Abstract
Free full text
The CRL4Cdt2 ubiquitin ligase targets the degradation of p21Cip1 to control replication licensing
Abstract
The faithful replication of genomic DNA is crucial for maintaining genome stability. In eukaryotes, DNA rereplication is prevented by the temporal regulation of replication licensing. Replication-licensing factors are required to form prereplicative complexes during G1 phase, but are inactivated in S phase to prevent rereplication. A vertebrate CUL4 CRL ubiquitin ligase (CRL4) complex containing Cdt2 as the substrate recognition subunit promotes proper DNA replication, in part, by degrading the replication-licensing factor Cdt1 during S phase. We show that the Caenorhabditis elegans CRL4Cdt2 complex has a conserved role in degrading Cdt1. Furthermore, we show that CRL4Cdt2 restrains replication licensing in both C. elegans and humans by targeting the degradation of the cyclin-dependent kinase (CDK) inhibitors CKI-1 and p21Cip1, respectively. Human CRL4Cdt2 targets the degradation of p21 in S phase, with the in vivo ubiquitylation of p21 by CRL4Cdt2 dependent on p21 binding to PCNA. Inactivation of Cdt2 induces rereplication, which requires the presence of the CDK inhibitor p21. Strikingly, coinactivation of CRL4Cdt2 and SCFSkp2 (which redundantly targets p21 degradation) prevents the nuclear export of the replication-licensing factor Cdc6 during S phase, and the block on nuclear export is dependent on p21. Our work defines the degradation of p21 as a critical aspect of replication licensing in human cells.
To maintain genome integrity, DNA replication must be tightly regulated so that replication only initiates once at each replication origin during S phase. The cellular pathway for preventing overreplication of genomic DNA (rereplication) is focused on blocking the reformation of prereplicative complexes (pre-RCs) in S phase (Arias and Walter 2007). The pre-RC consists of the six-member origin recognition complex (ORC), the replication-licensing factors Cdt1 and Cdc6, and the DNA helicase MCM2–7 complex. Pre-RC assembly occurs prior to S phase, either in late mitosis or G1 phase. The ORC binds to replication origins and recruits Cdt1 and Cdc6, which together recruit the putative replicative helicase, the MCM2–7 complex, to form the pre-RC. The formation of the pre-RC is said to provide the “license” for subsequent DNA replication. During S phase, specific pre-RC components are inactivated by various mechanisms (e.g., degradation, nuclear export, phosphorylation, or binding to inhibitors) so that pre-RCs cannot reform at replication origins that have already initiated DNA replication (Arias and Walter 2007).
In budding yeast, fission yeast, and Caenorhabditis elegans, multiple pre-RC components are regulated to redundantly prevent origin refiring during S phase (Arias and Walter 2007; Kim and Kipreos 2008). In vertebrates, three mechanisms have been implicated in preventing DNA rereplication: the degradation of the ORC component ORC1, the nuclear export of Cdc6, and the inactivation of Cdt1 either by degradation or by binding to the Cdt1 inhibitor geminin. The degradation of ORC1 may not be a consistent regulatory mechanism, as it is observed in some mammalian cells but not in others (Arias and Walter 2007). The importance of vertebrate Cdc6 nuclear export is not well established due to the observation that a substantial fraction of endogenous Cdc6 remains nuclear-localized throughout S phase (Kim and Kipreos 2008). In contrast, Cdt1 inactivation has been functionally linked to the prevention of DNA rereplication. In Xenopus and humans, limited rereplication is observed upon overexpression of a nondegradable Cdt1 mutant protein or inactivation of the Cdt1 inhibitor geminin (Vaziri et al. 2003; Melixetian et al. 2004; Zhu et al. 2004; Arias and Walter 2005; Li and Blow 2005; Yoshida et al. 2005). In C. elegans, Xenopus, humans, and fission yeast, Cdt1 is targeted for degradation during S phase by a cullin RING ubiquitin ligase (E3) complex (CRL) containing the cullin CUL4. In humans, Cdt1 is also redundantly degraded by the SCFSkp2 E3 complex (Kim and Kipreos 2007a).
In C. elegans, the inactivation of the cul-4 gene produces massive levels of DNA rereplication in proliferating cells, with DNA contents up to 100C (Zhong et al. 2003). C. elegans CUL-4 is required for both the degradation of CDT-1 and the nuclear export of CDC-6 during S phase (Zhong et al. 2003; Kim et al. 2007). CUL-4 promotes CDC-6 nuclear export by negatively regulating the level of the Cip/Kip family cyclin-dependent kinase (CDK) inhibitor CKI-1 (Kim et al. 2007). The deregulation of both CDT-1 and CDC-6 in C. elegans induces DNA rereplication (Kim et al. 2007). C. elegans CUL-4 is therefore a master regulator that negatively regulates both replication-licensing factors to prevent rereplication.
The structure of the CUL4 CRL complex (CRL4) is similar to that of other CRL complexes, and contains a RING H2 protein (Rbx1/Roc1), an adaptor protein (DDB1), and a substrate recognition subunit (SRS) (Higa and Zhang 2007; Lee and Zhou 2007; Bosu and Kipreos 2008). Rbx1 binds to the C terminus of cullins and promotes the binding of the ubiquitin-conjugating enzyme (E2) (Bosu and Kipreos 2008). DDB1 contains three β-propeller domains and binds to the N terminus of CUL4 (Angers et al. 2006). The SRS links to the complex through interaction with DDB1, and recruits substrates.
Recent proteomic approaches have identified a family of WD repeat proteins containing a conserved “WDXR” motif as potential SRSs for CRL4 complexes (Angers et al. 2006; He et al. 2006; Higa et al. 2006b; Jin et al. 2006). The WDXR protein family is predicted to contain ~90 members in mammals, of which at least 50 have been shown to be capable of physical association with CUL4 or DDB1 (Angers et al. 2006; He et al. 2006; Higa et al. 2006b; Jin et al. 2006). WDXR family members include the known CRL4 complex SRSs, suggesting that a majority of the WDXR family proteins will function as SRSs. In fission yeast and vertebrates, the WDXR protein Cdt2/DTL (Cdc10-dependent transcript 2/Denticleless) functions as a CRL4 SRS to target Cdt1 for degradation (Higa et al. 2006a; Jin et al. 2006; Ralph et al. 2006; Sansam et al. 2006). The degradation of Cdt1 is dependent on its interaction with PCNA (proliferating cell nuclear antigen), which is mediated by the PCNA-binding PIP-box motif of Cdt1 (Arias and Walter 2007).
In this work, we show that the C. elegans Cdt2 ortholog, CDT-2, functions in a CRL4CDT-2 complex to target the degradation of the replication-licensing factor CDT-1 and the CDK inhibitor CKI-1. We extend this to show that the human CRL4Cdt2 complex targets the degradation of the Cip/Kip family CDK inhibitor p21Cip1/Waf1/Sdi1. The CRL4Cdt2-mediated ubiquitylation of p21 requires p21 binding to PCNA. We present evidence that CRL4Cdt2 targets the degradation of p21 during S phase to control the nuclear export of the Cdc6 replication-licensing factor and prevent DNA rereplication.
