<<

Endocrine-Related (1999) 6 45-48 Activation of by oncogenes

S W Lowe

Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA

Abstract p53 is activated by a variety of cellular stresses, including DNA damage, hypoxia, and mitogenic oncogenes, but the extent to which each signal engages p53 as a tumour suppressor remains unknown. In non-immortal cells, the adenovirus E1A oncogene activates p53 to promote , whereas oncogenic ras activates p53 to promote cellular . Inactivation of p53 prevents E1A-induced apoptosis or Ras-induced senescence, allowing proliferation to continue unabated. In each instance, the ability of the oncogene to activate p53 involves the same functions as are required for their transforming potential, implying that p53 activation acts as a fail-safe mechanism to counter hyperproliferative signals. Furthermore, p19ARF is strictly required for oncogene signalling to p53. The fact that ARF - itself a tumour suppressor - acts as an intermediary in this response argues that the tumour suppressor activity of p53 can arise from its ability to eliminate oncogene-expressing cells.

Endocrine-Related Cancer (1999) 6 45-48

Introduction remarkable ability of E1A to enhance radio- and chemosensitivity (Lowe et al. 1993). Although E1A is a The high frequency of p53 in human cancer mitogenic oncogene, p53 acts to limit its oncogenic implies a central role for p53 in tumorigenesis, but the potential. Thus, p53-deficient primary fibroblasts express- signals that trigger p53 in suppressing tumour growth ing E1A are resistant to apoptosis and become oncogeni- remain poorly defined. p53 is a sequence-specific DNA cally transformed (Lowe et al. 1994b). The ability of E1A binding that promotes cell-cycle arrest or to activate p53 is not unique, as c- activates p53 to apoptosis in response to a variety of cellular stresses promote apoptosis (Hermeking & Eick 1994) and (Kastan et al. 1991, Graeber et al. 1994, Linke et al. 1996; oncogenic ras induces p53, leading to premature reviewed by Giaccia & Kastan 1998, Prives 1998). For senescence (Serrano et al. 1997). How oncogenic signals example, p53 levels and activity increase after DNA activate p53 is unknown, and it is conceivable that they damage, in part as a result of de novo phosphorylation and induce p53 by inadvertently damaging DNA. Never- the accompanying conformational changes (Shieh et al. theless, the general involvement of p53 in the cellular 1997, Siliciano et al. 1997). Phosphorylation at serine 15 response to oncogenes raises the possibility that these prevents the interaction of p53 with (Shieh et al. stimuli are relevant triggers of the tumour suppressor 1997), a protein that can downregulate p53 via ubiquitin- activity of p53. mediated proteolysis (Haupt et al. 1997, Kubbutat et al. The INK4a/ARF locus is second only to p53 in the 1997). In principle, failure of p53 to suppress proliferation frequency of its disruption in human cancer (reviewed by after DNA damage might indirectly promote tumour Haber 1997). This locus encodes p16INK4a, a cyclin- development by allowing the growth and survival of cells dependent kinase inhibitor that acts upstream of with mutations (Livingstone et al. 1992, Yin et al. 1992, protein (RB) to promote cell-cycle arrest Griffiths et al. 1997), but whether this provides the (Serrano et al. 1993). Whereas compelling evidence primary driving force for p53 in tumours is indicates that p16INK4a is an important tumour suppressor, unclear. the INK4a/ARF locus encodes a second protein translated Oncogenes can also induce p53, leading to increased in an alternate reading frame, designated p19ARF (Quelle apoptosis or premature senescence (Lowe & Ruley 1993, et al. 1995). p19ARF and p16INK4a are often co-deleted in Hermeking & Eick 1994, Serrano et al. 1997). For tumour cells, but mice lacking p19ARF alone are highly example, the adenovirus E1A oncogene induces p53 and susceptible to cancer (Kamijo et al. 1997; reviewed by promotes apoptosis in primary cells (Debbas & White Haber 1997). p19ARF promotes cell-cycle arrest (Quelle et 1993, Lowe & Ruley 1993, Querido et al. 1997, al. 1995), whereas ARF-null primary mouse embryo Samuelson & Lowe 1997), which is reflected by the fibroblasts (MEFs) do not undergo replicative senescence

Endocrine-Related Cancer (1999) 6 45-48 Online version via http://www.endocrinology.org 1351-0088/99/006-045 © 1999 Society for Endocrinology Printed in Great Britain

