Open access

Phosphorylation Mediated Regulation of Cdc25 Activity, Localization and Stability

Written By

C. Frazer and P.G. Young

Submitted: 21 November 2011 Published: 06 September 2012

DOI: 10.5772/48315

From the Edited Volume

Protein Phosphorylation in Human Health

Edited by Cai Huang

Chapter metrics overview

3,448 Chapter Downloads

View Full Metrics

1. Introduction

Dual specificity phosphatases of the Cdc25 family are critically important regulators of the cell cycle. They activate cyclin-dependent kinases (CDKs) at key cell cycle transitions such as the initiation of DNA synthesis and mitosis. They also represent key points of regulation for pathways monitoring DNA integrity, DNA replication, growth factor signaling and extracellular stress. Since their mis-regulation allows cells to function in a genetically unstable state, it is not surprising that these phosphatases are involved in transformation to a cancerous state. Cdc25 phosphatases are heavily regulated by phosphorylation. Many regulatory phosphorylation sites on Cdc25 influence catalytic activity, substrate specificity, subcellular localization and stability. This chapter summarizes the current literature on the phospho-regulation of these proteins.

Advertisement

2. Yeast genetics and Xenopus oocyte maturation – Setting the stage

The study of cell division in eukaryotes was dramatically changed with the isolation of temperature sensitive “cell division cycle” (cdc-) mutants of the yeasts Schizosaccharomyces pombe and Saccharomyces cerevisiae. These mutants arrested uniformly at a particular cell cycle stage and uncoupled cell growth from cell cycle progression [1-3]. S. pombe cells are cylindrical cells growing from the tips while keeping a constant diameter [3]. At the restrictive temperature, fission yeast cdc- mutants arrest at their restriction point and become abnormally elongated. Within the fission yeast cdc- collection was the cdc25-22 mutation which arrests at mitotic entry when incubated at its restrictive temperature [3]. The collection also included cdc9-50 which divided at approximately half the length of a wildtype cell; it was later renamed wee1-50 in reference to its small size [4]. The wee1-50 mutation suppresses the cell cycle phenotype of cdc25-22 demonstrating that these two proteins act in opposition during the G2/M transition [5]. Analysis of these mutations showed that Cdc2 is rate limiting for mitotic entry and is inhibited by Wee1 and activated by Cdc25 [6,7]. Regulation of Cdc2 by Wee1 and Cdc25 in fission yeast was one of the first connected pathways in cell cycle research in any organism. The gene names Cdc25 and Wee1 are used in almost all organisms today.

Concurrently, Maturation Promoting Factor (MPF) was discovered through a series of elegant cytoplasmic transfer experiments conducted on frog, starfish and sea urchin oocytes [8-10]. Immature Xenopus oocytes arrest at prophase I. Progesterone induced MPF activation induces maturation whereby oocytes complete meiosis I and arrest at meiotic metaphase II [9,11]. Microinjection of cytoplasm from a mature oocyte into an immature oocyte also induces maturation [8,9]. This bioassay for maturation promoting factor (MPF) activity allowed it to be tracked through the meiotic and later mitotic cycles. Activity falls after anaphase I, rising again as cells enter prophase II. Although MPF activity is high, mature oocytes arrest due to the presence of a second soluble factor called Cyto-Static Factor (CSF) [9]. Subsequent entry of the sperm nucleus causes an influx of extracellular calcium, ultimately removing CSF inhibition and serving as the trigger for completion of meiosis and the start of mitotic divisions. Extracts from prophase II arrested mature oocytes can be induced to undergo alternating DNA synthesis and mitotic phases by the addition of calcium, providing an excellent cell free system for studying MPF regulation [12]. A peak of MPF activity accompanies each round of mitosis, reaching maximal activity during meiotic prophase, followed by a catastrophic drop as chromosomes segregate at anaphase. Fertilization is followed by a rapid succession of 12 rounds of DNA synthesis and cell division with no intervening gap phases, driven solely by maternally derived mRNA [13]. Gap phases are re-established at the mid-blastula transition followed by typical somatic cell cycles [14]. MPF consists of three proteins: Cdk1 kinase, cyclin B (Cdc2 [19‐21]. and Cdc13 [22,23].respectively in fission yeast) and the small regulatory protein Suc1 [15-19]. The activator of both MPF in Xenopus and the Cdc2-Cdc13 complex in S. pombe is Cdc25 phosphatase [24,25].

