781–90.CrossRef
Shiozaki, EN, Shi, Y. Caspases, IAPs and Smac/DIABLO: mechanisms from structural biology. Trends in Biochemical Sciences 2004;29:486–94.CrossRef
MacFarlane, M, Merrison, W, Bratton, SB, Cohen, GM. Proteasome-mediated degradation of Smac during apoptosis: XIAP promotes Smac ubiquitination in vitro. Journal of Biological Chemistry 2002;277:36 611–16.CrossRefGoogle ScholarPubMed
Hu, S, Yang, X. Cellular inhibitor of apoptosis 1 and 2 are ubiquitin ligases for the apoptosis inducer Smac/DIABLO. Journal of Biological Chemistry 2003;278:10 055–60.CrossRefGoogle ScholarPubMed
Yang, Y, Fang, S, Jensen, JP, Weissman, AM, Ashwell, JD. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli. Science 2000;288:874–7.CrossRef
Suzuki, Y, Nakabayashi, Y, Takahashi, R. Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death. Proceedings of the National Academy of Sciences USA 2001;98:8662–7.CrossRef
Dan, HC, Sun, M, Kaneko, S, et al. Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP). Journal of Biological Chemistry 2004;279:5405–12.CrossRefGoogle Scholar
Hegde, R, Srinivasula, SM, Zhang, Z, et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. Journal of Biological Chemistry 2002;277:432–8.CrossRefGoogle ScholarPubMed
van Loo, G, van Gurp, M, Depuydt, B, et al. The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi interacts with caspase-inhibitor XIAP and induces enhanced caspase activity. Cell Death and Differentiation 2002;9:20–6.CrossRef
Martins, LM, Iaccarino, I, Tenev, T, et al. The serine protease Omi/HtrA2 regulates apoptosis by binding XIAP through a reaper-like motif. Journal of Biological Chemistry 2002;277:439–44.CrossRefGoogle ScholarPubMed
Yang, QH, Church-Hajduk, R, Ren, J, Newton, ML, Du, C. Omi/HtrA2catalytic cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and facilitates caspase activity in apoptosis. Genes and Development 2003;17:1487–96.CrossRef
Srinivasula, SM, Gupta, S, Datta, P, et al. Inhibitor of apoptosis proteins are substrates for the mitochondrial serine protease Omi/HtrA2. Journal of Biological Chemistry 2003;278:31 469–72.CrossRefGoogle ScholarPubMed
Straszewski-Chavez, SL, Visintin, IP, Karassina, N, et al. XAF1 mediates tumor necrosis factor-alpha-induced apoptosis and X-linked inhibitor of apoptosis cleavage by acting through the mitochondrial pathway. Journal of Biological Chemistry 2007;282:13 059–72.CrossRefGoogle ScholarPubMed
Vande Walle, L, Lamkanfi, M, Vandenabeele, P. The mitochondrial serine protease HtrA2/Omi: an overview. Cell Death and Differentiation 2008;15:453–60.CrossRef
Deveraux, QL, Reed, JC. IAP family proteins–suppressors of apoptosis. Genes and Development 1999;13:239–52.CrossRef
Johnson, DE, Gastman, BR, Wieckowski, E, et al. Inhibitor of apoptosis protein hILP undergoes caspase-mediated cleavage during T lymphocyte apoptosis. Cancer Research 2000;60:1818–23.