Results
CRL4CDT-2 is required for CKI-1 degradation in C. elegans
In C. elegans, cul-4 inactivation causes substantial rereplication in mitotically proliferating larval cells (Zhong et al. 2003). The rereplication in cul-4 mutants arises from a failure to negatively regulate the levels of the replication-licensing factor CDT-1 and the CDK inhibitor CKI-1 (Zhong et al. 2003; Kim and Kipreos 2007b; Kim et al. 2007). CKI-1 accumulates in rereplicating cul-4 mutant cells, and cki-1 RNAi depletion suppresses the rereplication, indicating that CKI-1 is essential for the rereplication to occur (Kim and Kipreos 2007b; Kim et al. 2007). In order to determine which SRS is responsible for the degradation of CKI-1 in C. elegans, we took a candidate gene approach. Fifty WDXR motif proteins have been identified as physical interactors with CRL4 components in mammals, and are therefore potential SRSs (Angers et al. 2006; He et al. 2006; Higa et al. 2006b; Jin et al. 2006). We identified 24 orthologs of these genes in C. elegans, and tested RNAi depletions of eight of the genes to determine if the RNAi would induce CKI-1 accumulation, as determined by immunofluorescence with anti-CKI-1 antibody. Of the tested WDXR motif genes, only the RNAi inactivation of the cdt-2 gene (the ortholog of mammalian CDT2) produced an accumulation of CKI-1 in larval blast cells (Fig. 1A; see the Materials and Methods section for the genes tested).
cdt-2(RNAi) larvae exhibit enlarged blast cells, including seam cells, that have increased DNA content, similar to what is observed in cul-4 or ddb-1 RNAi animals (Fig. 1A,B). The level of genomic DNA in the enlarged cdt-2(RNAi) cells is 14.8 ± 7.1 C DNA content for L4stage larvae (2 d post-hatch), relative to 2.0 ± 0.2 C for wild-type L4-stage larvae, and 25.9 ± 15.4 C for ddb-1(RNAi) arrested L2-stage larvae (3 d post-hatch). However, cdt-2(RNAi) larvae do not exhibit all of the cul-4 mutant phenotypes. Unlike cul-4 or ddb-1 mutants, cdt-2(RNAi) animals do not arrest at the L2 larval stage or exhibit a stereotypical degenerating germ cell phenotype. The observation of only a subset of cul-4 mutant phenotypes is consistent with the loss of a single class of CRL4 complex (as each class is defined by its associated SRS).
In vertebrates, Cdt2 functions as the CRL4 SRS to target the degradation of Cdt1 in S phase (Higa et al. 2006a; Jin et al. 2006; Sansam et al. 2006). We found that C. elegans CDT-2 is similarly required for CDT-1 degradation in S phase, with CDT-1GFP perduring in S-phase cdt-2(RNAi) seam cells (Fig. 1C). In cul-4 mutants, the rereplication phenotype arises from the failure to degrade CDT-1 and CKI-1 (Zhong et al. 2003; Kim and Kipreos 2007b). The observation that cdt-2(RNAi) blast cells have increased DNA content and accumulate both CDT-1 and CKI-1, strongly suggests that CDT-2 functions in the context of a CRL4 complex to prevent DNA rereplication.
CDT-2 functions as a CRL4 complex SRS to target CKI-1 degradation
To test if CDT-2 functions as an SRS, we analyzed the physical interactions of CDT-2 with CRL4 components and the putative substrates CDT-1 and CKI-1 using the yeast two-hybrid assay. The two-hybrid results are consistent with CDT-2 functioning as an SRS: CDT-2 interacted with the adaptor DDB-1 and with the substrates CKI-1 and CDT-1, but did not interact directly with CUL-4 (Fig. 2A [top panel], B). Also consistent with the CRL structure, the adaptor DDB-1 did not interact directly with the substrates CKI-1 or CDT-1 in the two-hybrid assay (Fig. 2A, top; data not shown). We tested whether CDT-2 can act as a bridge between DDB-1 and CKI-1 using the yeast three-hybrid test (Licitra and Liu 1996). CDT-2 was expressed from the GAL1 promoter, which is induced by galactose and repressed by glucose (Giniger et al. 1985). Yeast containing the GAL1 promoter/CDT-2 construct plus a DDB-1/GAL4 DNA-binding domain fusion and CKI-1/GAL4 activation domain fusion were able to grow on histidine(−) selection plates containing galactose but not glucose, indicating that CDT-2 can bridge interaction between DDB-1 and CKI-1 (Fig. 2A, bottom). A similar experiment showed that DDB-1 can mediate interaction between CUL-4 and CDT-2 (Fig. 2A, bottom).
To test if CKI-1 is a ubiquitylation substrate of CDT-2, we utilized a mammalian expression system. We observed that C. elegans CDT-2 can interact with TAP-tagged human DDB1, suggesting that it can interact with human CRL4 components (Fig. 2C). Coexpression of C. elegans CDT-2 with CKI-1 in HeLa cells stimulated the in vivo ubiquitylation of CKI-1, suggesting that CKI-1 is a direct substrate of the CRL4CDT-2 E3 complex (Fig. 2D). Overall, our data indicate that CDT-2 functions as a CRL4 SRS to catalyze the ubiquitin-mediated degradation of the replication-licensing factor CDT-1 and the Cip/Kip family CDK inhibitor CKI-1 in C. elegans.
Human CRL4Cdt2 targets p21Cip1 for degradation
The mammalian Cip/Kip family includes three members (p21Cip1, p27Kip1, and p57Kip2) that are each capable of inhibiting a wide-range of CDK holoenzymes (complexes of CDKs and cyclins) (Besson et al. 2008). To test if Cdt2 negatively regulates the levels of Cip/Kip family CDK inhibitors in human cells, we determined whether Cdt2 siRNA knockdown would lead to increases in the protein levels of the three mammalian CDK inhibitors. We observed that Cdt2 knockdown in HeLa cells produced a 5.6-fold increase in p21Cip1 protein levels (Fig. 3A). In contrast, Cdt2 knockdown did not increase the levels of the Cip/Kip family members p27Kip1 or p57Kip2 (Fig. 3A). It has been reported that the E3 SCFSkp2 can target p21, p27, and p57 for degradation (Yu et al. 1998; Carrano et al. 1999; Sutterluty et al. 1999; Tsvetkov et al. 1999; Bornstein et al. 2003; Kamura et al. 2003). In our experiments, Skp2 knockdown in HeLa cells increased the levels of p21 and p27, but did not affect p57 levels. Notably, coknockdown of Cdt2 and Skp2 increased p21 levels approximately twofold higher than either single knockdown (an 11-fold increase), indicating that Cdt2 and Skp2 redundantly regulate p21 (Fig. 3A). Unless otherwise noted, siRNA experiments were performed with pools of four siRNAs. Knockdowns with individual siRNAs for Cdt2 and Skp2 also produced increased p21 levels (Fig. 3B).
To determine if Cdt2 regulates p21 levels in S phase, we inactivated Cdt2 in asynchronous cells and in cells enriched in S phase by hydroxyurea (HU) treatment. The level of p21 was significantly increased in S-phase Cdt2 siRNA cells relative to asynchronous Cdt2 siRNA cells, suggesting that CRL4Cdt2 predominantly regulates p21 levels during S phase (Fig. 3C). A similar increase in p21 levels was also observed in aphidicolin-blocked Cdt2 siRNA cells (data not shown). In contrast to the Cdt2 knockdown, Skp2 siRNA treatment produced similar elevated levels of p21 in asynchronous and HU-treated cells, suggesting that SCFSkp2-mediated p21 degradation is not restricted to S phase (Fig. 3C). To determine if Cdt2 affects the rate of p21 protein turnover, we performed a time-course experiment in which Cdt2 knockdown or control GFP knockdown cells were arrested in S phase with HU in the presence of the proteasome inhibitor MG132. At time 0, HU and MG132 were removed while cycloheximide was added to block protein synthesis. We found that the half-life of endogenous p21 was significantly increased in Cdt2 knockdown cells, indicating that Cdt2 contributes to p21 turnover during S phase (Fig. 3D).
It has been observed that p21 levels decrease during S phase as serum-stimulated quiescent mammalian cells progress through the cell cycle (Dulic et al. 1998; Amador et al. 2007). We asked whether Cdt2 is required for this drop in p21 protein levels. To test this, serum-starved HeLa cells were treated with either Cdt2 siRNA or control GFP siRNA, and then stimulated with serum. As previously reported, p21 levels increased in control cells after serum stimulation and then decreased as cells progressed through S phase (Fig. 3E). Cdt2 protein levels were very low in quiescent control cells, but increased significantly upon serum stimulation, with the highest level observed at 9 h post-serum stimulation coincident with increased expression of Cyclin B1, indicating that the cells had entered S phase (Fig. 3E). The increase in Cdt2 levels correlated with the decrease in p21 levels. In Cdt2 knockdown cells, the induction of Cdt2 protein after serum stimulation was blocked, and p21 was markedly stabilized as cells progressed through S phase (Fig. 3E). Our results indicate that Cdt2 contributes significantly to the degradation of p21 during S phase in mitotically dividing cells.