The cytoplasmic transfer experiments showed that MPF activity correlates with increased protein phosphorylation in donor oocytes [26], targeting a large number of nuclear proteins [27,28]. MPF is a histone H1 kinase, an activity which is still used today to measure CDK function [29]. MPF induces many of the cytological changes associated with mitosis such as nuclear envelope breakdown, chromosome condensation and mitotic spindle formation [30-32]. Mass spectrometry has shown that the single Cdk1 homologue in budding yeast (CDC28) phosphorylates 547 sites on 308 proteins in vivo [33]. The regulation of only about 75 of these CDC28 substrates has been examined in any detail [34]. Undoubtedly, the network of CDK mediated phosphorylation events in vertebrate cells will turn out to be far more complex.

Advertisement

3. Taxonomic distribution, duplication and divergence of Cdc25 homologues

Cdc25 is present in all eukaryotic cells. The yeasts, S. cerevisiae and S. pombe, possess a single Cdc25 protein (referred to as MIH1 in the former) [35]. Duplication and divergence led to three Cdc25 paralogues in vertebrates, Cdc25A, Cdc25B, and Cdc25C [36,36-40]. Cdc25A acts early in the cell cycle, regulating the G1/S transition, whereas Cdc25B and Cdc25C act at G2/M. Human Cdc25A, Cdc25B and Cdc25C have several isoforms through alternative splicing [41-44]. Cdc25C was discovered based on sequence similarity to fission yeast Cdc25 by using degenerate PCR primers to conserved residues in the catalytic region [38]. Human Cdc25A and Cdc25B were cloned by complementation of the temperature sensitive cdc25-22ts allele of S. pombe, demonstrating the strong conservation of function in this protein family [37]. Xenopus Cdc25A and Cdc25C were initially discovered based on sequence similarity to human Cdc25C [40,45]. Injection of recombinant Xenopus Cdc25A or Cdc25C into oocytes induces their maturation, the first direct indication that this protein was a positive regulator of MPF [40,45,46]. Xenopus Cdc25C also complements the cdc25-22ts mutation in fission yeast [45]. Four Cdc25 homologues are present in Caenorhabditis [47], whereas Drosophila contains two, string and twine. [48,49]. The conserved C-terminal domain of string contains the intrinsic phosphatase activity against a Cdk1-derived peptide containing phosphorylated Y15 and against the synthetic substrate p-nitrophenyl phosphate [50]. Few plant Cdc25 homologues can be found by sequence comparison in the NCBI database and those only in unicellular algae, eg Ostreococcus tauri [51]. In Arabidopsis thaliana [52]. the full length protein is only 146 residues long representing only the phosphatase domain and it shows marginal similarity to vertebrate and fission yeast Cdc25. Overexpression of A. thaliana Cdc25 causes premature mitotic entry in fission yeast [53].

Advertisement

4. Regulation of cell cycle transitions by Cdc25

4.1. The fission yeast G2/M transition – The prototype Cdc25/cyclin-CDK circuit

Regulation of the transition from G2 to mitosis in S. pombe is typical of the positive feedback loops seen between Cdc25 and CDKs in all systems. Cdc2 (Cdk1) drives all cell cycle transitions in fission yeast [20]. During G2 and early mitosis Cdc2 complexes with the B-type cyclin Cdc13 [54,55], the only essential cyclin in fission yeast [56]. Whereas Cdc2 is constitutively expressed throughout the cell cycle [57], Cdc25 and Cdc13 begin to accumulate in G2 and are degraded during mitotic entry [58-60]. Prior to M-phase, the Cdc2-Cdc13 complex is kept inactive by phosphorylation of threonine 14 (T14) and tyrosine 15 (Y15) [61,62]. Y15 is phosphorylated by the S-phase specific kinase Mik1 [63,64] and T14 and Y15 are modified by Wee1 [63,65-67]. In G2 Cdc25 translocates to the nucleus via the importin-β homologue Sal3 [68]. After cells grow to a critical size for mitotic entry, Cdc25 becomes active and dephosphorylates Cdc2 Y15 [69]. Cdc25 is activated and hyperphosphorylated by Cdc2 as the cell enters mitosis [58, 70]. CDKs phosphorylate a serine or threonine residue in the context of a consensus site (S/T)PX(K/R) [71,72]. The positions of the Cdc2 phosphorylation sites on Cdc25 have not been determined, but mutagenesis of 15 potential CDK consensus sites abrogates phosphorylation by Cdc2-Cdc13 [73]. In S. pombe, the feedback loop between Cdc25 and Cdc2-Cdc13 is strengthened by the involvement of the S. pombe Polo kinase homologue Plo1 which phosphorylates Cdc25 downstream of Cdc2 activation [74]. After the metaphase-anaphase transition, Polo kinase activation encourages cyclin degradation through activation of the anaphase promoting complex (APC) [75]. Thus, this kinase simultaneously ensures that Cdc2 activation will be robust, and brief, as its cyclin partner Cdc13 is degraded by the APC immediately following anaphase initiation.