Downloaded from Bioscientifica.com at 09/30/2021 05:13:27PM via free access Lowe: Activation of p53 by oncogenes and are transformed by oncogenic ras alone (Kamijo et al. B), c-Jun-N-terminal kinase, Rac, the Rho , and 1997). Thus ARF is a bona fide tumour suppressor. NF-κB (Ridley et al. 1992, Finco & Baldwin 1993, Van Several observations suggest that p19ARF may Aelst et al. 1993, Derijard et al. 1994, Franke et al. 1997, function in a genetic and biochemical pathway that Robinson & Cobb 1997, Rodriguez-Viciana et al. 1997). involves p53. At the level of the organism, the In normal cells, Ras induces premature senescence consequences of deleting p53 and ARF are remarkably through activation of the MAPK cascade - the same similar (Donehower et al. 1992, Kamijo et al. 1997). In pathway important for Ras-induced mitogenesis in either case, the mutant mouse develops normally, but is immortal cells (Lin et al. 1998; see also Zhu et al. 1998). highly predisposed to malignant tumours of a similar In human fibroblasts, Ras ‘effector loop’ mutants that overall pattern and latency. At the cellular level, enforced retain their ability to bind Raf-1 promote premature expression of p19ARF can induce cell-cycle arrest in cells senescence and, among a series of Ras downstream harbouring wild-type, but not mutant, p53 (Kamijo et al. components examined, only activated Raf-1 and MEK 1997). In turn, p19ARF can physically associate with p53 induced p53, , and features of senescence. Moreover, itself, with Mdm2, or with both, to alter p53 levels and a MEK inhibitor (PD98059) prevents Ras-induced cell- activity (Kamijo et al. 1998, Pomerantz et al. 1998, Stott cycle arrest and senescence. In primary murine fibroblasts, et al. 1998, Zhang et al. 1998). Nevertheless, ARF is not activated MEK arrested wild-type MEFs, but forced required for the p53 response following DNA damage, uncontrolled mitogenesis and transformation when ex- because radiation induces G1 arrest in ARF-deficient pressed at comparable levels in p53-/- or INK4aex2-/- fibroblasts and apoptosis in ARF-deficient thymocytes MEFs. The precisely opposite response of normal and (Kamijo et al. 1997, 1998). Thus an understanding of the functionally immortal cells to constitutive - signals that activate p19ARF may help to explain its role as activated protein kinase (MAPK) activation implies that a tumour suppressor, in addition to that of p53. premature senescence acts as a fail-safe mechanism to limit the transforming potential of excessive Ras mitogenic signalling. Thus constitutive MAPK signalling E1A functions that activate p53 activates p53 and p16 as tumour suppressors. To determine how E1A induces p53 and promotes apoptosis, a series of E1A mutants were introduced into Oncogene signalling to p53 primary human and mouse fibroblasts using high-titre ARF recombinant , allowing analysis of E1A in involves p19 genetically normal cells outside the context of adenovirus Using MEFs derived from wild-type, ARF -/-, and p53-/- infection (Samuelson & Lowe 1997). In primary human mice, we investigated whether oncogene signalling to p53 and mouse fibroblasts, E1A mutations that prevented p53 involves p19ARF. E1A induces p19ARF mRNA and pro- accumulation and apoptosis separated into two tein, and the ability of E1A to induce p53 and its trans- complementation groups, which correlated precisely with criptional targets is severely compromised in ARF-null the ability of E1A to associate with either the p300/CRE cells (de Stanchina et al. 1998). ARF -/- cells expressing binding protein (CBP) or RB-related proteins. Furthe- E1A are resistant to apoptosis, albeit to a lesser extent then rmore, E1A mutants incapable of binding RB, p107, and their p53-null counterparts. Re-introduction of p19ARF p130 induce p53 and promote apoptosis in fibroblasts restores p53 accumulation and resensitizes ARF-null cells derived from RB-deficient mice, but not in fibroblasts to apoptotic signals. Like E1A, oncogenic ras induces from mice lacking p107 or p130. Hence, inactivation of p19ARF in wild-type cells. Moreover, Ras is unable to RB, but not p107 or p130, is required for p53 arrest ARF -/- cells, which continue to grow at rates similar accumulation and apoptosis induced by E1A. to p53-/- and INK4aex2-/- MEFs. Ras-induced p53 trans- Interestingly, the regions of E1A that promote apoptosis criptional targets are induced in wild-type, but not ARF -/- are precisely those required for the oncogenic potential of cells (unpublished observations). Thus ARF is required for E1A (Whyte et al. 1988), suggesting that apoptosis and p53 activation and cell-cycle arrest induced by oncogenic p53 accumulation are a response to oncogenic ‘stress’ Ras. Our data, together with those of others (Bates et al. rather than a direct effect of E1A. 1998, Palmero et al. 1998, de Stanchina et al. 1998, Zindy et al. 1998) argue that ARF mediates p53 activation by oncogenes. Ras functions that activate p53 We also examined the Ras signalling pathway(s) respon- sible for p53 activation and premature senescence of non- The ARF-p53 tumour immortal human and mouse fibroblasts. Known Ras suppressor pathway effectors include Raf-1 and components of the MAPK Although p53 integrates signals from multiple stress- cascade, phosphoinositide-3-kinase, Akt (protein kinase induced pathways, its action can, in turn, lead to several