Levkau, B, Garton, KJ, Ferri, N, et al. xIAP induces cell-cycle arrest and activates nuclear factor-kappaB: new survival pathways disabled by caspase-mediated cleavage during apoptosis of human endothelial cells. Circulation Research 2001;88:282–90.CrossRef
Dohi, T, Okada, K, Xia, F, et al. An IAP-IAP complex inhibits apoptosis. Journal of Biological Chemistry 2004;279:34 087–90.CrossRefGoogle ScholarPubMed
Arora, V, Cheung, HH, Plenchette, S, et al. Degradation of survivin by the X-linked inhibitor of apoptosis (XIAP)-XAF1 complex. Journal of Biological Chemistry 2007;282:26 202–9.CrossRefGoogle ScholarPubMed
Ceballos-Cancino, G, Espinosa, M, Maldonado, V, Melendez-Zajgla, J. Regulation of mitochondrial Smac/DIABLO-selective release by survivin. Oncogene 2007;26:7569–75.CrossRef
Hauser, HP, Bardroff, M, Pyrowolakis, G, Jentsch, S. A giant ubiquitin-conjugating enzyme related to IAP apoptosis inhibitors. Journal of Cell Biology 1998;141:1415–22.CrossRefGoogle ScholarPubMed
Chen, Z, Naito, M, Hori, S, et al. A human IAP-family gene, apollon, expressed in human brain cancer cells. Biochemical and Biophysical Research Communications 1999;264:847–54.CrossRef
Qiu, XB, Markant, SL, Yuan, J, Goldberg, AL. Nrdp1-mediated degradation of the gigantic IAP, BRUCE, is a novel pathway for triggering apoptosis. EMBO Journal 2004;23:800–10.CrossRef
Eckelman, BP, Salvesen, GS, Scott, FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Reports 2006;7:988–94.CrossRef
Hoffmann, EK, Simonsen, LO. Membrane mechanisms in volume and pH regulation in vertebrate cells. Physiological Reviews 1989;69:315–82.CrossRef
O’Neill, WC. Physiological significance of volume-regulatory transporters. American Journal of Physiology 1999;276:C995-C1011.CrossRefGoogle ScholarPubMed
Gomez-Angelats, M, Cidlowski, JA. Cell volume control and signal transduction in apoptosis. Toxicologic Pathology 2002;30:541–51.CrossRef
Lang, F, Busch, GL, Ritter, M, et al. Functional significance of cell volume regulatory mechanisms. Physiological Reviews 1998;78:247–306.CrossRef
Gomez-Angelats, M, Bortner, CD, Cidlowski, JA. Cell volume regulation in immune cell apoptosis. Cell and Tissue Research 2000;301:33–42.CrossRef
Kida, H, Miyoshi, T, Manabe, K, et al. Roles of aquaporin-3 water channels in volume-regulatory water flow in a human epithelial cell line. Journal of Membrane Biology 2005;208:55–64.CrossRefGoogle Scholar
Vu, CC, Bortner, CD, Cidlowski, JA. Differential involvement of initiator caspases in apoptotic volume decrease and potassium efflux during Fas- and UV-induced cell death. Journal of Biological Chemistry 2001;276:37 602–11.CrossRefGoogle ScholarPubMed
Thompson, GJ, Langlais, C, Cain, K, Conley, EC, Cohen, GM. Elevated extracellular [K+] inhibits death-receptor- and chemical-mediated apoptosis prior to caspase activation and cytochrome c release. Biochemical Journal 2001;357:137–45.CrossRef
Storey, NM, Gomez-Angelats, M, Bortner, CD, Armstrong, DL, Cidlowski, JA. Stimulation of Kv1.3 potassium channels by death receptors during apoptosis in Jurkat T lymphocytes. Journal of Biological Chemistry 2003;278:33319–26.CrossRefGoogle ScholarPubMed
Montague, JW, Bortner, CD, Hughes, FM,Cidlowski JA. A necessary role for reduced intracellular potassium during the DNA degradation phase of apoptosis. Steroids 1999;64:563–9.CrossRef
Cidlowski, JA, King, KL, Evans-Storms, RB, et al. The biochemistry and molecular biology of glucocorticoid-induced apoptosis in the immune system. Recent Progress in Hormone Research 1996;51:457–90; discussion 90–1.