4.2. Vertebrate cell cycle

While fission yeast Cdc25 is solely involved in the G2/M transition, Cdc25 orthologues in vertebrates also play a role in G1 and S-phase progression. Vertebrates have several CDK-cyclin complexes which participate in these transitions through positive feedback loops with Cdc25. In addition to associating with cyclin B, Cdk1 associates with cyclin A during late S-phase and G2 [76]. A second CDK, Cdk2, was discovered as a cDNA which could complement the loss of the budding yeast Cdk1 homologue, CDC28 [77]. Cdk2 functions early in the cell cycle and is likewise negatively regulated by phosphorylation of Y15 [78,79]. It forms a complex with Cyclin A and Cyclin E [80,81]. Cdk4 and Cdk6 operate early in G1 in association with D-type cyclins [82]. In many cases a particular cyclin class (ie. A, B, D, E) has multiple members. For the sake of clarity, cyclins will be referred to by their subtype only. Furthermore, only the CDKs and cyclins directly responsible for cell cycle transitions in concert with Cdc25 orthologues will be discussed. For instance, Cdk1/cyclin B is activated by a CDK-Activating Kinase (CAK), a complex of Cdk7 and cyclin H [83], but as CAKs are not activated by Cdc25 they are outside the scope of this review.

4.2.1. Cell cycle re-entry from Go

Most somatic cells spend their time in Go. Cells in Go may commit to entry into the cell cycle when they receive stimuli in the form of growth factors The cells then deactivate the cell cycle repressors which have kept them in Go and transcribe the positive regulators of the next cell cycle transition, G1/S.

In non-dividing cells, the Retinoblastoma protein (Rb) binds to E2F thus preventing transcription of genes required for cell cycle progression including cyclin D, cyclin A and Cdc25A [84-87]. (Figure 1) After exposure to growth factors, cells in Go re-enter the cell cycle through activation of the Ras pathway via the Raf/MAP (Mitogen Activated Protein) kinase pathway [88]. This leads to the degradation of the Cdk4 inhibitors p15INK4B and p16INK4A and the Cdk2 inhibitors p27KIP, p21CIP and induction of cyclin D. Cdk4 and Cdk6 bound to cyclin D inhibit the Rb protein [89] allowing transcription of cyclin E and cyclin A [89,90]. Cdc25A activates Cdk4-cyclin D but not Cdk6-cyclin D in vitro. [91]. Cyclin E associates with Cdk2 in late G1 and helps complete the inhibition of Rb [89,92].

4.2.2. The G1/S transition

Cdc25A is transcribed following relief of Rb-mediated transcriptional repression, reaching its maximal level at the end of G1 and dephosphorylating Y15 on Cdk2 [87,91]. Cdk2 is phosphorylated on Y15 by Wee1 [93]. Dephosphorylation of Cdk2 takes place via both Cdc25A and Cdc25B [94-97]. Cdk2 immunoprecipitated from cell lysates where Cdc25A has been overexpressed has high histone H1 kinase activity and low levels of Y15 phosphorylation [91]. Such overexpression accelerates entry into S-phase through activation of Cdk2-cyclin E [91,98]. DNA-synthesis can be blocked in these cells by injecting them with anti-Cdc25A antibodies [99]. Cdk2-cyclin E and Cdc25A are mutually activated by a positive feedback loop allowing passage of the G1/S boundary [95]. Depletion of Cdk2 or cyclin E prevents phosphorylation of Cdc25A and recombinant Cdc25A can be activated by Cdk2-cyclin E in vitro [95]. In addition phosphorylation of Cdc25A by Cdk2 destabilizes the phosphatase. Conversely, exposure of cells to CDK inhibitors roscovitine and olomoucine causes stabilization of Cdc25A [99].

