The p53 tumor suppressor participates in ... - Wiley Online Library

11 downloads 0 Views 216KB Size Report
LUCIANA E. GIONO AND JAMES J. MANFREDI*. Department of .... *Correspondence to: James J. Manfredi, Department of Oncolo- .... cated DNA (Taylor et al., 1999). ..... Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW. 1991.
JOURNAL OF CELLULAR PHYSIOLOGY 209:13–20 (2006)

The p53 Tumor Suppressor Participates in Multiple Cell Cycle Checkpoints LUCIANA E. GIONO AND JAMES J. MANFREDI* Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York The process of cell division is highly ordered and regulated. Checkpoints exist to delay progression into the next cell cycle phase only when the previous step is fully completed. The ultimate goal is to guarantee that the two daughter cells inherit a complete and faithful copy of the genome. Checkpoints can become activated due to DNA damage, exogenous stress signals, defects during the replication of DNA, or failure of chromosomes to attach to the mitotic spindle. Abrogation of cell cycle checkpoints can result in death for a unicellular organism or uncontrolled proliferation and tumorigenesis in metazoans (Nyberg et al., 2002). The tumor suppressor p53 plays a critical role in each of these cell cycle checkpoints and is reviewed here. J. Cell. Physiol. 209: 13–20, 2006. ß 2006 Wiley-Liss, Inc.

p53 is a sequence-specific transcriptional regulator that is widely conserved in metazoans. p53 has presumably appeared in evolution as a response in multicellular organisms to the problem of uncontrolled cellular division and the accompanying increased probability of mutation. This notion is supported by the fact that the p53 protein or pathway is mutated or inactivated in most human cancers (Vogelstein et al., 2000). Consistent with this, p53-deficient mice are highly susceptible to spontaneous development of different types of tumors (Donehower et al., 1992; Harvey et al., 1993; Purdie et al., 1994). Most of the tumor suppressor properties of p53 have been ascribed to its ability to function as a transcription factor. The human p53 gene encodes a 393-amino acid protein that is organized into several domains. The globular central domain of p53 (aa 102–292) has been shown to confer sequence-specific DNA binding activity to the protein (Wang et al., 1993). This domain contains the four conserved regions of p53 and the major mutation hot-spots found in tumor-derived mutants (Pavletich et al., 1993). An oligomerization domain has been mapped to residues 324–355 and p53 has been shown to bind DNA as a tetramer, more specifically a dimer of dimers (Friedman et al., 1993; Wang et al., 1994; McLure and Lee, 1998). The defined DNA consensus sequence for p53 binding consists of two repeats of the palindromic sequence 50 -RRRCA/TT/AGYYY-30 , separated by a 0–13-bp spacer (el-Deiry et al., 1992; Funk et al., 1992). With four pentamer repeats of 50 -RRRCW-30 , the organization of the p53 DNA binding site is consistent with p53 binding as a tetramer. The central domain of p53 also coordinates a zinc ion and metal chelators can inhibit DNA binding (Pavletich et al., 1993). Two independent acidic activation domains have been identified at the N-terminus of p53, between residues 1–42 and 43–73 (Fields and Jang, 1990; Venot et al., 1999). These have been shown to interact with factors of the basal transcription machinery such as TFIID (TBP) and TFIIH, as well as the Drosophila TAFII40 and TAFII60, and their human homologs TAFII31 and TAFII70 (Xiao et al., 1994; Lu and Levine, 1995; Thut et al., 1995; Farmer et al., 1996). The oncogenic protein Mdm2, the main negative regulator of p53 stability and activity, also binds to the N-terminal domain of p53 inhibiting its transcriptional activity (Oliner et al., 1993). Finally, a basic regulatory domain ß 2006 WILEY-LISS, INC.

is located at the carboxyl terminus of p53, between residues 363 and 393. This domain has been shown to bind to DNA in a non-specific manner and to negatively regulate specific DNA binding by the central domain (Anderson et al., 1997). Together with the N-terminal domain, the C-terminus of p53 concentrates most of the many post-translational modification sites that contribute to regulate p53 function (Appella and Anderson, 2001) (Fig. 1A). p53 can integrate signals from many different pathways that become activated as a result of a variety of stimuli such as DNA damage, hypoxia, and oncogene activation. In these conditions, p53 triggers various cellular responses that can lead to cell-cycle arrest, senescence, differentiation, DNA repair, apoptosis, and inhibition of angiogenesis. p53-dependent transcriptional regulation of p21, 14-3-3s, Cdc25C, and GADD45 has been proposed to mediate cell growth arrest (el-Deiry et al., 1993; Hermeking et al., 1997). p53 can stimulate DNA repair by inducing the expression of p21, GADD45, and the p48 xeroderma pigmentosum protein (Smith et al., 1994; Li et al., 1994a; Pan et al., 1995; Hwang et al., 1999). Several p53 target genes including Bax, PUMA, Noxa, p53-AIP, PIG3, Fas/APO1, and KILLER/DR5 have been implicated in p53-induced apoptosis (Miyashita and Reed, 1995; Owen-Schaub et al., 1995; Polyak et al., 1997; Wu et al., 1997; Oda et al., 2000a,b; Nakano and Vousden, 2001). Transcriptional upregulation of thrombospondin-1, glioblastomaderived angiogenesis inhibiting factor, and hypoxiainducible factor-1a have been proposed to be involved in the inhibition of angiogenesis by p53 (Dameron et al., 1994; Van Meir et al., 1994; Fukushima et al., 1998; Ravi

Contract grant sponsor: NIH/NCI; Contract grant numbers: CA86001, CA80058; Contract grant sponsor: DOD; Contract grant numbers: W81XWH-05-1-0109, W81XWH-05-1-00305. *Correspondence to: James J. Manfredi, Department of Oncological Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1130, New York, NY 10029. E-mail: [email protected] Received 6 April 2006; Accepted 14 April 2006 DOI: 10.1002/jcp.20689

14

GIONO AND MANFREDI

apoptotic genes. Alternatively, it has been proposed that specific post-translational modifications of p53 or the recruitment by p53 of specific co-activators could determine the different outcomes following cellular stress. Finally, although most of the studies on p53 function have focused on its transcriptional activity, p53 has been shown to play a transcriptional-independent role in the mitochondrial pathway of apoptosis (Mihara et al., 2003). p53 AND THE G1/S CHECKPOINT

Fig. 1. p53 structure and pathways. A: p53 is organized in several domains including two N-terminal transactivation domains (TA1, TA2), a proline-rich domain (PRO), a central DNA binding domain (DBD), and a C-terminal regulatory domain (REG). Multiple residues clustered at the N- and C-terminus of p53 are post-translationally modified by phosphorylation (circles), acetylation and ubiquitination (squares), and sumoylation (triangle). B: p53 is stabilized and activated following oncogene activation and a variety of cellular stress. p14ARF induction and post-translational modification of p53 by multiple kinases ultimately lead to disruption of the interaction of p53 with its E3 ligase Mdm2. Activated p53 transcriptionally regulates the expression of a number of target genes that are involved in cell growth arrest, apoptosis, DNA repair, anti-angiogenesis, and feedback regulation of p53 itself.