Bian, X, Hughes, FM, Huang, Y, Cidlowski, JA, Putney, JW. Roles of cytoplasmic Ca2+ and intracellular Ca2+ stores in induction and suppression of apoptosis in S49 cells. American Journal of Physiology 1997;272:C1241–9.CrossRefGoogle ScholarPubMed
Perez, GI, Maravei, DV, Trbovich, AM, et al. Identification of potassium-dependent and -independent components of the apoptotic machinery in mouse ovarian germ cells and granulosa cells. Biology of Reproduction 2000;63:1358–69.CrossRef
Maeno, E, Ishizaki, Y, Kanaseki, T, Hazama, A, Okada, Y. Normotonic cell shrinkage because of disordered volume regulation is an early prerequisite to apoptosis. Proceedings of the National Academy of Sciences USA 2000;97:9487–92.CrossRef
Maeno, E, Takahashi, N, Okada, Y. Dysfunction of regulatory volume increase is a key component of apoptosis. FEBS Letters 2006;580:6513–17.CrossRef
Lang, F, Gulbins, E, Szabo, I, et al. Cell volume and the regulation of apoptotic cell death. Journal of Molecular Recognition 2004;17:473–80.CrossRefGoogle ScholarPubMed
Friis, MB, Friborg, CR, Schneider, L, et al. Cell shrinkage as a signal to apoptosis in NIH 3T3 fibroblasts. Journal of Physiology 2005;567:427–43.CrossRefGoogle ScholarPubMed
Warr, D. Standard treatment of chemotherapy-induced emesis. Supportive Care in Cancer 1997;5:12–16.CrossRef
Evans-Storms, RB, Cidlowski, JA. Delineation of an antiapoptotic action of glucocorticoids in hepatoma cells: the role of nuclear factor-kappaB. Endocrinology 2000;141:1854–62.CrossRef
Scoltock, AB, Heimlich, G, Cidlowski, JA. Glucocorticoids inhibit the apoptotic actions of UV-C but not Fas ligand in hepatoma cells: direct evidence for a critical role of Bcl-xL. Cell Death and Differentiation 2007;14:840–50.CrossRef
Rhen, T, Cidlowski, JA. Anti-inflammatory action of glucocorticoids–new mechanisms for old drugs. New England Journal of Medicine 2005;353:1711–23.CrossRefGoogle ScholarPubMed
Schwartzman, RA, Cidlowski, JA. Glucocorticoid-induced apoptosis of lymphoid cells. International Archives of Allergy and Immunology 1994;105:347–54.CrossRef
Smets, LA, Salomons, G, van den Berg, J. Glucocorticoid induced apoptosis in leukemia. Advances in Experimental Medicine and Biology 1999;457:607–14.CrossRef
Tsujimoto, K, Ono, T, Sato, M, et al. Regulation of the expression of caspase-9 by the transcription factor activator protein-4 in glucocorticoid-induced apoptosis. Journal of Biological Chemistry 2005;280:27 638–44.CrossRefGoogle ScholarPubMed
Yamamoto, M, Fukuda, K, Miura, N, et al. Inhibition by dexamethasone of transforming growth factor beta1-induced apoptosis in rat hepatoma cells: a possible association with Bcl-xL induction. Hepatology 1998;27:959–66.CrossRef
Herr, I, Ucur, E, Herzer, K, et al. Glucocorticoid cotreatment induces apoptosis resistance toward cancer therapy in carcinomas. Cancer Research 2003;63:3112–20.
Huang, ST, Cidlowski, JA. Glucocorticoids inhibit serum depletion-induced apoptosis in T lymphocytes expressing Bcl-2. FASEB Journal 1999;13:467–76.CrossRef
Lu, J, Quearry, B, Harada, H. p38-MAP kinase activation followed by BIM induction is essential for glucocorticoid-induced apoptosis in lymphoblastic leukemia cells. FEBS Letters 2006;580:3539–44.CrossRef
Bailly-Maitre, B, de Sousa, G, Boulukos, K, Gugenheim, J, Rahmani, R. Dexamethasone inhibits spontaneous apoptosis in primary cultures of human and rat hepatocytes via Bcl-2 and Bcl-xL induction. Cell Death and Differentiation 2001;8:279–88.CrossRef
Hartmann, BL, Geley, S, Loffler, M, et al. Bcl-2 interferes with the execution phase, but not upstream events, in glucocorticoid-induced leukemia apoptosis. Oncogene 1999;18:713–19.CrossRef
Ploner, C, Schmidt, S, Presul, E, et al. Glucocorticoid-induced apoptosis and glucocorticoid resistance in acute lymphoblastic leukemia. Journal of Steroid Biochemistry and Molecular Biology 2005;93:153–60.CrossRefGoogle ScholarPubMed
Geley, S, Fiegl, M, Hartmann, BL, Kofler, R. Genes mediating glucocorticoid effects and mechanisms of their regulation. Reviews of Physiology, Biochemistry and Pharmacology 1996;128:1–97.CrossRef
Planey, SL, Abrams, MT, Robertson, NM, Litwack, G. Role of apical caspases and glucocorticoid-regulated genes in glucocorticoid-induced apoptosis of pre-B leukemic cells. Cancer Research 2003;63:172–8.