Figure 1.

Positive feedback loops between Cdk-cyclin complexes and Cdc25 family members drive cell cycle transitions.

S-phase initiation requires activation of DNA replication proteins by Cdk2-cyclin A. Injecting G1 cells with anti-cyclin A antibodies stops entry into S-phase [100]. Phosphorylation of the essential DNA replication initiator Cdc6 by Cdk2-cyclin A leads to its nuclear import [101,102]. Cdk2 is recruited to chromatin by the replication initiation factor Cdc45 where it phosphorylates histone H1 and induces chromosome de-condensation [103]. As S-phase progresses high Cdk2-cyclin A activity induces degradation of Cdc6, preventing re-initiation of DNA synthesis at origins which have already fired [102].

Cdc25B has a role late in S-phase as Cdc25B immunoprecipitated from late S-phase HeLa cell extracts is phosphorylated, activated, and able to dephosphorylate Cdk2-cyclin A [97]. The murine homologue of Cdc25B purified from S-phase extracts promotes cyclin A and cyclin E associated histone H1 kinase activity in vitro [94]. In addition, siRNA knockdown of human Cdc25B causes a delay in initiation of DNA synthesis [104]. The Cdk1-cyclin A complex regulates late origin firing in S-phase. A constitutively activated Cdk1 allele increases firing of late S-phase origins, whereas a loss of function temperature sensitive allele of Cdk1 has a defect in late origin firing at the restrictive temperature [76].

4.2.3. The G2/M transition

Unlike the simple circuit of Cdc25-Cdc2 activation in fission yeast, the vertebrate G2/M transition involves a series of interconnected loops with positive and negative inputs from a variety of pathways. This led to what could be considered the “traditional model” of G2/M transition in vertebrates with respect to Cdc25 regulation by CDK-cyclin complexes. In reality things may be more complex. Cdc25C is not explicitly required for mitotic entry; siRNA knockdown of Cdc25C does not prevent the G2/M transition [105]. In addition, mouse lines which lack Cdc25B and/or Cdc25C are viable with the only obvious phenotype being a defect in oocyte maturation observed in cdc25B-/- mice [106-108]. In addition, the creation of CDK and cyclin knockout mice has revealed a network of compensatory mechanisms which cloud the traditional model. Cdk2-/-, Cdk4-/-, or Cdk6-/- mice show some abnormalities as adults, but are not embryonic lethal indicating that these Cdks are not individually essential for cell division [92]. There is considerable redundancy. Mitotic entry is best viewed as a three step process. First, Cdc25B and CDK-cyclin A are activated followed by basal activation of Cdk1-cyclin B at the centrosome [109]. Second, Cdk1-cyclinB, Cdc25B and Cdc25C localize to the nucleus. Third, Cdk1-cyclin B and Cdc25C mutually activate. Cdk1-cyclin B then induces overt mitotic events such as breakdown of the nuclear envelope and spindle formation. [81,110,111].

4.2.4. Activation of Cdc25B and Cdk1/2-cyclinA

Phosphorylation of human Cdc25B by Cdk1-cyclin A during G2 causes Cdc25B activation but also destabilizes the protein [112,113]. Cdc25B activates Cdk2-cyclinA in a positive feedback loop [114]. Cdk2-cyclin A mediated destabilization of Cdc25B has not been reported, although the Cdk1 and Cdk2 kinase complexes modify Cdc25B to approximately the same degree and have a similar set of substrates [113,115]. However, Cdk2 is the preferred binding partner of cyclin A and is more active than Cdk1-cyclin A during G2 [80,100]. Cdk2 cyclin A has two peaks of activation, one during S-phase and one prior to G2/M. [80,116]. Inhibition of Cdk2-cyclin A delays mitotic entry [109]. Depletion of Cdk1-cyclin A, activation of the CDK inhibitor p21cip, or addition of an inhibitory ATP analogue destabilizes Cdc25B in Sf9 cell extracts [112]. Similarly, in cycloheximide treated cells Cdc25B, but not Cdc25A or Cdc25C, is unstable and uniquely labile during the cell cycle [117].