et al., 2000). Finally, several p53 target genes play a role in the regulation of the p53 through an auto-regulatory feedback loop: Mdm2, Pirh2, COP1, and cyclin G (Juven et al., 1993; Okamoto and Beach, 1994; Haupt et al., 1997; Okamoto et al., 2002; Leng et al., 2003; Dornan et al., 2004) (Fig. 1B). The responses elicited by p53, mainly growth arrest and apoptosis, are frequently stimuli- and cell typespecific. The way p53 activates one pathway or the other, however, is poorly understood. It has been proposed that the decision may be driven by affinity (Vousden, 2000). According to this model, growth arrest target genes are regulated through high affinity p53 response elements, whereas the promoters of apoptotic target genes contain low affinity response elements or have additional requirements for transcriptional activation (Inga et al., 2002). In this setting, low levels of stress or DNA damage would induce levels of p53 that upregulate high-affinity growth arrest genes. In contrast, when levels of stress hit a cell leaving it in a situation beyond repair, the higher levels of induced p53 activate Journal of Cellular Physiology DOI 10.1002/jcp

The G1/S checkpoint prevents initiation of DNA replication in cells that have damaged DNA. p53 plays a prominent role in the G1/S checkpoint. Expression of p53 ectopically or following DNA damage, arrests cells at the G1/S transition (Kastan et al., 1991; Lin et al., 1992). Cell cycle progression is driven by phosphorylation events mediated by cyclin/cdk complexes. Cyclin D/cdk4, cyclin E/cdk2, and cyclin A/cdk2 complexes sequentially phosphorylate the tumor suppressor pRb and its family members, resulting in release of the E2F family of transcription factors and transactivation of genes involved in DNA replication. These cyclin/cdk complexes are thus the main targets of the effectors of the G1/S checkpoint (Sherr, 1994). Following activation by DNA damage, p53 induces the expression of the cdk inhibitor p21. p21 is induced and localizes to the nucleus in p53 wild-type cells undergoing G1 arrest, but not in cells expressing mutant p53 (el-Deiry et al., 1993, 1994). p21 was originally thought to bind to and inhibit the activity of all G1 cyclin/cdk complexes (Harper et al., 1993). It was later established that although p21 does, in fact, inhibit cdk2 complexes, not only does it not inhibit cyclin D/cdk4, but it promotes the assembly of the complex (LaBaer et al., 1997). Expression of p21, even in the absence of p53, was sufficient to induce growth arrest at the G1 and G2 phases (Rousseau et al., 1999). Furthermore, p21 is required for efficient G1 arrest. Mouse embryo fibroblasts derived from p21-deficient mice have an impaired DNA damage induced-G1 arrest (Brugarolas et al., 1995; Deng et al., 1995). Finally, deletion of p21 by homologous recombination in the colon cancer cell line HCT116 completely abrogated the G1 arrest following DNA damage (Waldman et al., 1995). Despite the clear role of p53 in the establishment of the G1 arrest, there is evidence that the first delay following DNA damage is p53-independent. This is consistent with the observation of a transient decrease in cyclin E/A-cdk2 activities after irradiation in the absence of p53 (D’Anna et al., 1997). Evidence has been reported that the phosphatase Cdc25A mediates this delay. Cdc25A regulates the activity of cdk2 by removing inhibitory phosphates near its amino terminus. A DNA damage-activated ATM/Chk2 pathway was shown to lead to phosphorylation of Cdc25A, resulting in its ubiquitin- and proteasome-dependent degradation, and consequently, inhibition of cdk2 (Mailand et al., 2000). Moreover, cyclin D1 has been shown to be degraded following genotoxic stress, and this was required for initiation of the G1 arrest. In addition, upon cyclin D1 degradation, p21 is released and redistributed to cdk2 complexes which are then inhibited by p21 (Agami and Bernards, 2000). The second and p53-dependent G1 delay would take place later after DNA damage, since it requires gene transcription, and it also involves the p53 activating kinases ATM, ATR, and Chk2 (Fig. 2). It is proposed that the p53-dependent arrest in G1 extends the delay initiated by Cdc25A, providing the cell with

p53 AND CHECKPOINTS

15

Fig. 2. G1/S and G2/M checkpoints. Sequential activation of the G1/S cyclins D, E, and A result in hyperphosphorylation of pRb, which then releases E2F, allowing transcription of genes required for entry into S-phase. Cell growth arrest at the G1/S border is initiated by destruction of cyclin D1 and the cdk2 activator Cdc25A. p53 maintains this arrest by inducing the expression of the cdk inhibitor p21 that blocks this progression by inhibiting cdk2 complexes. The G2/M transition is driven by the cdc2/cyclin B complex. Following cellular

stress, the G2/M arrest is initiated by phosphorylation of the cdc2/Bactivating phosphatase, Cdc25C, which is then sequestrated in the cytoplasm by 14-3-3 proteins. A number of p53 target genes ensure the maintenance of this arrest: p21 inhibits the cdc2/B complex, as well as its activating kinase CAK; GADD45 promotes dissociation of the cdc2/ B complex; 14-3-3 s sequesters cdc2/B in the cytoplasm; Cdc25C expression is repressed by p53.

sufficient time to repair the damaged DNA (Nyberg et al., 2002). The reversibility of the p53-induced G1 arrest, however, appears to be cell-type specific (Di Leonardo et al., 1994; Agarwal et al., 1995; Carr, 2000).

significant proportion of p53-null cells continued to enter mitosis, although after a much longer duration of the S phase. These mitotic cells, scored by high levels of phosphorylated H1b, contained less than a 4N DNA complement. Based on these results, p53 was proposed to ensure that cells do not enter mitosis with unreplicated DNA (Taylor et al., 1999). Downregulation of Cdc7, a kinase that is involved in the firing of replication origins, arrested normal human fibroblasts in S-phase, although without strong activation of checkpoint pathways, measured by low levels of active ATM/ATR and Chk1/Chk2 kinases. Despite undetectable phosphorylation at Ser15, p53 expression was induced, followed by p21 upregulation. When the cells arrested in S-phase were subsequently transfected with p53 siRNA, DNA replication resumed in a fraction of cells with subsequent caspase activation and apoptotis. These results suggest that p53 is required for maintenance of S-phase arrest induced by Cdc7 downregulation (Montagnoli et al., 2004). Furthermore, inactivation of ATR (by expression of a catalytically inactive mutant) and p53 (by expression of Mdm2 or HPV E6) synergized at promoting premature chromatin condensation, a hallmark of mammalian cells that enter mitosis before completing DNA replication (Nghiem et al., 2002).