46

Downloaded from Bioscientifica.com at 09/30/2021 05:13:27PM via free access Endocrine-Related Cancer (1999) 6 45-48 possible anti-proliferative responses, depending upon importance in suppressing proliferation of oncogene- cellular context. For example, enforced ARF expression in expressing cells. MEFs induces cell-cycle arrest, but cells overexpressing ARF p19 together with E1A or Myc undergo apoptosis (de Acknowledgements Stanchina et al. 1998, Zindy et al. 1998), which is potentiated by withdrawal of serum survival factors (Evan The author is a Kimmel Scholar and supported by grant et al. 1992, Lowe & Ruley 1993, Lowe et al. 1994a). ARF- CA13106 from the National Cancer Institute (USA). null MEFs are resistant to both E1A- and Myc-induced apoptosis and escape Ras-induced senescence, bypassing References the p53-dependent fail-safe mechanism that normally Bates S, Phillips AC, Clark PA, Stott F, Peters G, Ludwig RL & protects them from these oncogenic signals, and thereby Vousden KH 1998 links the tumour suppressors RB enabling mitogenic oncogenes to function as pure growth and p53 [letter]. Nature 395 124-125. promoters. Debbas M & White E 1993 Wild-type p53 mediates apoptosis by Like p53, ARF has no overt role in normal E1A, which is inhibited by E1B. and Development 7 control or development; hence, the physiological 546-554. circumstances in which it might become activated to DeGregori J, Leone G, Miron A, Jakoi L & Nevins JR 1997 inhibit proliferation or suppress tumour growth are not Distinct roles for E2F proteins in control and apoptosis. Proceedings of the National Academy of Sciences obvious. Our studies suggest that p19ARF can be activated of the USA 94 7245-7250. to suppress proliferation by the E1A oncogene through Derijard B, Hibi M, Wu IH, Barrett T, Su B, Deng T, Karin M & mechanisms that correlate with its binding to both p300/ Davis RJ 1994 JNK1: a protein kinase stimulated by UV light CBP and RB (de Stanchina et al. 1998). These same and Ha-Ras that binds and phosphorylates the c-Jun activation functions are required for E1A to induce p53 and to domain. Cell 76 1025-1037. promote apoptosis in primary fibroblasts (Samuelson & Donehower LA, Harvey M, Slagle BL, McArthur MJ, Lowe 1997) and, remarkably, are also required for the Montgomery CA, Butel JA & Bradley A 1992 Mice deficient transforming potential of E1A (Whyte et al. 1988, 1989). for p53 are developmentally normal but susceptible to Loss of RB greatly increases ARF expression, consistent spontaneous tumours. Nature 356 215-220. with the possibility that ARF is an E2F-responsive Evan GI, Wyllie AH, Gilbert CS, Littlewood TD, Land H, Brooks (DeGregori et al. 1997). Indeed, enforced expression of M, Waters C, Penn LZ & Hancock DC 1992 Induction of E2F-1 induces p19ARF and, conversely, ARF-null cells are apoptosis in fibroblasts by c-myc protein. Cell 69 119-128. resistant to E2F-1-induced apoptosis (Bates et al. 1998, Finco TS & Baldwin AS Jr 1993 Kappa B site-dependent Zindy et al. 1998). In contrast, Ras induces ARF through induction of by diverse inducers of nuclear the MAPK cascade (unpublished observations), but factor kappa B requires Raf-1. Journal of Biological whether this acts as a direct or indirect effect remains to be Chemistry 268 17676-17679. established. In any case, p19ARF function, like that of p53, Franke TF, Kaplan DR, Cantley LC & Toker A 1997 Direct regulation of the Akt proto-oncogene product by depends upon the mutational status of RB, and upon both phosphatidylinositol-3,4-bisphosphate. Science 275 665-668. c-myc and ras proto-oncogene activities. Irrespective of Giaccia AJ & Kastan MB 1998 The complexity of p53 the precise outcome, ARF mutations compromise p53 modulation: emerging patterns from divergent signals. Genes activation and reduce its ability to counter uncontrolled and Development 12 2973-2983. proliferation. Graeber TG, Peterson JF, Tsai M, Monica K, Fornace AJ & Our studies provide additional insights into the role of Giaccia AJ 1994 Hypoxia induces accumulation of p53 p53 in tumour suppression. The predominant view of p53 protein, but activation of a g(1)-phase checkpoint by low- action centres around its ability to function in the cellular oxygen conditions is independent of p53 status. Molecular response to DNA damage. Although this stimulus is and Cellular Biology 14 6264-6277. undoubtedly important for the tumour suppressor activity Griffiths SD, Clarke AR, Healy LE, Ross G, Ford AM, Hooper ML, Wyllie AH & Greaves M 1997 Absence of p53 permits of p53 and may contribute to the outcome of cancer propagation of mutant cells following genotoxic damage. therapy (Lowe et al. 1993, 1994a), p53 activation in Oncogene 14 523-531. response to oncogenes provides an alternative pressure to Haber DA 1997 Splicing into senescence: the curious case of p16 mutate p53 during tumorigenesis (Lowe et al. 1994b, and p19ARF. Cell 91 555-558. Symonds et al. 1994). In this view, p53 normally acts to Haupt Y, Maya R, Kazaz A & Oren M 1997 Mdm2 promotes the limit the consequences of uncontrolled mitogenesis by rapid degradation of p53. Nature 387 296-299. promoting cell-cycle arrest or apoptosis, whereas its loss Hermeking H & Eick D 1994 Mediation of c-myc induced allows proliferation to continue unabated. The fact that apoptosis by p53. Science 265 2091-2093. disruption of the ARF-p53 pathway occurs in the vast Kamijo T, Zindy F, Roussel MF, Quelle DE, Downing JR, majority of human underscores its global Ashmun RA, Grosveld G & Sherr CJ 1997 Tumor suppression