Cyclin B accumulates throughout G2 but Cdc25A and Cdc25B are required to induce formation of the Cdk1-cyclin B complex at G2/M [118]. Cdk1-cyclin B interaction occurs earlier in G2 when Cdc25A or Cdc25B are overexpressed. Cdk1 and cyclin B are almost exclusively cytoplasmic during interphase with a small portion of the complex associating with the centrosome at G2/M [119-121]. A population of Cdk2-cyclinA is likewise localized to the centrosomes prior to prophase [116]. In Xenopus, centrosomal localization of Cdk1-cyclin B requires Aurora kinase [122]. Aurora is involved in a diverse set of mitotic events such as spindle assembly, centrosome maturation, and cytokinesis [123]. Aurora can also phosphorylate Cdc25B at S353 in vivo, activating the phosphatase [124]. (Table 1) Although some Cdk1-cyclin B is activated by Cdc25B [120,125], this does not represent a full mitotic activation and is only sufficient for progression as far as prophase. Injection of a dominant-negative mutant of Cdc25B into HeLa cells causes arrest in prophase with condensed chromosomes and disassembled nucleoli, but without nuclear envelope breakdown [109]. Microinjection of Cdk1-cyclin B lets cells pass this block and complete mitosis.

Table 1.

Summary of known phosphorylation sites on Cdc25 family members and the kinases responsible

4.2.5. Everybody into the nucleus

Cyclin B has a cytoplasmic retention sequence which is sufficient to induce cytoplasmic localization of the normally nuclear protein and contains a nuclear export signal (NES) [126]. Nuclear export is blocked by phosphorylation of S126 at the end of prophase [127-129]. Cyclin B is phosphorylated by Cdk1-Cyclin B as starfish oocytes pass the prophase II to metaphase II arrest [130]. Human cyclin B S126 is followed by a proline residue suggesting Cdk1-cyclin B autophosphorylation. Cyclin B is phosphorylated by the Xenopus Polo kinase homologue Plx1 on S133 and S147 (homologous to human cyclin B S177 and S181) enhancing its nuclear import [125,131]. Initial Cdc25B mediated activation of Cdk1-cyclin B may result in a priming autophosphorylation of S126 followed by docking of Polo kinase and inhibition of nuclear export. Such a Cdk1 mediated priming phosphorylation has been shown to induce Plx1 phosphorylation of Cdc25B (see below). Human Polo kinase Plk1 also deactivates Cdk1-cyclin B inhibitory kinases. Plk1 inhibits the cytoplasmic Cdk1 inhibitory kinase Myt1 [132,133]. Wee1 is phosphorylated on S53 and S123 by Plk1 and Cdk1-cyclin B, respectively, resulting in its degradation by β-TrCP [134].

The activated cytoplasmic pool of Cdk1-cyclin B phosphorylates Cdc25B on S160 inducing its nuclear import [135] (All phosphorylated residues on human Cdc25B are numbered according to the sequence of the longest splice variant Cdc25B3). Overexpression of human Cdc25B causes an increase in cells with condensed chromatin, whereas overexpression of Cdc25B-S160G does not induce mitotic entry. S160 phosphorylation does not affect the in vitro activity of human Cdc25B against a fluorescein diphosphate substrate, but instead positively regulates its nuclear import. In vitro phosphorylation assays show Cdk1-cyclin B can phosphorylate residues on Cdc25B which are not targeted by Cdk1-cyclin A or Cdk2-cyclin A [113]. Interestingly, Cdk1-cyclin A cannot phosphorylate Cdc25B that has previously been phosphorylated by Cdk1-cyclin B [112].