p53 AND THE S-PHASE CHECKPOINT

The S-phase checkpoint actually involves three distinct modes that despite some differences, share components and are mutually coordinated. First is the replication checkpoint that is activated by stalling of replication forks due to nucleotide depletion or polymerase inhibition. The second is the intra-S-phase checkpoint that is activated when damaged DNA is detected during S-phase, outside of the replicons, and is therefore replication-independent. Finally, the S/M checkpoint prevents division before the cell has completely duplicated its genome (Bartek et al., 2004). In addition, stalled replication forks frequently collapse, resulting in DNA damage. A role for p53 at the S-phase checkpoint has been argued in several reports. Cells differing in their p53 status were treated with inhibitors of DNA replication, aphidicolin (which blocks DNA polymerase), hydroxyurea (HU, which inhibits ribonucleotide synthase). In contrast to p53 expressing cells, a Journal of Cellular Physiology DOI 10.1002/jcp

16

GIONO AND MANFREDI

With the frequent occurrence of stalled replication forks and double strand breaks during DNA replication, it has been suggested that a transient delay in S-phase with the absence of a mechanism for maintenance could actually be beneficial to the cells. Non-error prone DNA repair by homologous recombination requires the presence of sister chromatids and therefore can only occur during S or G2 phases. Accordingly, a prolonged blockade in S phase would limit the possibility of efficient repair. In addition, it has been suggested that during a prolonged S-phase arrest, replication origins can regain competence, possibly leading to rereplication (Bartek and Lukas, 2001). Consistent with this idea, downregulation of cdk2 triggered S-phase arrest, accompanied by p53 and p21 induction and ATM/ATR activation. While some cells died of apoptosis, cdk2 inactivation lead to increased loading of MCM complexes onto chromatin, and cells acquired a >4N DNA content, indicating rereplication. However, arguing for a role of p53 at the S-phase checkpoint, this phenotype was enhanced when cdk2 inactivation was combined with downregulation of p53 (Zhu et al., 2004). Finally, the combination of activated p53 and E2F-1 found during S phase, can result in apoptosis (Bartek and Lukas, 2001; Gottifredi et al., 2001). This possibility was raised by studies where p53 was shown to become stabilized after HU. However, in contrast to irradiation, HU-induced p53 appeared to be transcriptionally inactive, as it failed to upregulate a number of target genes that are usually induced after irradiation. Moreover, in contrast to irradiation, p53 activation and Ser15 phosphorylation induced by HU did not require ATM (Gottifredi et al., 2001). More recently, a new p53 isoform was described that results from alternative splicing at exons 7 to 9. Despite lacking 39 residues of the core domain, this Dp53 isoform was shown to bind to promoters and transactivate a subset of p53 target genes, in particular the p21 and 14-3-3s growth arrest genes, but not the apoptotic PIG3 gene. Even more interesting is the fact that Dp53 is active only during Sphase in G1/S UV irradiated cells, when full-length p53 is inactive. The data presents a scenario where Dp53 and p53 work in a consecutive and almost mutually exclusive manner (Rohaly et al., 2005). The appearance of this apparently apoptosis-defective isoform of p53 could reconcile the idea that active p53 during S-phase could be deleterious to the cell due to induction of apoptosis, with the several reports that argue in favor of a role for p53 at the S-phase checkpoint. The novelty of the Dp53 isoform, combined with the unprecedented circumstances that surround it, although very promising and exciting, ask for more extensive and careful examination of the involvement of p53 at the S-phase checkpoint (Prives and Manfredi, 2005). p53 AND THE G2/M CHECKPOINT

G2/M progression is driven by the maturationpromoting factor MPF, a complex of cyclin B1/cdc2. Cyclin B1 and cdc2 accumulate and associate in the cytoplasm during G2. The complex is kept inactive by phosphorylation of cdc2 at Thr14 and Tyr15. The entry into mitosis is triggered by translocation of the complex to the nucleus and activation by dephosphorylation by the Cdc25 phosphatases. Cdc2 is further activated by phosphorylation at Thr161 by the cdk-activating kinase CAK (Nurse, 1990). DNA damage activates checkpoint mechanisms that arrest cells at G1/S, but also at G2/M. The critical cyclin B1/cdc2 complex is the main target of the G2 checkpoint Journal of Cellular Physiology DOI 10.1002/jcp

pathways that involve activation of the ATM and ATR and their downstream substrates Chk1 and Chk2 (Nyberg et al., 2002). In addition to the G1/S arrest, p53 has been shown to participate in the G2/M checkpoint (Stewart et al., 1995). Similarly to what has been proposed for the G1 arrest, p53 is involved in the maintenance rather than the initiation of the G2 arrest. For example, wild-type p53 HCT116 cells were able to sustain a G2 arrest following IR, whereas the isogenic p53-null derivative arrested initially in G2 but then escaped and entered mitosis (Bunz et al., 1998). Several p53 target genes were shown to play a role in the p53-induced G2 arrest (Fig. 2). Cdc25C, the phosphatase that promotes mitosis, is inhibited after DNA damage through phosphorylation at Ser216 by Chk1, Chk2, and other kinases. This modification creates a binding site for 14-3-3 proteins and this association sequesters Cdc25C in the cytoplasm and/or inhibits its phosphatase activity (Lin et al., 1992; Lopez-Girona et al., 2001). Cdc25C has been shown to be a target of repression by p53 following DNA damage (St Clair et al., 2004). In addition, a particular 14-3-3 isoform, 14-3-3s, is also a p53 target gene and is upregulated following DNA damage (Hermeking et al., 1997). 14-3-3s prevents proper nuclear localization of cyclin B1/cdc2 after DNA damage. Further, deletion of 14-3-3s by homologous recombination in HCT116 cells, resulted in cell death in response to irradiation (Chan et al., 1999). Although it had been suggested that p21 is a poor inhibitor of cdc2 in vitro, compared to other cyclindependent kinases (Harper et al., 1995), p21 was also implicated in the sustained G2 arrest, through inhibition of the cyclin B1/cdc2 complex activity (Bunz et al., 1998). Early studies had reported that p21 mRNA has a bimodal expression in cycling cells, peaking in G1, decreasing during S-phase, and accumulating again in G2 (Li et al., 1994b). Consistent with that, p21 was detected in the nucleus in late G2, and this correlated with a delayed entry into mitosis that was not observed in cells lacking p53 or p21. At this stage, p21 was found associated with inactive cyclin A/cdk2 complexes, and to a lesser extent those containing cyclin B1. It was proposed that p21, by inducing a pause before the onset of mitosis, could facilitate G2/M checkpoint implementation (Dulic et al., 1998). Using p53-null cells expressing wild-type p21, or a p21 mutant incapable of binding PCNA, it was shown that the ability of p21 to interact with PCNA is required for a sustained G2 arrest. p21 coimmunoprecipitated with the cyclin B1/cdc2 complex and PCNA in cells arrested in G2 after DNA damage. In contrast, in cells with no p21, the p21 mutant, or in the absence of DNA damage, PCNA was found associated with Cdc25C. Because the binding of PCNA to Cdc25C or p21 appeared to be mutually exclusive, the authors hypothesized that p21 may contribute to the G2 arrest by preventing the interaction of Cdc25C with the cyclin B1/cdc2 complex (Ando et al., 2001). Also, p21 was reported to retain cyclin B1/cdc2 complexes in the nucleus, preventing its activation by Cdc25C and recruitment to the centrosomes (Charrier-Savournin et al., 2004). Another study, however, showed that p21 inhibits the cyclin B1/complex not by preventing dephosphorylation at the inhibitory Thr14/Tyr15, but by preventing activation of cdc2 by CAK (Smits et al., 2000). In contrast to these observations, a novel positive regulation of the cyclin B1/cdc2 complex by p21 has been described. A transient hyperphosphorylated form of p21