To determine if Cdt2 directly targets the degradation of p21, we tested for physical interaction between p21 and Cdt2. We observed that when expressed in HEK293T cells, Flag-Cdt2 and Myc-p21 reciprocally coimmunoprecipitate with each other, while Flag-Cdt2 did not show significant interaction with Myc-p27Kip1 (Fig. 4A). We also asked whether the expression of Cdt2 would allow interaction between p21 and DDB1, as expected for an SRS. We observed that significantly more p21 associated with expressed TAP-tagged DDB1 in the presence of Flag-Cdt2 than in its absence (a 6.7-fold difference) (Fig. 4B). The observed low-level of p21 bound to TAP-DDB1 in the absence of Flag-Cdt2 was potentially mediated by interaction with the endogenous Cdt2.
We next sought to determine if Cdt2 facilitated p21 ubiquitylation. We observed that expression of Flag-Cdt2 stimulated the in vivo conjugation of HA-ubiquitin to endogenous p21 (Fig. 5A). Expression of Flag-Skp2 also stimulated the in vivo ubiquitylation of p21, consistent with the siRNA data that showed that both E3 complexes regulate p21 levels (Fig. 5A). The CRL4Cdt2 complex can catalyze the in vitro ubiquitylation of p21 (Fig. 5B). Taken together, our results suggest that p21Cip1 is a direct target for ubiquitylation by the CRL4Cdt2 E3 ligase.
p21 degradation is PIP-box-dependent
The ubiquitylation of vertebrate Cdt1 by CRL4Cdt2 is dependent on the interaction of Cdt1 with PCNA, mediated by the PIP-box sequence of Cdt1 (Higa et al. 2006a; Hu and Xiong 2006; Jin et al. 2006; Nishitani et al. 2006; Senga et al. 2006). p21 has a C-terminal PIP-box motif and can bind to PCNA (Waga et al. 1994). We observed that the precipitated Flag-Cdt2 immunocomplex, which was capable of in vitro ubiquitylation of Myc-p21, contains PCNA (Fig. 5C; data not shown). To determine the importance of the PIP-box for the physical interaction between p21 and Cdt2, we converted the PIP-box sequence of p21 (QTSMTDFY) to eight alanine residues. The p21 PIP-box mutant protein coprecipitated with Flag-Cdt2 at levels equivalent to that of wild-type p21 (Fig. 5D). As expected, PCNA was only coprecipitated above background levels with wild-type p21 but not the p21 PIP-box mutant, suggesting that Cdt2 can bind p21 independently of PCNA association (Fig. 5D).
To determine if the PIP-box of p21 is required for its in vivo ubiquitylation, we expressed wild-type p21 or PIP-box mutant p21 along with HA-ubiquitin and Flag-Cdt2. We observed that the in vivo ubiquitylation of wild-type p21 was significantly greater than the ubiquitylation of the p21 PIP-box mutant (Fig. 5E). This suggests that p21 ubiquitylation depends on PCNA binding, providing a mechanistic link between the CRL4Cdt2-mediated degradation of the two substrates Cdt1 and p21, with both ubiquitylation reactions dependent on interaction with PCNA.
p21 is required for the rereplication induced upon inactivation of CRL4Cdt2
We observed that Cdt2 siRNA treatment of HeLa cells induces rereplication, producing increased numbers of cells with DNA content greater than 4C (Fig. 6), similar to what has been reported previously (Jin et al. 2006). Notably, Skp2 siRNA did not produce cells with DNA content greater than 4C, and coinactivation of Cdt2 and Skp2 did not increase the percentage of cells with greater than 4C DNA content beyond that of Cdt2 siRNA alone (Fig. 6). Thus, Cdt2 is required to prevent DNA rereplication in HeLa cells, while Skp2 does not significantly contribute to the prevention of overreplication. Strikingly, the rereplication observed with both Cdt2 knockdown and the coknockdown of Cdt2 and Skp2 was abolished upon coinactivation of p21 (Fig. 6). Thus, p21 is essential for the rereplication that occurs when Cdt2 is inactivated.
CRL4Cdt2 and SCFSkp2 redundantly promote Cdc6 nuclear export in a p21-dependent manner
In C. elegans, CUL4 promotes the nuclear export of the replication-licensing factor CDC-6 by negatively regulating the levels of CKI-1, which allows CDKs to phosphorylate CDC-6 and induce its nuclear export (Kim et al. 2007). Based on our observation that CRL4Cdt2 targets the degradation of p21, we wanted to determine if the regulation of p21 levels would similarly control Cdc6 nuclear export in humans. We followed the subcellular localization of endogenous Cdc6 in HeLa cells using immunofluorescence with anti-Cdc6 antibody. Cells were costained with anti-Cyclin A antibody to mark cells in S/G2 phase. In the control siRNA population, Cdc6 is predominantly nuclear localized in Cyclin A-negative cells (G1 phase), while Cdc6 is primarily cytoplasmic in Cyclin A-positive cells (S/G2 phase), indicating that the majority of Cdc6 is exported to the cytoplasm during S phase (Fig. 7A,B; data not shown). The majority of Cdt2 siRNA cells have cytoplasmic-localized Cdc6 during S/G2 phase, although an increased percentage of cells exhibit equal distributions of nuclear and cytoplasmic Cdc6. The percentage of the total cellular Cdc6 signal that is nuclear-localized upon Cdt2 knockdown is modestly higher than in control GFP knockdown cells (31 ± 1.8% vs. 25 ± 1.7%; P = 0.05, Student’s t-test). Skp2 knockdown led to a greater reduction in Cdc6 nuclear export, with over half the cells showing equivalent levels of nuclear and cytoplasmic Cdc6 (Fig. 7A,B). Strikingly, when Cdt2 and Skp2 were inactivated together, a majority of Cyclin A-positive cells exhibited nuclear-localized Cdc6, indicating an almost total failure of Cdc6 nuclear export (Fig. 7A,B). Therefore, both Cdt2 and Skp2 are redundantly required to allow Cdc6 nuclear export during S phase.
We hypothesized that the failure to degrade p21 upon inactivating both Cdt2 and Skp2 contributed to the block on Cdc6 nuclear export during S phase. Knockdown of p21 by itself has no effect on the subcellular distribution of Cdc6 relative to control cells (data not shown). To test if p21 was required for the failure of Cdc6 nuclear export in Cdt2 and Skp2 double-knockdown cells, we coinactivated p21, Cdt2, and Skp2. Western blotting demonstrated the high efficiency of the triple siRNA knockdown (Fig. 7C). As we hypothesized, coinactivation of p21 with Cdt2 and Skp2 markedly increased the percentage of Cdc6 that was cytoplasmic-localized in S/G2-phase cells, indicating that p21 is required for the block on Cdc6 nuclear export (Fig. 7A,B). We note that the p21, Cdt2, Skp2 triple-knockdown cells retain higher nuclear Cdc6 levels than Cdt2 knockdown cells even though the triple-knockdown cells do not undergo rereplication while a substantial percentage of Cdt2 knockdown cells rereplicate. This suggests that increased Cdc6 nuclear localization is not sufficient to explain how p21 contributes to rereplication, and implies that the presence of p21 in S-phase cells deregulates additional replication-licensing pathway(s).
Discussion
In this study, we identified the C. elegans WDXR motif protein CDT-2 as an SRS for a CRL4 E3 complex that targets the CDK inhibitor CKI-1 and the replication-licensing factor CDT-1 for degradation. CDT-2 can bind to both CKI-1 and CDT-1 and negatively regulate their protein levels in C. elegans. CDT-2 also promotes the ubiquitylation of CKI-1 when expressed in mammalian cells. We extended our findings by demonstrating that the human CRL4Cdt2 complex has a conserved function in targeting the degradation of the CDK inhibitor p21Cip1. Inactivation of Cdt2 in HeLa cells causes a significant increase in p21 levels, but does not affect the levels of the related CDK inhibitors p27Kip1 and p57Kip2. Cdt2 overexpression stimulates the in vivo ubiquitylation of p21. The CRL4Cdt2 complex binds p21 in vivo and can ubiquitylate p21 in vitro. Therefore, we propose that p21Cip1 is a direct substrate of the CRL4Cdt2 E3 complex.