Plk1 is involved in human Cdc25B nuclear import following the initial activation of Cdk1-cyclin B. After addition of the Plk1 inhibitor thiophene benzimidazole, nuclear accumulation of GFP-Cdc25B is reduced. Conversely, expression of a constitutively active Plk1 mutant enhances Cdc25 nuclear localization [136]. Co-overexpression of Plk1 and Cdc25B in U2OS osteosarcoma cells induces chromosome condensation to a greater degree compared to cells expressing Plk1 or Cdc25B alone. This is partially dependent on the presence of a functional nuclear localization signal (NLS) in Cdc25B. Plk1 docking to Cdc25B requires prior phosphorylation of S50 by Cdk1/cyclin B [137]. Mass spectrometry identified thirteen phosphorylated Plk1 sites on Cdc25B in vitro and showed that T167, S209, T404, S465 and S513 appear to be particularly strong targets [137]. Plk1 is required for Cdk1-cyclin B activation, at least in part by negatively regulating Cdc25C nuclear export. Depletion of Plx1 from oocyte extracts prevents the activation of Cdc25C and Cdk1-cyclin B [138]. Unlike Cdc25B, phosphorylation of Cdc25C by Cdk1 is not a prerequisite for Plk1 targeting the phosphatase; Plk1 affects Cdc25C phosphatase activity and its localization. In vitro phosphorylation of Cdc25C enhances its ability to dephosphorylate kinase dead Cdc2 on Y15 [139]. Plk1 phosphorylates human Cdc25C on S198 which resides within the nuclear export signal (NES) of the phosphatase, promoting its nuclear localization [140]. Polo kinase family member Plk3 also interacts with human Cdc25C and phosphorylates it on S191, and S198 to a lesser degree [141]. Substitutions S191D, S198D or Plk3 overexpression result in constitutive nuclear localization, while siRNA knockdown of Plk3 or substitutions S191A and S198A leads to nuclear exclusion [141]. Another polo family member, Plk4, phosphorylates human Cdc25C on undetermined sites [142].

4.2.6. Full activation of Cdk1-CyclinB and Cdc25C

Activated Cdk1-cyclin B phosphorylates human Cdc25C on T48, T67, S122, T130, S168 and S214 in vitro and in vivo, driving a positive feedback loop which culminates in the phosphorylation of mitotic Cdk1 substrates [42,143]. Cdk1 phosphorylated Cdc25C has an increased Cdk1 Y15 phosphatase activity [143]. Recombinant Cdc25C can activate Cdk1 thereby increasing its histone H1 kinase activity [144]. Use of phospho-specific antibodies recently showed that phosphorylation of T48, T67 and T130 occur on spatially separate pools of human Cdc25C [145]. T67-phosphorylated Cdc25C is chromatin associated from prophase until telophase while T130-phosphorylated Cdc25C localizes to the centrosomes. T130 phosphorylation creates a Plk1 binding site on Cdc25C. Three distinct pools of T48, T67 and T130 mono-phosphorylated Cdc25C protein can be detected by immunoprecipitation with each individual phospho-specific antibody; Cdc25C pulled down with one antibody is not bound by the other two. In Xenopus oocyte extracts Cdc25C is heavily phosphorylated while cells undergo germinal vesicle breakdown (meiosis I) [45]. Xenopus Cdc25C residues T48, T67, T138, S205 and S285 are phosphorylated by Cdk1-cyclin A and Cdk1-cyclin B in vitro [146]. Mutation of the major targets, T48, T67 and T138, to alanine prevents Xenopus Cdc25C activation in vitro [147]. Although G2/M is not normally associated with Cdc25A function, it also contributes to Cdk1-cyclin B activation. Depleting cells of Cdc25A reduces Cdk1-cyclin B activation by approximately fifty percent [148]. In cells arrested in mitosis by addition of nocodazole, a microtubule polymerization inhibitor and spindle poison, Cdc25A is phosphorylated by Cdk1-cyclin B on S18 and S116. Substitution of these residues with alanine leads to Cdc25A instability. Cdc25A S40, S88, S261 and S283 are also Cdk1-cyclin B phosphorylated in vitro [149].

4.2.7. Mitotic exit

Following chromosome alignment on the metaphase plate a cascade of APC mediated degradation events occurs to reset conditions for the start of the next cell cycle [150]. The APC regulates two important processes required for completion of mitosis. First, it targets Securin, the inhibitory subunit of Separase, which is responsible for Cohesin cleavage and chromosome separation. Second, it targets cyclin A and cyclin B for destruction, inactivating Cdk1. Cyclin B is degraded after the metaphase-anaphase transition while cyclin A is degraded during metaphase. [121]. Cdk1 mediated phosphorylation of Cdc25 paralogues is reversed by Cdc14 family phosphatases. Cdk1-cyclin B phosphorylates human Cdc25A S18, S40, S88, S116, S261 and S283 in vitro [149]. Of these sites, human Cdc14A dephosphorylates S116, Cdc14B targets S88 and S261, while both phosphatases can dephosphorylate S40. Cdc14A was independently identified as also dephosphorylating Cdc25A S321 [151]. siRNA knockdown of Cdc14B leads to accumulation of phosphorylated Cdc25B and Cdc25C [149]. In S. pombe Cdc14 homologue Clp1 dephosphorylates Cdc25 and this is required for its ubiquitination and APC mediated proteolysis at the end of mitosis [73]. Similarly, in vertebrates Cdc25A and Cdc25B are targeted by the APC at mitotic exit [152,153].