17

p53 AND CHECKPOINTS

was detected in the nucleus in early G2, preceding the phosphorylation of Cdc25C, dephosphorylation of cdc2 at Thr14/Tyr15, and the nuclear accumulation of cyclin B1. The authors propose a model in which p21, following translocation to the nucleus during S-phase, becomes phosphorylated at Thr57, possibly by cdk2. The presence of p21 correlated with increased cyclin B1associated kinase activity. Using cell extracts and working with mutants, examination of the formed complexes by immunoprecipitation revealed that hyperphosphorylation of p21 facilitates binding of p21 to Ser126-phosphorylated cyclin B1 and promotes the formation of the cyclin B1/cdc2 kinase complex (Dash and El-Deiry, 2005). Another p53 target gene involved in the G2 arrest is GADD45. GADD45 has been shown to interact with PCNA (Smith et al., 1994; Kearsey et al., 1995). GADD45 also interacts with cdc2 and inhibits its kinase activity, presumably by causing dissociation of the cyclin B1/cdc2 complex (Zhan et al., 1999). Further, induction of GADD45 resulted in G2 arrest associated with increased cytoplasmic cyclin B1 (Jin et al., 2002), and overexpression of cyclin B1 and Cdc25C abrogated GADD45-induced G2. Of note, induction of G2 arrest by GADD45 requires the presence of wild-type p53 and depends on the type of DNA damage. Human cells with downregulated GADD45 expression or lymphocytes from GADD45 knock-out mice failed to arrest in G2 following UV or the radiomimetic MMS, but had an intact response after irradiation (Wang et al., 1999). p53 AND THE SPINDLE ASSEMBLY CHECKPOINT VERSUS A G1 TETRAPLOID CHECKPOINT

The spindle assembly checkpoint ensures that cells will not enter anaphase, where chromosome segregation occurs, until all chromosomes are aligned at the equator and attached to the microtubules of the mitotic spindle. Through activation of Mad1/2 and other proteins, spindle defects or the presence of unattached kinetochores will result in inactivation of the anaphase promoting complex or cyclosome, APC/C. This complex regulates the degradation by the proteasome of a number of proteins that regulate sister chromatid cohesion, and spindle elongation, as well as cyclin B1. Degradation of these proteins is a prerequisite for progression of mitosis (Hardwick, 1998). However, even in the absence of a functional mitotic spindle and chromosome segregation, MPF can undergo spontaneous degradation, and cells can enter an apparent state of interphase, a process known as adaptation or mitotic slippage (Rieder and Maiato, 2004). Failure of the mitotic spindle checkpoint can lead to cells exiting mitosis and entering the next S phase with a 4N DNA content, resulting in endoreduplication. p53 was initially believed to control the mitotic spindle checkpoint because p53-deficient fibroblasts failed to arrest after treatment with microtubule inhibitors, undergoing a second round of DNA synthesis in the absence of division and becoming octaploid (Cross et al., 1995; Di Leonardo et al., 1997). In addition, it was thought that spindle components themselves could be under regulation of p53, since p53 binds to centrosomes (Brown et al., 1994) and p53/ mouse embryo fibroblasts contain an abnormal number of centrosomes (Fukasawa et al., 1996). More detailed examination showed that expression of p53 occurred after cells had exited mitosis and progressed to a G1-like state (Minn et al., 1996). The time spent in mitosis by cells treated Journal of Cellular Physiology DOI 10.1002/jcp

with nocodazole was the same, regardless of their p53 status. In addition, there were strong similarities between the p53-dependent checkpoint following nocodazole and the p53-dependent G1 arrest following irradiation: requirement of p21, high cyclin E and low cyclin B1 protein levels, hypophosphorylated pRb, and flat morphology (Lanni and Jacks, 1998). The nature of the signal that causes G1 arrest due to spindle malfunction is unclear. A p53-dependent G1 arrest with a 4N DNA content was also observed when actin assembly was inhibited, preventing cytokinesis even when spindle formation and chromosome segregation could proceed normally (Andreassen et al., 2001). This suggests that it is tetraploidy itself, and not the activation of the mitotic checkpoint, which causes G1 arrest. However, another study argues that a prolonged mitotic arrest is required for tetraploid G1 arrest. HCT116 cells where Mad1/2 function was diminished by downregulation or by a dominant negative approach had a compromised spindle checkpoint following nocodazole treatment. They were able to escape mitosis and endoreduplicate despite expressing p53. However, when mitotic Mad1/2-deficient cells were forced into a prolonged mitotic arrest by treatment with a proteasome inhibitor, a reduced proportion of cells undergoing endoreduplication was observed. These results in turn suggest that not only functional p53 but also an intact spindle assembly checkpoint is required for the 4N G1 arrest (Vogel et al., 2004). These results show that p53 plays a critical role in preventing aneuploidy by blocking endoreduplication of tetraploid cells that result from mitotic failure. ROLE OF p53 IN DNA REPAIR

In addition to growth arrest and apoptosis, the ability of p53 to regulate DNA repair after genotoxic stress represents another mechanism by which p53 helps maintaining genomic integrity. p53-deficient cells have impaired nucleotide-excision repair (NER). More specifically, the global genomic repair (GGR) rather than the transcription-coupled repair (TCR) appears to be affected by loss of p53 (Ford and Hanawalt, 1995). The participation of p53 in DNA repair involves transcriptional regulation as well as direct interaction with mediators of DNA repair. For example, in addition to binding and inhibiting cyclin-dependent kinases, the p53 target p21 has been shown to bind to PCNA, the DNA polymerase d loading, and processivity factor. Through this association, p21 was reported to block replication (Waga et al., 1994), but not PCNA-dependent NER (Li et al., 1994a). However, the role of p21 in DNA repair remains controversial (Adimoolam et al., 2001; Stivala et al., 2001; Wani et al., 2002; Bendjennat et al., 2003). As mentioned earlier, GADD45 is another p53regulated gene that also binds to PCNA (Smith et al., 1994). GADD45 stimulated DNA excision repair in vitro, and downregulation of GADD45 affected DNA repair and sensitized cells to killing by UV and genotoxic drugs (Smith et al., 1996). Mouse embryo fibroblasts from p53 and GADD45 knock-out mice presented impaired GGR, whereas loss of p21 had only a marginal effect (Smith et al., 2000). The xeroderma pigmentosum p48 gene is transactivated by p53 and shown to be required for efficient GGR as well (Hwang et al., 1999). Regulation of deoxynucleotide triphosphate pools appears to be characteristic of DNA repair, since their limited availability increases damage sensitivity (Nyberg et al., 2002). For instance, the ribonucleotide reductase-like protein p53R2 was reported to be induced