The CDK inhibitor p21 has a central role as an effector of cell cycle arrest when it is transactivated by the p53 tumor suppressor protein in response to DNA damage (Besson et al. 2008). While p21 is primarily considered an inhibitor of CDK/cyclin complexes, it also has CDK-independent functions involving the regulation of the cytoskeleton, transcription, and apoptosis (Besson et al. 2008). During the cell cycle, p21 levels are high in G1 phase, decrease significantly during S phase, and increase again during G2 phase (Dulic et al. 1998; Amador et al. 2007). p21 has been shown to undergo proteasome-mediated degradation through the actions of a number of E3s: SCFSkp2, MDM2, MDMX, and APC/CCdc20 (Yu et al. 1998; Carrano et al. 1999; Jin et al. 2003, 2008; Amador et al. 2007). APC/CCdc20 targets the degradation of p21 during prometaphase (Amador et al. 2007). MDM2 and MDMX promote the proteasomal turnover of p21 predominantly in G1 and early S phases (Jin et al. 2008). SCFSkp2 has been shown to target p21 degradation in S-phase cells (Bornstein et al. 2003); however, our study suggests that this degradation is not restricted to S phase. In contrast, we observed that CRL4Cdt2-mediated degradation of p21 occurs primarily in S phase.
We showed that the in vivo ubiquitylation of p21 by CRL4Cdt2 is dependent on p21 binding to PCNA. The binding of Cdt1 to chromatin-associated PCNA has been proposed as a mechanism to ensure that Cdt1 degradation is S-phase-specific (Arias and Walter 2006; Senga et al. 2006), and a similar mechanism may explain the S-phase specificity of the CRL4Cdt2-mediated p21 degradation. The significance of the PCNA–p21 interaction is underlined by the independent discovery that CRL4Cdt2 targets p21 degradation in response to DNA damage in a PCNA-dependent manner (T. Abbas and A. Dutta, pers. comm.).
The observation that the CRL4Cdt2-mediated degradation of both Cdt1 and p21 substrates is PCNA-dependent suggests that Cdt2 may have evolved to specifically target substrates in the context of PCNA binding. It is possible that CRL4Cdt2-mediated ubiquitylation in response to PCNA binding will be a general mechanism for degrading S-phase substrates, analogous to the degradation of mitotic substrates by APC/C. The most obvious candidates for this regulation are the numerous identified PCNA-binding proteins (Moldovan et al. 2007). p21 has previously been shown to bind PCNA, and this interaction was suggested as a mechanism to inactivate PCNA after DNA damage (Waga et al. 1994). The PCNA–p21 interaction has recently been shown to promote the targeting of DNA repair proteins to sites of DNA damage (Cazzalini et al. 2008; Masih et al. 2008). However, the role of PCNA–p21 interaction in normal cell cycle progression had not been determined (Moldovan et al. 2007). Our results suggest that the interaction of PCNA and p21 during S phase promotes p21 degradation to allow the proper temporal regulation of DNA replication licensing.
The inactivation of Cdt2 induces rereplication in HeLa cells (Jin et al. 2006). Strikingly, we found that the rereplication induced by Cdt2 knockdown is abolished by the coinactivation of p21. Although both CRL4Cdt2 and SCFSkp2 target the degradation of Cdt1 and p21, there are clear differences in the relevance of the two E3 complexes for the prevention of DNA rereplication. Inactivation of Cdt2 induces substantial rereplication, while inactivation of Skp2 does not produce rereplication. It is possible either that CRL4Cdt2 and SCFSkp2 target different subpopulations of Cdt1 and p21, or that there are additional targets of CRL4Cdt2 whose degradation restrains DNA replication licensing.
In C. elegans, the failure to export Cdc6 during S phase in cul-4 mutants is directly linked to the rereplication phenotype (Kim et al. 2007). In HeLa cells, inactivating either Cdt2 or Skp2 alone produces only partial effects on Cdc6 nuclear export during S phase. However, coinactivation of both Cdt2 and Skp2 produces an almost complete block on Cdc6 nuclear export, indicating that the nuclear export is redundantly controlled by the two E3 ligases. The coinactivation of p21 with Cdt2 and Skp2 partially suppresses the Cdc6 nuclear export defect, suggesting that CRL4Cdt2 and SCFSkp2 regulate Cdc6 nuclear export, at least in part, via degradation of p21.
In vertebrates, it has not been determined whether the nuclear export of Cdc6 inhibits its activity during S phase because a substantial fraction of endogenous Cdc6 remains in the nucleus throughout S phase (Kim and Kipreos 2008). Our results do not clarify this issue, as we observed rereplication in Cdt2 knockdown cells in which there was only a modest increase in Cdc6 nuclear retention; and so it is not clear if the limited increase in nuclear Cdc6 allowed the rereplication or whether the increase was immaterial. However, we can conclude that Cdc6 nuclear retention is not sufficient to explain how p21 promotes rereplication. This is based on the observation that while coinactivation of p21 with Cdt2 and Skp2 blocks rereplication, these triple-knockdown cells have higher nuclear Cdc6 levels than Cdt2 single-knockdown cells, which undergo rereplication. Therefore, an elevated level of nuclear Cdc6 is not sufficient to induce rereplication in the absence of p21 in Cdt2 and Skp2 double-knockdown cells. This implies that p21 promotes rereplication through additional pathways beyond its effect on Cdc6 localization.
The increased level of p21 in Cdt2 knockdown cells may promote rereplication either by inhibiting CDK/cyclin activity or through CDK-independent mechanism(s). CDK/cyclin activity has been shown to inhibit the reinitiation of DNA replication during S phase in a number of eukaryotes (Arias and Walter 2007; Kim and Kipreos 2008). In budding yeast, CDK/cyclins block pre-RC formation through at least three pathways: induction of Cdc6 degradation, inactivation of ORC components, and the nuclear export of the Mcm2–7 helicase (Nguyen et al. 2001). In mammalian cells, siRNA knockdown of both CDK1 and CDK2 induces rereplication, suggesting that CDK activity is an essential aspect of restricting replication licensing (Machida and Dutta 2007). However, the relevant mammalian CDK/cyclin targets have not been identified, but may include Cdc6, whose subcellular localization is regulated by CDK-mediated phosphorylation (Kim and Kipreos 2008). Our results imply that Cdc6 cannot be the sole relevant target of CDK/cyclins, and that other functional CDK/cyclin targets exist, whose phosphorylation prevents rereplication. The additional mechanism(s) by which elevated p21 fosters rereplication are not known, but could involve promoting Cdt1 activity or more novel pathways to initiate pre-RC reformation.
From an evolutionary perspective, it is striking that the CRL4Cdt2 complex has been conserved as a master regulator of DNA replication licensing. In both nematodes and humans, CRL4Cdt2 controls the licensing factor Cdt1 (via degradation) and the licensing factor Cdc6 (via localization). Further, in both species, CRL4Cdt2 catalyzes the degradation of a CDK inhibitor in order to prevent the reinitiation of DNA replication. We propose that actively reducing CDK inhibitor activity during S phase is a central mechanism among metazoa for the prevention of DNA rereplication. Our results suggest that in humans, the degradation of p21 in S-phase cells performs this role.
Materials and methods
C. elegans analysis
The strains used were as follows: N2, wild type; ET341, unc-119(ed4); ekEx23[pPD95.75/Pwrt-2CDT-1GFP + pPD95.67/Prnr-1tdTomato + pRF4]; syIs78[AJM-1GFP + unc-119(+)]. RNAi was performed using the feeding RNAi protocol as described (Kamath et al. 2001). L4-stage larvae were placed on RNAi and their progeny were analyzed. cdt-2 RNAi was performed with E. coli strain HT115 containing plasmid pPD129.36/cdt-2, which contains a full-length cdt-2 cDNA situated between double T7 promoters. RNAi depletion of the following WDXR genes were analyzed by anti-CKI-1 immunofluorescence: D2030.9, F47D12.9, F53C11.7, R11D1.1, T01C3.1 (cdt-2), Y73E7A.9, ZK430.7, and ZK1251.9. Anti-CKI-1 immunofluorescence and quantitation of DNA content were performed as described (Kim and Kipreos 2007b). The isolation of larvae at set times post-hatch for the observation of CDT-1GFP in seam cells in G1 or S phase was performed as described (Zhong et al. 2003).