Advertisement

5. Cdc25 phosphorylation by the DNA damage and replication checkpoint

DNA damage causes activation of checkpoints which delay cell cycle transitions to allow sufficient time for repair. Stalling of replication forks causes a similar cell cycle arrest, with additional need for stabilizing replication forks and/or modulating replication origin firing until the cause of the stalling is eliminated. Checkpoint effector kinases impinge on the central cell cycle machinery by phosphorylating Cdc25. This modification variously inhibits Cdc25 phosphatase activity, induces degradation or creates binding sites for 14-3-3 proteins which modify localization of the protein.

5.1. Fission yeast

Cell cycle arrest following DNA damage requires that Cdc2 is kept in a Y15- phosphorylated, inhibited state [154]. Cdc25 is inhibited through phosphorylation by Chk1 and Cds1 kinases in response to DNA damage and replication fork arrest, respectively [155-157]. Cells over-expressing Cdc25 or expressing a Y15F phospho-mimetic mutation of Cdc2 fail to arrest cell cycle progression after exposure to ionizing radiation [154]. In the absence of Cds1, Chk1 can cause cell cycle arrest following stalling of replication forks by hydroxyurea (HU) exposure. Cells lacking both kinases are unable to arrest [155]. Cds1 also phosphorylates multiple substrates to stabilize stalled replication forks and prevent the occurrence of inappropriate recombination events [158]. Upstream regulation of the DNA damage and replication checkpoint pathway occurs through activation of the ATM (Ataxia-telangiectasia mutated) homologue Rad3 through a well conserved signaling cascade [159].

Phosphorylation of Cdc25 by Chk1 and Cds1 creates binding sites for the 14-3-3 homologues Rad24 and Rad25 [160,161]. Phosphorylation and 14-3-3 binding stabilizes Cdc25, a phenomena referred to as “stockpiling”, thought to allow the cell to rapidly re-enter the cell cycle once replication or DNA damage arrest has been lifted [70]. The first Chk1/Cds1 phosphorylated Cdc25 residue identified, S99, partially impairs the replication and DNA damage checkpoint when mutated to alanine [157]. S99 modified Cdc25 is also phosphorylated on S192 and S359 by Cds1 and Chk1 in vivo and in vitro [156]. By phosphorylating Cdc25 in vitro with Cds1, nine additional sites were identified by mass spectrometry (S148, S178, S204, S206, T226, S234, T561, S567, T569) as well as the three sites previously known [162]. Nine sites between S99 and S359 are distributed through the poorly conserved N-terminal two thirds of the protein, while three sites reside in the extreme C-terminus. Alanine substitutions of all nine sites in the amino two thirds of Cdc25, cdc25(9A), overrides the DNA replication checkpoint when expressed under control of a relatively weak heterologous promoter [162]. However, when cdc25(9A) is expressed from the native cdc25+ locus under the control of its own promoter it does not cause a cell cycle phenotype, and the cell is checkpoint competent [60]. In addition, Cdc25(9A) is unstable following replication arrest suggesting redundancy in checkpoint control, such that Cdc25 which cannot be inhibited by Cds1 is eliminated from the cell. The S-phase specific Cdc2-Y15 kinase Mik1 is sufficient to prevent mitotic entry in these cells. Alanine substitutions of only the three C-terminal Cdc25 sites (T561, S567, T569) have a clear replication checkpoint defect in a mik1- background and appear to be involved in maintenance, but not establishment, of the DNA damage checkpoint. [305].

5.2. Vertebrate Cdc25 regulation by DNA damage and replication checkpoints

In vertebrate cells detection of DNA damage is relayed through ATM and ATR (ATM-Related) to two checkpoint effectors, Chk1 and Chk2. While the S. pombe ATM homologue Rad3 is involved in activation of both Chk1 and Cds1; DNA damage signaling in vertebrates shows separation of ATR-Chk1 and ATM-Chk2 axes [163,164]. The target of these effector proteins is determined by the cell cycle stage at which the damage occurs and the nature of the damage itself. ATM-Chk2 signaling is initiated by double strand breaks, while ATR-Chk1 is activated by stalled replication forks and single stranded breaks. The p38 MAP kinase pathway is critical for cell cycle arrest following UV induced DNA damage. (Figure 2)