18

GIONO AND MANFREDI

by p53 after DNA damage by ionizing radiation, UV, and DNA damaging agents. Moreover, inhibition of p53R2 reduced DNA repair and cell survival after genotoxic stress. (Nakano et al., 2000; Tanaka et al., 2000). p53 can directly interact with proteins involved in DNA repair. p53 binds to replication protein A (RPA), a single-stranded DNA binding protein involved in DNA replication, homologous recombination (HR), and NER (Janus et al., 1999). The association of p53 and RPA is disrupted after UV, and the released RPA can then participate in NER (Abramova et al., 1997). p53 can also bind to members of the TFIIH complex that couples transcription with NER. These include XPB, XPC, and the cdk7/cyclin H/p36 (CAK) complex. p53 is regulated by CAK through phosphorylation (Ko et al., 1997), linking the DNA repair machinery with p53 activation. The role of these interactions, however, is not clear. It is possible that they could help recruiting p53 to sites of DNA repair or transcription (Janus et al., 1999). Also, p53 inhibits the helicase activity of XPB, XPC and the repair protein Cockayne Syndrome B protein (CSB), but the mechanistic role of that has yet to be found. In addition to binding to DNA repair proteins, p53 interacts directly with a variety of DNA structures. p53 binds to double-stranded and single-stranded DNA in a non-specific way, to ends of double-strand breaks, to Holliday junctions, and to DNA bulges caused by DNA mismatches. Furthermore, p53 has been reported to possess DNA-reannealing and DNA end-joining activities, the ability to promote DNA strand transfer, and a 30 to 50 exonuclease activity. In particular, studies on p53 exonuclease activity raised some interesting issues. The C-terminus of p53 appears to inhibit its exonuclease activity (Janus et al., 1999). Furthermore, p53 appears to provide proof-reading capacity to DNA polymerase aprimase complex and to HIV1 reverse transcriptase in vitro (Bakhanashvili, 2001; Melle and Nasheuer, 2002). Although still speculative, it is tempting to hypothesize that in its non-induced state in the absence of DNA damage p53 could participate in maintaining genome integrity, perhaps by enhancing the fidelity of DNA synthesis (Albrechtsen et al., 1999). CONCLUSIONS

After so many years of viewing p53 almost exclusively as a stress-activated sequence-specific transcriptional regulator, more and more faces of this versatile protein are revealed: an increasingly complex regulation, a growing number of interacting proteins and target genes, some unexpected functions. For instance, the possibility of p53 performing a maintenance role in the absence of stress is still an open question. Technical advances and increased understanding of the mechanisms set in place by p53 will certainly be of great value in optimizing cancer therapies. ACKNOWLEDGMENTS

A review examining an aspect of p53 biology is always difficult, given the extensive published literature. The authors apologize for selective citations which, by necessity, resulted in their failure to acknowledge all those who contributed to our current understandings. We thank Luis Carvajal, Dana Lukin, Anthony Mastropietro, Sejal Patel, Lois Resnick-Silverman, and Shohreh Varmeh-Ziaie for helpful discussions and their support. The research in the authors’ laboratory was supported by grants from the National Cancer Institute (CA86001 and CA80058) and the Department of Defense Prostate Cancer Research Program Journal of Cellular Physiology DOI 10.1002/jcp

(W81XWH-05-1-0109) and Breast Cancer Research Program (W81XWH-05-1-00305). LITERATURE CITED Abramova NA, Russell J, Botchan M, Li R. 1997. Interaction between replication protein A and p53 is disrupted after UV damage in a DNA repair-dependent manner. Proc Natl Acad Sci USA 94(14):7186–7191. Adimoolam S, Lin CX, Ford JM. 2001. The p53-regulated cyclin-dependent kinase inhibitor, p21 (cip1, waf1, sdi1), is not required for global genomic and transcription-coupled nucleotide excision repair of UV-induced DNA photoproducts. J Biol Chem 276(28):25813–25822. Agami R, Bernards R. 2000. Distinct initiation and maintenance mechanisms cooperate to induce G1 cell cycle arrest in response to DNA damage. Cell 102(1): 55–66. Agarwal ML, Agarwal A, Taylor WR, Stark GR. 1995. p53 controls both the G2/M and the G1 cell cycle checkpoints and mediates reversible growth arrest in human fibroblasts. Proc Natl Acad Sci USA 92(18):8493–8497. Albrechtsen N, Dornreiter I, Grosse F, Kim E, Wiesmuller L, Deppert W. 1999. Maintenance of genomic integrity by p53: Complementary roles for activated and non-activated p53. Oncogene 18(53):7706–7717. Anderson ME, Woelker B, Reed M, Wang P, Tegtmeyer P. 1997. Reciprocal interference between the sequence-specific core and nonspecific C-terminal DNA binding domains of p53: Implications for regulation. Mol Cell Biol 17(11): 6255–6264. Ando T, Kawabe T, Ohara H, Ducommun B, Itoh M, Okamoto T. 2001. Involvement of the interaction between p21 and proliferating cell nuclear antigen for the maintenance of G2/M arrest after DNA damage. J Biol Chem 276(46):42971–42977. Andreassen PR, Lohez OD, Lacroix FB, Margolis RL. 2001. Tetraploid state induces p53-dependent arrest of nontransformed mammalian cells in G1. Mol Biol Cell 12(5):1315–1328. Appella E, Anderson CW. 2001. Post-translational modifications and activation of p53 by genotoxic stresses. Eur J Biochem 268(10):2764–2772. Bakhanashvili M. 2001. p53 enhances the fidelity of DNA synthesis by human immunodeficiency virus type 1 reverse transcriptase. Oncogene 20(52):7635– 7644. Bartek J, Lukas J. 2001. Mammalian G1- and S-phase checkpoints in response to DNA damage. Curr Opin Cell Biol 13(6):738–747. Bartek J, Lukas C, Lukas J. 2004. Checking on DNA damage in S phase. Nat Rev Mol Cell Biol 5(10):792–804. Bendjennat M, Boulaire J, Jascur T, Brickner H, Barbier V, Sarasin A, Fotedar A, Fotedar R. 2003. UV irradiation triggers ubiquitin-dependent degradation of p21(WAF1) to promote DNA repair. Cell 114(5):599–610. Brown CR, Doxsey SJ, White E, Welch WJ. 1994. Both viral (adenovirus E1B) and cellular (hsp 70, p53) components interact with centrosomes. J Cell Physiol 160(1):47–60. Brugarolas J, Chandrasekaran C, Gordon JI, Beach D, Jacks T, Hannon GJ. 1995. Radiation-induced cell cycle arrest compromised by p21 deficiency. Nature 377(6549):552–557. Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW, Vogelstein B. 1998. Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science 282(5393):1497–1501. Carr AM. 2000. Cell cycle. Piecing together the p53 puzzle. Science 287(5459): 1765–1766. Chan TA, Hermeking H, Lengauer C, Kinzler KW, Vogelstein B. 1999. 14-33Sigma is required to prevent mitotic catastrophe after DNA damage. Nature 401(6753):616–620. Charrier-Savournin FB, Chateau MT, Gire V, Sedivy J, Piette J, Dulic V. 2004. p21-Mediated nuclear retention of cyclin B1-Cdk1 in response to genotoxic stress. Mol Biol Cell 15(9):3965–3976. Cross SM, Sanchez CA, Morgan CA, Schimke MK, Ramel S, Idzerda RL, Raskind WH, Reid BJ. 1995. A p53-dependent mouse spindle checkpoint. Science 267(5202):1353–1356. D’Anna JA, Valdez JG, Habbersett RC, Crissman HA. 1997. Association of G1/S-phase and late S-phase checkpoints with regulation of cyclindependent kinases in Chinese hamster ovary cells. Radiat Res 148(3):260– 271. Dameron KM, Volpert OV, Tainsky MA, Bouck N. 1994. Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science 265(5178):1582– 1584. Dash BC, El-Deiry WS. 2005. Phosphorylation of p21 in G2/M promotes cyclin BCdc2 kinase activity. Mol Cell Biol 25(8):3364–3387. Deng C, Zhang P, Harper JW, Elledge SJ, Leder P. 1995. Mice lacking p21CIP1/ WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82(4):675–684. Di Leonardo A, Linke SP, Clarkin K, Wahl GM. 1994. DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts. Genes Dev 8(21):2540–2551. Di Leonardo A, Khan SH, Linke SP, Greco V, Seidita G, Wahl GM. 1997. DNA rereplication in the presence of mitotic spindle inhibitors in human and mouse fibroblasts lacking either p53 or pRb function. Cancer Res 57(6):1013– 1019. Donehower LA, Harvey M, Slagle BL, McArthur MJ, Montgomery CA, Jr., Butel JS, Bradley A. 1992. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356(6366):215–221. Dornan D, Wertz I, Shimizu H, Arnott D, Frantz GD, Dowd P, O’Rourke K, Koeppen H, Dixit VM. 2004. The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature 429(6987):86–92. Dulic V, Stein GH, Far DF, Reed SI. 1998. Nuclear accumulation of p21Cip1 at the onset of mitosis: A role at the G2/M-phase transition. Mol Cell Biol 18(1):546–557. el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B. 1992. Definition of a consensus binding site for p53. Nat Genet 1(1):45–49. el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B. 1993. WAF1, a potential mediator of p53 tumor suppression. Cell 75(4):817–825.