Yeast two- and three-hybrid assays
For two-hybrid analysis, the full-length cul-4, ddb-1, cdt-2, cki-1, and cdt-1 cDNAs were cloned into the vectors pACT2 (which encodes GAL4 activation domain fusion) and pAS2 (encodes GAL4 DNA-binding domain fusion) (Clontech). Plasmids were transformed into the Saccharomyces cerevisiae strain pJ69-4A (James et al. 1996) using standard procedures (Ausubel et al. 1995). Interaction in the two-hybrid system was assayed on histidine-deficient selective media containing 1 mM 3-amino-1,2,4-triazole (3-AT, Sigma). For three-hybrid tests, cdt-2 and ddb-1 cDNAs were cloned into the pYES2 vector (Invitrogen), in which the gene of interest is under the control of the GAL1 promoter. Two-hybrid bait and prey plasmids as well as the pYES2 construct were cotransformed into pJ69-4A. Interaction in the three-hybrid system was determined on histidine-deficient selective media containing 1 mM 3-AT in the presence of 2% glucose or galactose.
Expression constructs
To express Flag-tagged proteins in mammalian systems, cDNAs of human Cdt2 and C. elegans cki-1 were cloned into pCMV-Tag2 (Stratagene). For Myc-tagged proteins, cDNAs of human p21 and p27 were cloned into pCS3-MT (Hallier et al. 1998), and the C. elegans cdt-2 cDNA was cloned into pcDNA3.1/Myc-His (Invitrogen). The expression constructs pcDNA3/FlagSkp2 (Bashir et al. 2004), pCMV/HA-UB (Furukawa et al. 2005), p3XFlag-CMV10/Hs Cdt2 (Higa et al. 2006b), pCS2/p21 (Sheaff et al. 2000), and pGLUE-hDDB1 (Angers et al. 2006) were gifts from M. Pagano, Y. Xiong, H. Zhang, B. Clurman, and S. Angers, respectively. The PIP-box in p21 was mutated by overlapping PCR mutagenesis (Mikaelian and Sergeant 1992) to convert the PIP-box sequence (QTSMTDFY) to eight alanine residues.
Mammalian cell culture
HeLa and HEK293T cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (GIBCO-BRL). For expression in HeLa and HEK293T cells, expression constructs were transfected using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. Flag-Cdt2 and wild-type p21 were expressed in an HEK293T cell line stably expressing TAP-DDB1. To isolate HEK293T cell lines stably expressing TAP-DDB1, cells were transfected with pGLUE-hDDB1, and selected in a medium containing 0.7 μg/mL puromycine (Sigma). For the study of p21 protein turnover, HeLa cells were treated with GFP or Cdt2 siRNA and then incubated with 2.5 mM HU (Sigma) for 19 h, and with the proteasome inhibitor MG132 (Sigma) for the last 7 h of the HU incubation. At time 0, HU and MG132 were removed from the culture media, and 25 μg/mL cycloheximide (Sigma) was added. For the analysis of p21 levels upon cell cycle entry, HeLa cells were serum-starved for 54 h by incubation in DMEM containing 0.02% fetal calf serum (FCS) in order to induce G0 entry. Cells were transfected with GFP or Cdt2 siRNA for the last 18 h of the serum starvation. At time 0, cells were stimulated with 15% FCS plus a second siRNA transfection. At set time points, cells were harvested for Western blot analysis.
Antibodies, immunofluorescence, and immunoprecipitation
For Western blots, whole-cell lysates were prepared by treating with NP-40 buffer: 1% or 0.5% NP-40, 0.15 M NaCl, 10 mM Sodium phosphate (pH 7.5), 10% glycerol, Protease Inhibitor Cocktail (Roche), 2 mM DTT, 1 mM EDTA, and 15 μM MG132. Monoclonal antibodies used were anti-p21 (Upstate Biotechnologies), anti-Cdc6 (DCS-180, Upstate Biotechnologies), anti-Myc (9E10, Covance), anti-Flag (M2, Sigma), anti-Cyclin A (Ab-6, Thermo Scientific), anti-Cyclin B1 (GNS1, Santa Cruz Biotechnologies), and anti-α-tubulin (DM1A, Sigma). Rabbit polyclonal antibodies used were anti-Cdt2 (Bethyl Laboratories), anti-Skp2 (H-435, Santa Cruz Biotechnologies), anti-p27 (Upstate Biotechnologies), anti-p57 (Upstate Biotechnologies), anti-Cyclin A (H-432, Santa Cruz Biotechnologies), anti-CUL4 (H-66, Santa Cruz Biotechnologies), anti-PCNA (PC10, Santa Cruz Biotechnologies), and anti-HA (ab9110, Abcam). For immunofluorescence, HeLa cells were fixed in 4.0% paraformaldehyde in PBS for 10 min at room temperature, washed twice with 0.4% Triton X-100 in PBS, and stained with monoclonal anti-Cdc6 (1:100) and rabbit polyclonal anti-Cyclin A (1:250). Anti-mouse Alexa-Fluor 488 (Molecular Probes) and anti-rabbit Alexa-Fluor 546 (Molecular Probes) were used as secondary antibodies. Alternatively, cells were stained with monoclonal anti-Cdc6 (1:100) and monoclonal anti-Cyclin A (1:40) using the Zenon Mouse Tricolor IgG1 labeling kit (Invitrogen). DNA was stained with 1 μg/mL Hoechst 33258 dye (Sigma). Immunofluorescence images were taken with the same exposure times and the images were processed identically using Adobe Photoshop software. Quantitation of nuclear and total immunofluorescence signal was performed with OpenLab software (version 5.0; Improvision). For the quantitation of Cdc6 localization in Figure 7B, 21–23 cells were analyzed for each condition.
Immunoprecipitation using anti-Myc and anti-Flag antibodies was performed as described (Kim and Kipreos 2007b). The purification of TAP-tagged DDB1 with associated proteins was modified from Angers et al. (2006). Briefly, HEK293T cells stably expressing TAP-DDB1 were cultured in a six-well plate and transfected using Lipofectamine 2000. After an 8-h incubation at 37°C, cells were treated with 2.5 mM HU and 10 μM MG132, and then harvested after an additional 10 h of incubation at 37°C. Whole-cell lysates were prepared with NP-40 buffer. Cell extracts were incubated at 4°C with 20 μL of packed streptavidin resin (Amersham); the resin was washed, and associated proteins were analyzed by Western blot using the ECL Advance chemiluminescence kit (Amersham). The immunoprecipitation of Flag-Cdt2 shown in Figure 5D was performed from a chromatin fraction of HeLa cells, prepared essentially as in Sarcinella et al. (2007), except that micrococcal nuclease (Sigma) treatment of nuclei was for 2 min at 37°C, and the nuclear extract was further prepared by osmotic lysis of nuclei. Determining the intensity of Western blot protein bands was performed with unsaturated images, and involved subtracting the mean background signal and standardizing to the level of α-tubulin in the same sample.