47

Downloaded from Bioscientifica.com at 09/30/2021 05:13:27PM via free access Lowe: Activation of p53 by oncogenes

at the mouse INK4a locus mediated by the alternative reading Rodriguez-Viciana P, Warne PH, Khwaja A, Marte BM, Pappin frame product p19ARF. Cell 91 649-659. D, Das P, Waterfield MD, Ridley A & Downward J 1997 Role Kamijo T, Weber JS, Zambetti G, Zindy F, Roussel MF & Sherr of phosphoinositide 3-OH kinase in cell transformation and CJ 1998 Interactions of the ARF tumor suppressor with p53 control of the actin cytoskeleton by Ras. Cell 89 457-467. and Mdm2. Proceedings of the National Academy of Sciences Samuelson AV & Lowe SW 1997 Selective induction of p53 and of the USA 95 8292-8297. chemosensitivity in RB-deficient cells by E1A mutants unable Kastan MB, Onyekwere O, Sidransky D, Vogelstein B & Craig to bind the RB-related proteins. Proceedings of the National RW 1991 Participation of p53 protein in the cellular response Academy of Sciences of the USA 94 12094-12099. to DNA damage. 51 6304-6311. Serrano M, Hannon GJ & Beach D 1993 A new regulatory motif Kubbutat MH, Jones SN & Vousden KH 1997 Regulation of p53 in cell-cycle control causing specific inhibition of / stability by Mdm2. Nature 387 299-303. CDK4. Nature 366 704-707. Lin AW, Barradas M, Stone JC, van Aelst L, Serrano M & Lowe Serrano M, Lin AW, McCurrach ME, Beach D & Lowe SW 1997 SW 1998 Premature senescence involving p53 and p16 is Oncogenic ras provokes premature cell senescence associated INK4a activated in response to constitutive MEK/MAPK mitogenic with accumulation of p53 and p16 . Cell 88 593-602. signaling. Genes and Development 12 3008-3019. Shieh SY, Ikeda M, Taya Y & Prives C 1997 DNA damage- Linke SP, Clarkin KC, Di Leonardo A, Tsou A & Wahl GM 1996 induced phosphorylation of p53 alleviates inhibition by A reversible, p53-dependent G0/G1 cell cycle arrest induced MDM2. Cell 91 325-334. by ribonucleotide depletion in the absence of detectable DNA Siliciano JD, Canman CE, Taya Y, Sakaguchi K, Appella E & damage. Genes and Development 10 934-947. Kastan MB 1997 DNA damage induces phosphorylation of Livingstone LR, White A, Sprouse J, Livanos E, Jacks T & Tlsty the amino terminus of p53. Genes and Development 11 3471- TD 1992 Altered cell cycle arrest and gene amplification 3481. potential accompany loss of wild-type p53. Cell 70 923-935. de Stanchina E, McCurrach ME, Zindy F, Shieh SY, Ferbeyre G, Samuelson AV, Prives C, Roussel MF, Sherr CJ & Lowe SW Lowe SW & Ruley HE 1993 Stabilization of the p53 tumor 1998 E1A signaling to p53 involves the p19ARF tumor suppressor is induced by adenovirus E1A and accompanies suppressor. Genes and Development 12 2434-2442. apoptosis. Genes and Development 7 535-545. Stott FJ, Bates S, James MC, McConnell BB, Starborg M, Lowe SW, Ruley HE, Jacks T & Housman DE 1993 p53- Brookes S, Palmero I, Ryan K, Hara E, Vousden KH & Peters dependent apoptosis modulates the cytotoxicity of anticancer G 1998 The alternative product from the human CDKN2A agents. Cell 