p53 AND CHECKPOINTS el-Deiry WS, Harper JW, O’Connor PM, Velculescu VE, Canman CE, Jackman J, Pietenpol JA, Burrell M, Hill DE, Wang Y, et al. 1994. WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 54(5):1169–1174. Farmer G, Colgan J, Nakatani Y, Manley JL, Prives C. 1996. Functional interaction between p53, the TATA-binding protein (TBP), andTBP-associated factors in vivo. Mol Cell Biol 16(8):4295–4304. Fields S, Jang SK. 1990. Presence of a potent transcription activating sequence in the p53 protein. Science 249(4972):1046–1049. Ford JM, Hanawalt PC. 1995. Li-Fraumeni syndrome fibroblasts homozygous for p53 mutations are deficient in global DNA repair but exhibit normal transcription-coupled repair and enhanced UV resistance. Proc Natl Acad Sci USA 92(19):8876–8880. Friedman PN, Chen X, Bargonetti J, Prives C. 1993. The p53 protein is an unusually shaped tetramer that binds directly to DNA. Proc Natl Acad Sci USA 90(8):3319–3323. Fukasawa K, Choi T, Kuriyama R, Rulong S, Vande Woude GF. 1996. Abnormal centrosome amplification in the absence of p53. Science 271(5256):1744– 1747. Fukushima Y, Oshika Y, Tsuchida T, Tokunaga T, Hatanaka H, Kijima H, Yamazaki H, Ueyama Y, Tamaoki N, Nakamura M. 1998. Brain-specific angiogenesis inhibitor 1 expression is inversely correlated with vascularity and distant metastasis of colorectal cancer. Int J Oncol 13(5):967–970. Funk WD, Pak DT, Karas RH, Wright WE, Shay JW. 1992. A transcriptionally active DNA-binding site for human p53 protein complexes. Mol Cell Biol 12(6): 2866–2871. Gottifredi V, Shieh S, Taya Y, Prives C. 2001. p53 accumulates but is functionally impaired when DNA synthesis is blocked. Proc Natl Acad Sci USA 98(3):1036– 1041. Hardwick KG. 1998. The spindle checkpoint. Trends Genet 14(1):1–4. Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge SJ. 1993. The p21 Cdkinteracting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 75(4):805–816. Harper JW, Elledge SJ, Keyomarsi K, Dynlacht B, Tsai LH, Zhang P, Dobrowolski S, Bai C, Connell-Crowley L, Swindell E, et al. 1995. Inhibition of cyclin-dependent kinases by p21. Mol Biol Cell 6(4):387–400. Harvey M, McArthur MJ, Montgomery CA, Jr., Bradley A, Donehower LA. 1993. Genetic background alters the spectrum of tumors that develop in p53-deficient mice. Faseb J 7(10):938–943. Haupt Y, Maya R, Kazaz A, Oren M. 1997. Mdm2 promotes the rapid degradation of p53. Nature 387(6630):296–299. Hermeking H, Lengauer C, Polyak K, He TC, Zhang L, Thiagalingam S, Kinzler KW, Vogelstein B. 1997. 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Mol Cell 1(1):3–11. Hwang BJ, Ford JM, Hanawalt PC, Chu G. 1999. Expression of the p48 xeroderma pigmentosum gene is p53-dependent and is involved in global genomic repair. Proc Natl Acad Sci USA 96(2):424–428. Inga A, Storici F, Darden TA, Resnick MA. 2002. Differential transactivation by the p53 transcription factor is highly dependent on p53 level and promoter target sequence. Mol Cell Biol 22(24):8612–8625. Janus F, Albrechtsen N, Dornreiter I, Wiesmuller L, Grosse F, Deppert W. 1999. The dual role model for p53 in maintaining genomic integrity. Cell Mol Life Sci 55(1):12–27. Jin S, Tong T, Fan W, Fan F, Antinore MJ, Zhu X, Mazzacurati L, Li X, Petrik KL, Rajasekaran B, Wu M, Zhan Q. 2002. GADD45-induced cell cycle G2-M arrest associates with altered subcellular distribution of cyclin B1 and is independent of p38 kinase activity. Oncogene 21(57):8696–8704. Juven T, Barak Y, Zauberman A, George DL, Oren M. 1993. Wild type p53 can mediate sequence-specific transactivation of an internal promoter within the mdm2 gene. Oncogene 8(12):3411–3416. Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW. 1991. Participation of p53 protein in the cellular response to DNA damage. Cancer Res 51(23 Pt 1):6304–6311. Kearsey JM, Coates PJ, Prescott AR, Warbrick E, Hall PA. 1995. Gadd45 is a nuclear cell cycle regulated protein which interacts with p21Cip1. Oncogene 11(9):1675–1683. Ko LJ, Shieh SY, Chen X, Jayaraman L, Tamai K, Taya Y, Prives C, Pan ZQ. 1997. p53 is phosphorylated by CDK7-cyclin H in a p36MAT1-dependent manner. Mol Cell Biol 17(12):7220–7229. LaBaer J, Garrett MD, Stevenson LF, Slingerland JM, Sandhu C, Chou HS, Fattaey A, Harlow E. 1997. New functional activities for the p21 family of CDK inhibitors. Genes Dev 11(7):847–862. Lanni JS, Jacks T. 1998. Characterization of the p53-dependent postmitotic checkpoint following spindle disruption. Mol Cell Biol 18(2):1055–1064. Leng RP, Lin Y, Ma W, Wu H, Lemmers B, Chung S, Parant JM, Lozano G, Hakem R, Benchimol S. 2003. Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 112(6):779–791. Li R, Waga S, Hannon GJ, Beach D, Stillman B. 1994a. Differential effects by the p21 CDK inhibitor on PCNA-dependent DNA replication and repair. Nature 371(6497):534–537. Li Y, Jenkins CW, Nichols MA, Xiong Y. 1994b. Cell cycle expression and p53 regulation of the cyclin-dependent kinase inhibitor p21. Oncogene 9(8):2261– 2268. Lin D, Shields MT, Ullrich SJ, Appella E, Mercer WE. 1992. Growth arrest induced by wild-type p53 protein blocks cells prior to or near the restriction point in late G1 phase. Proc Natl Acad Sci USA 89(19):9210–9214. Lopez-Girona A, Kanoh J, Russell P. 2001. Nuclear exclusion of Cdc25 is not required for the DNA damage checkpoint in fission yeast. Curr Biol 11(1):50– 54. Lu H, Levine AJ. 1995. Human TAFII31 protein is a transcriptional coactivator of the p53 protein. Proc Natl Acad Sci USA 92(11):5154–5158. Mailand N, Falck J, Lukas C, Syljuasen RG, Welcker M, Bartek J, Lukas J. 2000. Rapid destruction of human Cdc25A in response to DNA damage. Science 288(5470):1425–1429. McLure KG, Lee PW. 1998. How p53 binds DNA as a tetramer. EMBO J 17(12): 3342–3350. Melle C, Nasheuer HP. 2002. Physical and functional interactions of the tumor suppressor protein p53 and DNA polymerase alpha-primase. Nucleic Acids Res 30(7):1493–1499.