siRNA and cell cycle analysis
The following ON-TARGETplus siRNAs were generated by Dharmacon (overhanging bases are in lowercase): siGFP, GCUGACCCUGAAGUUCAUCUGdTdT; siCdt2(#2), GAAU UAUACUGCUUAUCGAuu; siCdt2(#3), GCGCUUGAAUAGA GGCUUAuu; siCdt2(#4), GUCAAGACCUGGCCUAGUAuu; siCdt2(#5), ACUCCUACGUUCUCUAUUAuu; siSkp2(#1), GC AUGUACAGGUGGCUGUUuu; siSkp2(#2), UGUCAAUACU CUCGCAAAAuu; siSkp2(#3), UCGGUGCUAUGAUAUAA UAuu; siSkp2(#4), GGUAUCGCCUAGCGUCUGAuu; siSkp2(#5), GGAUGUGACUGGUCGGUUGuu; sip21(#1), CUUCGACUU UGUCACCGAGuu; sip21(#2), AGACCAGCAUGACAGAUU Uuu; sip21(#3), CGACUGUGAUGCGCUAAUGuu; sip21(#4), CCUAAUCCGCCCACAGGAAuu; and sip21(#5), CGUCAG AACCCAUGCGGCAuu. The Cdt2(#1) siRNA, CAAUGGACA CCAGAACUCUACCUUUdTdT, was generated by Invitrogen. ON-TARGETplus SMARTpools of siRNAs (comprised of individual siRNAs #2–#5) were used for Cdt2, Skp2, and p21 knockdowns in all experiments except Figure 3E, which used individual Cdt2(#1) siRNA. Both pooled and individual siRNAs were used for the experiments in Figures. 3B, ,6,6, and and7A,7A, for which the individual siRNAs gave results that were similar to the pooled siRNAs. siRNA SMARTpools were transfected once at 100 nM (individual siRNAs in each pool were at 25 nM), and individual (nonpool) siRNAs were transfected two or three times sequentially at 100 nM. siRNAs were transfected with Oligofectamine (Invitrogen) according to the manufacturer’s instructions. HeLa cells were analyzed 60 h after transfection. For flow cytometry to determine DNA content, HeLa cells were fixed with 70% ethanol and stained with 0.5 mg/mL propidium iodide and 20 μg/mL RNAse A (Sigma). Propidium iodide-stained cells were analyzed using a FACSCalibur flow cytometer (Beckton-Dickinson), and the cell cycle phase distributions were determined with ModFit software (Verity).
Ubiquitylation assays
In vivo and in vitro ubiquitylation assays were performed essentially as described (Starostina et al. 2007). For in vivo ubiquitylation assays, Flag-Cdt2 and Flag-Skp2, or C. elegans Myc-CDT-2 and Flag-CKI-1, were coexpressed in HeLa cells (six-well plates) with HA-ubiquitin for 18 h. To prevent premature degradation of endogenous p21, HeLa cells were treated with 10 μm MG132 8 h after plasmid transfection, and then harvested 10 h after MG132 treatment. Lysates were prepared by boiling cells with 1% SDS, then diluted 10-fold with buffer composed of 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.1% NP-40, 1 mM DTT, 1 mM EDTA, Protease Inhibitor Cocktail, 50 μM LLnL, 10 mM N-ethyl maleimide (Sigma), and 10 mM iodoacetimide (Sigma). Endogenous p21 or Flag-CKI-1 was immunoprecipitated with anti-p21 or anti-Flag monoclonal antibodies, respectively, and analyzed by Western blot with anti-HA antibody for HA-ubiquitin detection or with either anti-p21 or anti-Flag antibodies. The in vivo ubiquitylation reaction of Figure 5E, which involved coexpressing Flag-Cdt2 and nontagged wild-type or PIP-box mutant p21, was performed as above, except the immunoprecipitation was from a nuclear fraction that was isolated by washing cells with buffer A (20 mM HEPES at pH 7.8; 10 mM KCl, 1.5 mM MgCl2, 0.34 M sucrose, 10% glycerol, 1 mM DTT, 1 mM EDTA) (Sarcinella et al. 2007), and then incubating cells for 5 min in 4°C buffer A containing 0.2% Triton X-100, Protease Inhibitor Cocktail, 50 μM LLnL, 10 mM N-ethyl maleimide, 10 mM iodoacetimide, 15 μM MG132, and 50 μM LLnL. After removing supernatant by low-speed centrifugation, nuclei were washed in the same buffer without Triton X-100, then boiled with 1% SDS in preparation for immunoprecipitation.
For in vitro ubiquitylation assays, 3xFlag-Cdt2 and p21 were coexpressed in HEK293T cells for 48 h. Eight hours before harvesting, 10 μM of MG132 and 2.5 mM HU were added to the cell culture. Cells were lysed with NP-40 Buffer supplemented with 1 mM Na3VO4, 100 μg/mL of DNase I (Roche), and 10 mM MgCl2. Flag-Cdt2 was immunoprecipitated from cell lysates with anti-Flag antibody in the presence of 10 mM EDTA and 2.5 mM o-phenanthroline. To each precipitated Flag-Cdt2 immunocomplex, 20 μL of ubiquitylation reaction mix was added, which contained 50 mM Tris-HCl (pH 7.4), 5 mM MgCl2, 2 mM sodium fluoride, 10 nM okadaic acid, 2 mM ATP, 0.6 μL of energy regeneration solution (Biomol Int., LP), 0.6 mM DTT, 1 μg HA-ubiquitin (Boston Biochem), 100 ng E1 (Biomol International, LP), 400 ng E2-UbcH5a, 200 ng UbcH3 (Biomol International, LP), 2 μM ubiquitin aldehyde (Biomol International, LP), and 15 μM MG132. Reactions were incubated for 90 min at 30°C and terminated by boiling with 1% SDS. p21 was secondarily immunoprecipitated and analyzed by Western blot.
Acknowledgments
We are grateful to S. Angers, B.E. Clurman, J. Jin, M. Pagano, Y. Xiong, and H. Zhang for reagents; J.G. Nelson and the UGA Flow Cytometry facility for DNA content analysis; the Caenorhabditis Genetics Center for strains; and members of the Kipreos laboratory for critical reading of the manuscript. This work was supported by grants from the NIH National Institute of General Medical Sciences (NIGMS) (R01GM055297 and R01GM074212) to E.T.K.