Chauhan, D, Auclair, D, Robinson, EK, et al. Identification of genes regulated by dexamethasone in multiple myeloma cells using oligonucleotide arrays. Oncogene 2002;21:1346–58.CrossRef
McColl, A, Michlewska, S, Dransfield, I, Rossi, AG. Effects of glucocorticoids on apoptosis and clearance of apoptotic cells. Scientific World Journal 2007;7:1165–81.CrossRef
McKay, LI, Cidlowski, JA. Molecular control of immune/inflammatory responses: interactions between nuclear factor-kappa B and steroid receptor-signaling pathways. Endocrine Reviews 1999;20:435–59.
Tao, Y, Williams-Skipp, C, Scheinman, RI. Mapping of glucocorticoid receptor DNA binding domain surfaces contributing to transrepression of NF-kappa B and induction of apoptosis. Journal of Biological Chemistry 2001;276:2329–32.CrossRefGoogle ScholarPubMed
Helmberg, A, Auphan, N, Caelles, C, Karin, M. Glucocorticoid-induced apoptosis of human leukemic cells is caused by the repressive function of the glucocorticoid receptor. EMBO Journal 1995;14:452–60.
Tissing, WJ, Meijerink, JP, den Boer, ML, Pieters, R. Molecular determinants of glucocorticoid sensitivity and resistance in acute lymphoblastic leukemia. Leukemia 2003;17:17–25.CrossRef
Erlacher, M, Michalak, EM, Kelly, PN, et al. BH3-only proteins Puma and Bim are rate-limiting for gamma-radiation- and glucocorticoid-induced apoptosis of lymphoid cells in vivo. Blood 2005;106:4131–8.CrossRef
Bouillet, P, Metcalf, D, Huang, DC, et al. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 1999;286:1735–8.CrossRef
Petrella, A, Ercolino, SF, Festa, M, et al. Dexamethasone inhibits TRAIL-induced apoptosis of thyroid cancer cells via Bcl-xL induction. European Journal of Cancer 2006;42:3287–93.CrossRefGoogle ScholarPubMed
Laane, E, Panaretakis, T, Pokrovskaja, K, et al. Dexamethasone-induced apoptosis in acute lymphoblastic leukemia involves differential regulation of Bcl-2 family members. Haematologica 2007;92:1460–9.CrossRef
Mann, CL, Hughes, FM., Cidlowski, JA. Delineation of the signaling pathways involved in glucocorticoid-induced and spontaneous apoptosis of rat thymocytes. Endocrinology 2000;141:528–38.CrossRef
Mann, CL, Bortner, CD, Jewell, CM, Cidlowski, JA. Glucocorticoid-induced plasma membrane depolarization during thymocyte apoptosis: association with cell shrinkage and degradation of the Na(+)/K(+)-adenosine triphosphatase. Endocrinology 2001;142:5059–68.CrossRef
Mann, CL, Cidlowski, JA. Glucocorticoids regulate plasma membrane potential during rat thymocyte apoptosis in vivo and in vitro. Endocrinology 2001;142:421–9.CrossRef
Schaaf, MJ, Cidlowski, JA. Molecular mechanisms of glucocorticoid action and resistance. Journal of Steroid Biochemistry and Molecular Biology 2002;83:37–48.CrossRefGoogle ScholarPubMed
Chae, HJ, Chae, SW, Kang, JS, et al. Dexamethasone suppresses tumor necrosis factor-alpha-induced apoptosis in osteoblasts: possible role for ceramide. Endocrinology 2000;141:2904–13.CrossRef
Lu, NZ, Cidlowski, JA. Translational regulatory mechanisms generate N-terminal glucocorticoid receptor isoforms with unique transcriptional target genes. Molecular Cell 2005;18:331–42.CrossRef
Lu, NZ, Collins, JB, Grissom, SF, Cidlowski, JA. Selective regulation of bone cell apoptosis by translational isoforms of the glucocorticoid receptor. Molecular and Cellular Biology 2007;27:7143–60.CrossRef
Lu, NZ, Cidlowski, JA. The origin and functions of multiple human glucocorticoid receptor isoforms. Annals of the New York Academy of Science 2004;1024:102–23.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.

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.

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.

Available formats
×