Journal of Cellular Physiology DOI 10.1002/jcp

19

Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, Moll UM. 2003. p53 has a direct apoptogenic role at the mitochondria. Mol Cell 11(3): 577–590. Minn AJ, Boise LH, Thompson CB. 1996. Expression of Bcl-xL and loss of p53 can cooperate to overcome a cell cycle checkpoint induced by mitotic spindle damage. Genes Dev 10(20):2621–2631. Miyashita T, Reed JC. 1995. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80(2):293–299. Montagnoli A, Tenca P, Sola F, Carpani D, Brotherton D, Albanese C, Santocanale C. 2004. Cdc7 inhibition reveals a p53-dependent replication checkpoint that is defective in cancer cells. Cancer Res 64(19):7110–7116. Nakano K, Vousden KH. 2001. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7(3):683–694. Nakano K, Balint E, Ashcroft M, Vousden KH. 2000. A ribonucleotide reductase gene is a transcriptional target of p53 and p73. Oncogene 19(37):4283–4289. Nghiem P, Park PK, Kim Ys YS, Desai BN, Schreiber SL. 2002. ATR is not required for p53 activation but synergizes with p53 in the replication checkpoint. J Biol Chem 277(6):4428–4434. Nurse P. 1990. Universal control mechanism regulating onset of M-phase. Nature 344(6266):503–508. Nyberg KA, Michelson RJ, Putnam CW, Weinert TA. 2002. Toward maintaining the genome: DNA damage and replication checkpoints. Annu Rev Genet 36: 617–656. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T, Tokino T, Taniguchi T, Tanaka N. 2000a. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288(5468):1053– 1058. Oda K, Arakawa H, Tanaka T, Matsuda K, Tanikawa C, Mori T, Nishimori H, Tamai K, Tokino T, Nakamura Y, Taya Y. 2000b. p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell 102(6):849–862. Okamoto K, Beach D. 1994. Cyclin G is a transcriptional target of the p53 tumor suppressor protein. EMBO J 13(20):4816–4822. Okamoto K, Li H, Jensen MR, Zhang T, Taya Y, Thorgeirsson SS, Prives C. 2002. Cyclin G recruits PP2A to dephosphorylate Mdm2. Mol Cell 9(4):761–771. Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW, Vogelstein B. 1993. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature 362(6423):857–860. Owen-Schaub LB, Zhang W, Cusack JC, Angelo LS, Santee SM, Fujiwara T, Roth JA, Deisseroth AB, Zhang WW, Kruzel E, et al. 1995. Wild-type human p53 and a temperature-sensitive mutant induce Fas/APO-1 expression. Mol Cell Biol 15(6):3032–3040. Pan ZQ, Reardon JT, Li L, Flores-Rozas H, Legerski R, Sancar A, Hurwitz J. 1995. Inhibition of nucleotide excision repair by the cyclin-dependent kinase inhibitor p21. J Biol Chem 270(37):22008–22016. Pavletich NP, Chambers KA, Pabo CO. 1993. The DNA-binding domain of p53 contains the four conserved regions and the major mutation hot spots. Genes Dev 7(12B):2556–2564. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. 1997. A model for p53induced apoptosis. Nature 389(6648):300–305. Prives C, Manfredi JJ. 2005. The continuing saga of p53—More sleepless nights ahead. Mol Cell 19(6):719–721. Purdie CA, Harrison DJ, Peter A, Dobbie L, White S, Howie SE, Salter DM, Bird CC, Wyllie AH, Hooper ML, et al. 1994. Tumour incidence, spectrum and ploidy in mice with a large deletion in the p53 gene. Oncogene 9(2):603–609. Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A. 2000. Regulation of tumor angiogenesis by p53induced degradation of hypoxia-inducible factor 1alpha. Genes Dev 14(1):34– 44. Rieder CL, Maiato H. 2004. Stuck in division or passing through: What happens when cells cannot satisfy the spindle assembly checkpoint. Dev Cell 7(5):637– 651. Rohaly G, Chemnitz J, Dehde S, Nunez AM, Heukeshoven J, Deppert W, Dornreiter I. 2005. A novel human p53 isoform is an essential element of the ATR-intra-S phase checkpoint. Cell 122(1):21–32. Rousseau D, Cannella D, Boulaire J, Fitzgerald P, Fotedar A, Fotedar R. 1999. Growth inhibition by CDK-cyclin and PCNA binding domains of p21 occurs by distinct mechanisms and is regulated by ubiquitin-proteasome pathway. Oncogene 18(30):4313–4325. Sherr CJ. 1994. G1 phase progression: Cycling on cue. Cell 79(4):551–555. Smith ML, Chen IT, Zhan Q, Bae I, Chen CY, Gilmer TM, Kastan MB, O’Connor PM, Fornace AJ, Jr. 1994. Interaction of the p53-regulated protein Gadd45 with proliferating cell nuclear antigen. Science 266(5189):1376–1380. Smith ML, Kontny HU, Zhan Q, Sreenath A, O’Connor PM, Fornace AJ, Jr. 1996. Antisense GADD45 expression results in decreased DNA repair and sensitizes cells to u.v.-irradiation or cisplatin. Oncogene 13(10):2255–2263. Smith ML, Ford JM, Hollander MC, Bortnick RA, Amundson SA, Seo YR, Deng CX, Hanawalt PC, Fornace AJ, Jr. 2000. p53-mediated DNA repair responses to UV radiation: Studies of mouse cells lacking p53, p21, and/or gadd45 genes. Mol Cell Biol 20(10):3705–3714. Smits VA, Klompmaker R, Vallenius T, Rijksen G, Makela TP, Medema RH. 2000. p21 inhibits Thr161 phosphorylation of Cdc2 to enforce the G2 DNA damage checkpoint. J Biol Chem 275(39):30638–30643. St Clair S, Giono L, Varmeh-Ziaie S, Resnick-Silverman L, Liu WJ, Padi A, Dastidar J, DaCosta A, Mattia M, Manfredi JJ. 2004. DNA damage-induced downregulation of Cdc25C is mediated by p53 via two independent mechanisms: One involves direct binding to the cdc25C promoter. Mol Cell 16(5):725– 736. Stewart N, Hicks GG, Paraskevas F, Mowat M. 1995. Evidence for a second cell cycle block at G2/M by p53. Oncogene 10(1):109–115. Stivala LA, Riva F, Cazzalini O, Savio M, Prosperi E. 2001. p21(waf1/cip1)-null human fibroblasts are deficient in nucleotide excision repair downstream the recruitment of PCNA to DNA repair sites. Oncogene 20(5):563–570. Tanaka H, Arakawa H, Yamaguchi T, Shiraishi K, Fukuda S, Matsui K, Takei Y, Nakamura Y. 2000. A ribonucleotide reductase gene involved in a p53dependent cell-cycle checkpoint for DNA damage. Nature 404(6773):42–49. Taylor WR, Agarwal ML, Agarwal A, Stacey DW, Stark GR. 1999. p53 inhibits entry into mitosis when DNA synthesis is blocked. Oncogene 18(2):283–295.