References
- Amador V., Ge S., Santamaria P.G., Guardavaccaro D., Pagano M. APC/C(Cdc20) controls the ubiquitin-mediated degradation of p21 in prometaphase. Mol. Cell. 2007;27:462–473. [Europe PMC free article] [Abstract] [Google Scholar]
- Angers S., Li T., Yi X., MacCoss M.J., Moon R.T., Zheng N. Molecular architecture and assembly of the DDB1–CUL4A ubiquitin ligase machinery. Nature. 2006;443:590–593. [Abstract] [Google Scholar]
- Arias E.E., Walter J.C. Replication-dependent destruction of Cdt1 limits DNA replication to a single round per cell cycle in Xenopus egg extracts. Genes & Dev. 2005;19:114–126. [Europe PMC free article] [Abstract] [Google Scholar]
- Arias E.E., Walter J.C. PCNA functions as a molecular platform to trigger Cdt1 destruction and prevent re-replication. Nat. Cell Biol. 2006;8:84–90. [Abstract] [Google Scholar]
- Arias E.E., Walter J.C. Strength in numbers: Preventing rereplication via multiple mechanisms in eukaryotic cells. Genes & Dev. 2007;21:497–518. [Abstract] [Google Scholar]
- Aspock G., Kagoshima H., Niklaus G., Burglin T.R. Caenorhabditis eleganshas scores of hedgehog-related genes: Sequence and expression analysis. Genome Res. 1999;9:909–923. [Abstract] [Google Scholar]
- Ausubel F.M., Brent R., Kingston R.E., Moore D.D., Seidman J.G., Smith J.A., Struhl K., editors. Current protocols in molecular biology. Wiley; New York: 1994. Saccharomyces cerevisiae; pp. 13.0.1–13.14.17. [Google Scholar]
- Bashir T., Dorrello N.V., Amador V., Guardavaccaro D., Pagano M. Control of the SCF(Skp2–Cks1) ubiquitin ligase by the APC/C(Cdh1) ubiquitin ligase. Nature. 2004;428:190–193. [Abstract] [Google Scholar]
- Besson A., Dowdy S.F., Roberts J.M. CDK inhibitors: Cell cycle regulators and beyond. Dev. Cell. 2008;14:159–169. [Abstract] [Google Scholar]
- Bornstein G., Bloom J., Sitry-Shevah D., Nakayama K., Pagano M., Hershko A. Role of the SCFSkp2 ubiquitin ligase in the degradation of p21Cip1 in S phase. J. Biol. Chem. 2003;278:25752–25757. [Abstract] [Google Scholar]
- Bosu D.R., Kipreos E.T. Cullin-RING ubiquitin ligases: Global regulation and activation cycles. Cell Div. 2008;3:7. 10.1186/1747-1028-3-7. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
- Carrano A.C., Eytan E., Hershko A., Pagano M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nat. Cell Biol. 1999;1:193–199. [Abstract] [Google Scholar]
- Cazzalini O., Perucca P., Savio M., Necchi D., Bianchi L., Stivala L.A., Ducommun B., Scovassi A.I., Prosperi E. Interaction of p21(CDKN1A) with PCNA regulates the histone acetyltransferase activity of p300 in nucleotide excision repair. Nucleic Acids Res. 2008;36:1713–1722. [Europe PMC free article] [Abstract] [Google Scholar]
- Dulic V., Stein G.H., Far D.F., Reed S.I. Nuclear accumulation of p21Cip1 at the onset of mitosis: A role at the G2/M-phase transition. Mol. Cell. Biol. 1998;18:546–557. [Europe PMC free article] [Abstract] [Google Scholar]
- Furukawa M., Andrews P.S., Xiong Y. Assays for RING family ubiquitin ligases. Methods Mol. Biol. 2005;301:37–46. [Abstract] [Google Scholar]
- Giniger E., Varnum S.M., Ptashne M. Specific DNA binding of GAL4, a positive regulatory protein of yeast. Cell. 1985;40:767–774. [Abstract] [Google Scholar]
- Hallier M., Lerga A., Barnache S., Tavitian A., Moreau-Gachelin F. The transcription factor Spi-1/PU.1 interacts with the potential splicing factor TLS. J. Biol. Chem. 1998;273:4838–4842. [Abstract] [Google Scholar]
- He Y.J., McCall C.M., Hu J., Zeng Y., Xiong Y. DDB1 functions as a linker to recruit receptor WD40 proteins to CUL4–ROC1 ubiquitin ligases. Genes & Dev. 2006;20:2949–2954. [Europe PMC free article] [Abstract] [Google Scholar]
- Higa L.A., Zhang H. Stealing the spotlight: CUL4–DDB1 ubiquitin ligase docks WD40-repeat proteins to destroy. Cell Div. 2007;2:5. 10.1186/1747-1028-2-5. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
- Higa L.A., Banks D., Wu M., Kobayashi R., Sun H., Zhang H. L2DTL/CDT2 interacts with the CUL4/DDB1 complex and PCNA and regulates CDT1 proteolysis in response to DNA damage. Cell Cycle. 2006a;5:1675–1680. [Abstract] [Google Scholar]
- Higa L.A., Wu M., Ye T., Kobayashi R., Sun H., Zhang H. CUL4–DDB1 ubiquitin ligase interacts with multiple WD40-repeat proteins and regulates histone methylation. Nat. Cell Biol. 2006b;8:1277–1283. [Abstract] [Google Scholar]
- Hu J., Xiong Y. An evolutionarily conserved function of proliferating cell nuclear antigen for Cdt1 degradation by the Cul4–Ddb1 ubiquitin ligase in response to DNA damage. J. Biol. Chem. 2006;281:3753–3756. [Abstract] [Google Scholar]
- James P., Halladay J., Craig E.A. Genomic libraries and a host strain designed for highly efficient two-hybrid selection in yeast. Genetics. 1996;144:1425–1436. [Europe PMC free article] [Abstract] [Google Scholar]
- Jin Y., Lee H., Zeng S.X., Dai M.S., Lu H. MDM2 promotes p21waf1/cip1 proteasomal turnover independently of ubiquitylation. EMBO J. 2003;22:6365–6377. [Europe PMC free article] [Abstract] [Google Scholar]
- Jin J., Arias E.E., Chen J., Harper J.W., Walter J.C. A family of diverse Cul4–Ddb1-interacting proteins includes Cdt2, which is required for S phase destruction of the replication factor Cdt1. Mol. Cell. 2006;23:709–721. [Abstract] [Google Scholar]
- Jin Y., Zeng S.X., Sun X.X., Lee H., Blattner C., Xiao Z., Lu H. MDMX promotes proteasomal turnover of p21 at G1 and early S phases independently of, but in cooperation with, MDM2. Mol. Cell. Biol. 2008;28:1218–1229. [Europe PMC free article] [Abstract] [Google Scholar]
- Kamath R.S., Martinez-Campos M., Zipperlen P., Fraser A.G., Ahringer J. Effectiveness of specific RNAmediated interference through ingested double-stranded RNA in Caenorhabditis elegans. Genome Biol. 2001;2:RESEARCH0002.1–RESEARCH0002.10. 10.1186/gb-2000-2-1-research0002. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
- Kamura T., Hara T., Kotoshiba S., Yada M., Ishida N., Imaki H., Hatakeyama S., Nakayama K., Nakayama K.I. Degradation of p57Kip2 mediated by SCFSkp2-dependent ubiquitylation. Proc. Natl. Acad. Sci. 2003;100:10231–10236. [Europe PMC free article] [Abstract] [Google Scholar]
- Kim Y., Kipreos E.T. Cdt1 degradation to prevent DNA re-replication: Conserved and non-conserved pathways. Cell Div. 2007a;2:18. 10.1186/1747-1028-2-18. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
- Kim Y., Kipreos E.T. The Caenorhabditis elegans replication licensing factor CDT-1 is targeted for degradation by the CUL-4/DDB-1 complex. Mol. Cell. Biol. 2007b;27:1394–1406. [Europe PMC free article] [Abstract] [Google Scholar]
- Kim J., Kipreos E.T. Control of the Cdc6 replication licensing factor in metazoa: The role of nuclear export and the CUL4 ubiquitin ligase. Cell Cycle. 2008;7:146–150. [Abstract] [Google Scholar]
- Kim J., Feng H., Kipreos E.T. C. elegans CUL-4 prevents rereplication by promoting the nuclear export of CDC-6 via a CKI-1-dependent pathway. Curr. Biol. 2007;17:966–972. [Europe PMC free article] [Abstract] [Google Scholar]
- Lee J., Zhou P. DCAFs, the missing link of the CUL4–DDB1 ubiquitin ligase. Mol. Cell. 2007;26:775–780. [Abstract] [Google Scholar]
- Li A., Blow J.J. Cdt1 downregulation by proteolysis and geminin inhibition prevents DNA re-replication in Xenopus. EMBO J. 2005;24:395–404. [Europe PMC free article] [Abstract] [Google Scholar]
- Licitra E.J., Liu J.O. A three-hybrid system for detecting small ligand–protein receptor interactions. Proc. Natl. Acad. Sci. 1996;93:12817–12821. [Europe PMC free article] [Abstract] [Google Scholar]