74 954-967. locus, p14ARF, participates in a regulatory feedback loop with Lowe SW, Bodis S, McClatchey A, Remington L, Ruley HE, p53 and MDM2. EMBO Journal 17 5001-5014. Fisher D, Housman DE & Jacks T 1994a p53 status and the Symonds H, Krall L, Remington L, Saenzrobles M, Lowe S, efficacy of cancer therapy in vivo. Science 266 807-810. Jacks T & Van Dyke T 1994 p53-dependent apoptosis Lowe SW, Jacks T, Housman DE & Ruley HE 1994b Abrogation suppresses tumor growth and progression in vivo. Cell 78 703- of oncogene-associated apoptosis allows transformation of 711. p53-deficient cells. Proceedings of the National Academy of Van Aelst L, Barr M, Marcus S, Polverino A & Wigler M 1993 Sciences of the USA 91 2026-2030. Complex formation between RAS and RAF and other protein ARF Palmero I, Pantoja C & Serrano M 1998 p19 links the tumour kinases. Proceedings of the National Academy of Sciences of suppressor p53 to Ras [letter]. Nature 395 125-126. the USA 90 6213-6217. Pomerantz J, Schreiber-Agus N, Liegeois NJ, Silverman A, Whyte P, Ruley HE & Harlow E 1988 Two regions of the Alland L, Chin L, Potes J, Chen K, Orlow I, Lee HW, Cordon- adenovirus early region 1A proteins are required for Cardo C & DePinho RA 1998 The Ink4a tumor suppressor transformation. Journal of 62 257-265. ARF , p19 , interacts with MDM2 and neutralizes Whyte P, Williamson NM & Harlow E 1989 Cellular targets for MDM2’s inhibition of p53. Cell 92 713-723. transformation by the adenovirus E1A proteins. Cell 56 67-75. Prives C 1998 Signaling to p53: breaking the MDM2-p53 circuit. Yin Y, Tainsky MA, Bischoff FZ, Strong LC & Wahl GM 1992 Cell 95 5-8. Wild-type p53 restores cell cycle control and inhibits gene Quelle DE, Zindy F, Ashmun RA & Sherr CJ 1995 Alternative amplification in cells with mutant p53 alleles. Cell 70 937- reading frames of the INK4a encode 948. two unrelated proteins capable of inducing cell cycle arrest. Zhang Y, Xiong Y & Yarbrough WG 1998 ARF promotes Cell 83 993-1000. MDM2 degradation and stabilizes p53: ARF-INK4a locus Querido E, Teodoro JG & Branton PE 1997 Accumulation of p53 deletion impairs both the Rb and p53 tumor suppression induced by the adenovirus E1A protein requires regions pathways. Cell 92 725-734. involved in the stimulation of DNA synthesis. Journal of Zhu J, Woods D, McMahon M & Bishop JM 1998 Senescence of Virology 71 3526-3533. human fibroblasts induced by oncogenic Raf. Genes and Ridley AJ, Paterson HF, Johnston CL, Diekmann D & Hall A Development 12 2997-3007. 1992 The small GTP-binding protein rac regulates growth Zindy F, Eischen CM, Randle DH, Kamijo T, Cleveland JL, Sherr factor-induced membrane ruffling. Cell 70 401-410. CJ & Roussel MF 1998 Myc signaling via the ARF tumor Robinson MJ & Cobb MH 1997 Mitogen-activated protein suppressor regulates p53-dependent apoptosis and kinase pathways. Current Opinions in Cell Biology 9 180-186. immortalization. Genes and Development 12 2424-2433.

48

Downloaded from Bioscientifica.com at 09/30/2021 05:13:27PM via free access