20

GIONO AND MANFREDI

Thut CJ, Chen JL, Klemm R, Tjian R. 1995. p53 transcriptional activation mediated by coactivators TAFII40 and TAFII60. Science 267(5194):100– 104. Van Meir EG, Polverini PJ, Chazin VR, Su Huang HJ, de Tribolet N, Cavenee WK. 1994. Release of an inhibitor of angiogenesis upon induction of wild type p53 expression in glioblastoma cells. Nat Genet 8(2):171–176. Venot C, Maratrat M, Sierra V, Conseiller E, Debussche L. 1999. Definition of a p53 transactivation function-deficient mutant and characterization of two independent p53 transactivation subdomains. Oncogene 18(14):2405–2410. Vogel C, Kienitz A, Hofmann I, Muller R, Bastians H. 2004. Crosstalk of the mitotic spindle assembly checkpoint with p53 to prevent polyploidy. Oncogene 23(41):6845–6853. Vogelstein B, Lane D, Levine AJ. 2000. Surfing the p53 network. Nature 408(6810):307–310. Vousden KH. 2000. p53: Death star. Cell 103(5):691–694. Waga S, Hannon GJ, Beach D, Stillman B. 1994. The p21 inhibitor of cyclindependent kinases controls DNA replication by interaction with PCNA. Nature 369(6481):574–578. Waldman T, Kinzler KW, Vogelstein B. 1995. p21 is necessary for the p53mediated G1 arrest in human cancer cells. Cancer Res 55(22):5187–5190. Wang Y, Reed M, Wang P, Stenger JE, Mayr G, Anderson ME, Schwedes JF, Tegtmeyer P. 1993. p53 domains: Identification and characterization of two autonomous DNA-binding regions. Genes Dev 7(12B):2575–2586.

Journal of Cellular Physiology DOI 10.1002/jcp

Wang P, Reed M, Wang Y, Mayr G, Stenger JE, Anderson ME, Schwedes JF, Tegtmeyer P. 1994. p53 domains: Structure, oligomerization, and transformation. Mol Cell Biol 14(8):5182–5191. Wang XW, Zhan Q, Coursen JD, Khan MA, Kontny HU, Yu L, Hollander MC, O’Connor PM, Fornace AJ, Jr., Harris CC. 1999. GADD45 induction of a G2/M cell cycle checkpoint. Proc Natl Acad Sci USA 96(7):3706–3711. Wani MA, Wani G, Yao J, Zhu Q, Wani AA. 2002. Human cells deficient in p53 regulated p21(waf1/cip1) expression exhibit normal nucleotide excision repair of UV-induced DNA damage. Carcinogenesis 23(3):403–410. Wu GS, Burns TF, McDonald ER, 3rd, Jiang W, Meng R, Krantz ID, Kao G, Gan DD, Zhou JY, Muschel R, Hamilton SR, Spinner NB, Markowitz S, Wu G, elDeiry WS. 1997. KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet 17(2):141–143. Xiao H, Pearson A, Coulombe B, Truant R, Zhang S, Regier JL, Triezenberg SJ, Reinberg D, Flores O, Ingles CJ, et al. 1994. Binding of basal transcription factor TFIIH to the acidic activation domains of VP16 and p53. Mol Cell Biol 14(10):7013–7024. Zhan Q, Antinore MJ, Wang XW, Carrier F, Smith ML, Harris CC, Fornace AJ, Jr. 1999. Association with Cdc2 and inhibition of Cdc2/Cyclin B1 kinase activity by the p53-regulated protein Gadd45. Oncogene 18(18):2892–2900. Zhu Y, Alvarez C, Doll R, Kurata H, Schebye XM, Parry D, Lees E. 2004. Intra-Sphase checkpoint activation by direct CDK2 inhibition. Mol Cell Biol 24(14): 6268–6277.