- Machida Y.J., Dutta A. The APC/C inhibitor, Emi1, is essential for prevention of rereplication. Genes & Dev. 2007;21:184–194. [Europe PMC free article] [Abstract] [Google Scholar]
- Masih P.J., Kunnev D., Melendy T. Mismatch Repair proteins are recruited to replicating DNA through interaction with proliferating cell nuclear antigen (PCNA) Nucleic Acids Res. 2008;36:67–75. [Europe PMC free article] [Abstract] [Google Scholar]
- Melixetian M., Ballabeni A., Masiero L., Gasparini P., Zamponi R., Bartek J., Lukas J., Helin K. Loss of Geminin induces rereplication in the presence of functional p53. J. Cell Biol. 2004;165:473–482. [Europe PMC free article] [Abstract] [Google Scholar]
- Mikaelian I., Sergeant A. A general and fast method to generate multiple site directed mutations. Nucleic Acids Res. 1992;20:376. [Europe PMC free article] [Abstract] [Google Scholar]
- Moldovan G.L., Pfander B., Jentsch S. PCNA, the maestro of the replication fork. Cell. 2007;129:665–679. [Abstract] [Google Scholar]
- Nishitani H., Sugimoto N., Roukos V., Nakanishi Y., Saijo M., Obuse C., Tsurimoto T., Nakayama K.I., Nakayama K., Fujita M., et al Two E3 ubiquitin ligases, SCF–Skp2 and DDB1–Cu14, target human Cdt1 for proteolysis. EMBO J. 2006.;25:1126–1136. [Europe PMC free article] [Abstract] [Google Scholar]
- Nguyen V.Q., Co C., Li J.J. Cyclin-dependent kinases prevent DNA re-replication through multiple mechanisms. Nature. 2001;411:1068–1073. [Abstract] [Google Scholar]
- Pines J., Hunter T. Cyclins A and B1 in the human cell cycle. Ciba Found. Symp. 1992;170:187–196. [Abstract] [Google Scholar]
- Ralph E., Boye E., Kearsey S.E. DNA damage induces Cdt1 proteolysis in fission yeast through a pathway dependent on Cdt2 and Ddb1. EMBO Rep. 2006;7:1134–1139. [Europe PMC free article] [Abstract] [Google Scholar]
- Sansam C.L., Shepard J.L., Lai K., Ianari A., Danielian P.S., Amsterdam A., Hopkins N., Lees J.A. DTL/CDT2 is essential for both CDT1 regulation and the early G2/M checkpoint. Genes & Dev. 2006;20:3117–3129. [Europe PMC free article] [Abstract] [Google Scholar]
- Sarcinella E., Zuzarte P.C., Lau P.N., Draker R., Cheung P. Monoubiquitylation of H2A.Z distinguishes its association with euchromatin or facultative heterochromatin. Mol. Cell. Biol. 2007;27:6457–6468. [Europe PMC free article] [Abstract] [Google Scholar]
- Senga T., Sivaprasad U., Zhu W., Park J.H., Arias E.E., Walter J.C., Dutta A. PCNA is a cofactor for Cdt1 degradation by CUL4/DDB1-mediated N-terminal ubiquitination. J. Biol. Chem. 2006;281:6246–6252. [Abstract] [Google Scholar]
- Sheaff R.J., Singer J.D., Swanger J., Smitherman M., Roberts J.M., Clurman B.E. Proteasomal turnover of p21Cip1 does not require p21Cip1 ubiquitination. Mol. Cell. 2000;5:403–410. [Abstract] [Google Scholar]
- Starostina N.G., Lim J.M., Schvarzstein M., Wells L., Spence A.M., Kipreos E.T. A CUL-2 ubiquitin ligase containing three FEM proteins degrades TRA-1 to regulate C. elegans sex determination. Dev. Cell. 2007;13:127–139. [Europe PMC free article] [Abstract] [Google Scholar]
- Sutterluty H., Chatelain E., Marti A., Wirbelauer C., Senften M., Muller U., Krek W. p45SKP2 promotes p27Kip1 degradation and induces S phase in quiescent cells. Nat. Cell Biol. 1999;1:207–214. [Abstract] [Google Scholar]
- Tsvetkov L.M., Yeh K.H., Lee S.J., Sun H., Zhang H. p27(Kip1) ubiquitination and degradation is regulated by the SCF(Skp2) complex through phosphorylated Thr187 in p27. Curr. Biol. 1999;9:661–664. [Abstract] [Google Scholar]
- Vaziri C., Saxena S., Jeon Y., Lee C., Murata K., Machida Y., Wagle N., Hwang D.S., Dutta A. A p53-dependent checkpoint pathway prevents rereplication. Mol. Cell. 2003;11:997–1008. [Abstract] [Google Scholar]
- Waga S., Hannon G.J., Beach D., Stillman B. The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature. 1994;369:574–578. [Abstract] [Google Scholar]
- Yoshida K., Takisawa H., Kubota Y. Intrinsic nuclear import activity of geminin is essential to prevent re-initiation of DNA replication in Xenopuseggs. Genes Cells. 2005;10:63–73. [Abstract] [Google Scholar]
- Yu Z.K., Gervais J.L., Zhang H. Human CUL-1 associates with the SKP1/SKP2 complex and regulates p21(CIP1/WAF1) and cyclin D proteins. Proc. Natl. Acad. Sci. 1998;95:11324–11329. [Europe PMC free article] [Abstract] [Google Scholar]
- Zhong W., Feng H., Santiago F.E., Kipreos E.T. CUL-4 ubiquitin ligase maintains genome stability by restraining DNA-replication licensing. Nature. 2003;423:885–889. [Abstract] [Google Scholar]
- Zhu W., Chen Y., Dutta A. Rereplication by depletion of geminin is seen regardless of p53 status and activates a G2/M checkpoint. Mol. Cell. Biol. 2004;24:7140–7150. [Europe PMC free article] [Abstract] [Google Scholar]
Articles from Genes & Development are provided here courtesy of Cold Spring Harbor Laboratory Press
Full text links
Read article at publisher's site: https://doi.org/10.1101/gad.1703708
Read article for free, from open access legal sources, via Unpaywall: http://genesdev.cshlp.org/content/22/18/2507.full.pdf
Free after 12 months at www.genesdev.org
http://www.genesdev.org/cgi/reprint/22/18/2507
Free to read at www.genesdev.org
http://www.genesdev.org/cgi/content/abstract/22/18/2507
Free after 12 months at www.genesdev.org
http://www.genesdev.org/cgi/content/full/22/18/2507
Citations & impact
Impact metrics
Citations of article over time
Alternative metrics
Article citations
USP44 inactivation accelerates the progression of thyroid cancer by inducing ubiquitylation and degradation of p21.
Int J Biol Sci, 20(13):5223-5238, 23 Sep 2024
Cited by: 0 articles | PMID: 39430240 | PMCID: PMC11489182
DCAF2 regulates the proliferation and differentiation of mouse progenitor spermatogonia by targeting p21 and thymine DNA glycosylase.
Cell Prolif, 57(10):e13676, 04 Jun 2024
Cited by: 1 article | PMID: 38837535 | PMCID: PMC11471390
Deciphering the role of neddylation in tumor microenvironment modulation: common outcome of multiple signaling pathways.
Biomark Res, 12(1):5, 08 Jan 2024
Cited by: 4 articles | PMID: 38191508 | PMCID: PMC10773064
Review Free full text in Europe PMC
DCAF14 regulates CDT2 to promote SET8-dependent replication fork protection.
Life Sci Alliance, 7(1):e202302230, 08 Nov 2023
Cited by: 1 article | PMID: 37940188 | PMCID: PMC10631547
Pan-cancer analysis and experimental validation of DTL as a potential diagnosis, prognosis and immunotherapy biomarker.
BMC Cancer, 23(1):328, 10 Apr 2023
Cited by: 2 articles | PMID: 37038185 | PMCID: PMC10088150
Go to all (173) article citations
Other citations
Wikipedia
Data
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
PCNA-dependent regulation of p21 ubiquitylation and degradation via the CRL4Cdt2 ubiquitin ligase complex.
Genes Dev, 22(18):2496-2506, 01 Sep 2008
Cited by: 270 articles | PMID: 18794347 | PMCID: PMC2546691
Selective ubiquitylation of p21 and Cdt1 by UBCH8 and UBE2G ubiquitin-conjugating enzymes via the CRL4Cdt2 ubiquitin ligase complex.
Mol Cell Biol, 31(15):3136-3145, 31 May 2011
Cited by: 36 articles | PMID: 21628527 | PMCID: PMC3147600
Inactivation of the CRL4-CDT2-SET8/p21 ubiquitylation and degradation axis underlies the therapeutic efficacy of pevonedistat in melanoma.
EBioMedicine, 10:85-100, 16 Jun 2016
Cited by: 41 articles | PMID: 27333051 | PMCID: PMC5006603
Regulation of DNA Replication Licensing and Re-Replication by Cdt1.
Int J Mol Sci, 22(10):5195, 14 May 2021
Cited by: 9 articles | PMID: 34068957 | PMCID: PMC8155957
Review Free full text in Europe PMC
Funding
Funders who supported this work.
NIGMS NIH HHS (4)
Grant ID: R01GM055297
Grant ID: R01 GM055297
Grant ID: R01GM074212
Grant ID: R01 GM074212