5.2.1. G1/S and Intra-S checkpoints

The G1/S checkpoint prevents the start of DNA synthesis in the presence of DNA damage while the Intra-S checkpoint protects replication forks, prevents activation of late replication origins, and keeps the cell from entering mitosis until S-phase is completed. G1-S checkpoint arrest is manifested through inhibition of Cdc25A, thus preventing activation of Cdk4-cyclin D and Cdk2-cyclin E. The checkpoint also activates p53 resulting in the induction of the Cdk2 inhibitor p21CIP and the targeting of Cyclin E to the SCF complex to reinforce Cdk2 inhibition [165]. In rat fibroblasts UV induced DNA damage during G1 results in cell cycle arrest at the G1/S transition requiring inhibition of Cdc25A and phosphorylation of Cdk4-Y14 [166]. In U2OS osteosarcoma cells Cdk2-cyclin E kinase activity decreases and Y15 phosphorylation increases coincident with Cdc25A degradation following UV exposure [167]. Conversely, Cdc25A overexpression in UV exposed U2OS osteosarcoma cells results in bypass of the checkpoint and dephosphorylation of Cdk2-cyclin E. Cdc25A inhibition involves a combination of destabilization and inhibition of phosphatase activity by Chk1 [167]. Treatment with caffeine (an ATM/ATR inhibitor) or the Chk1 inhibitor UNC-01, or depletion of Chk1, stabilizes the phosphatase [167,168]. In humans, Chk1 phosphorylates Cdc25A S76, S124, S178, and T507 [168-170]. Cdc25A catalytic activity is reduced three-fold when it is phosphorylated by hChk1 in vitro [169]. S76 and S124 are phosphorylated following ionizing radiation resulting in Cdc25A instability [168,169,171-173]. Mutation of S76 to alanine stabilizes human Cdc25A [171,172]; however, neither S76A nor S124A overrides checkpoint arrest following ionizing radiation or UV [168]. Mouse cells homozygous for the Cdc25A S124A mutation display Cdc25A stabilization following ionizing radiation; [174] however, there are no changes in proportion of cells in S-phase, or radiation resistant DNA synthesis indicating their S-phase checkpoint is intact.

Human Cdc25A S76, S124 and/or S178 are identified in several publications as “S75, S123 and S177,” respectively [168,169,174-176]. In the original cloning of human Cdc25A [37], there are several substitutions in the N-terminus (Accession: AAA58415.1) and a one residue gap corresponding to residue R12 in all other full length human Cdc25A sequences in the NCBI database (Accession: P30304). Residues have been re-numbered as per “P30304” for the sake of consistency.

Cdc25A S76, S124, S178 and T507 match the consensus site for 14-3-3 binding, RXXpS/T [177]. However, only the Chk1 dependent phosphorylation of S178 and T507 results in association with 14-3-3 [170]. Substitution of these residues to alanine results in a complete loss of 14-3-3 interaction in vivo. Phosphorylation of T507 in particular, and subsequent 14-3-3 binding, interferes with Cdk1-cyclin B association by blocking a cyclin B docking site. A recent study suggests that a ternary complex between Cdc25A, 14-3-3γ and Chk1 is formed following ionizing radiation [178]. The Chk1/14-3-3γ interaction requires auto-phosphorylation of Chk1 S296. Substituting Chk1 S296 for alanine precludes 14-3-3 binding and Cdc25A S76 phosphorylation and deactivates the DNA damage checkpoint [178]. 14-3-3 proteins preferably exist as thermostable homo- and heterodimers. Each isoform in a heterodimer binding a different protein provides a common mechanism for bringing enzymes and their substrate proteins into close proximity [179,180].

Defects in the Intra-S checkpoint allow replication of damaged DNA [173,176,181]. In mammalian cells, DNA damage results in destabilization of Cdc25A and inhibition of Cdk2 [173]. Cdk2 is involved in loading the Cdc45 origin binding factor. Inhibition of Cdc25A stops further origin firing once DNA damage is detected [182]. Cdc25A is also unstable after HU induced replication fork stalling, which unlike in fission yeast, is controlled by activation of Chk1 in mammalian cells [183].

Figure 2.

Inhibitory phosphorylation of Cdc25 family members following DNA damage and replication arrest

5.2.1.1. Phosphorylation mediated degradation of Cdc25A by the SCF- βTrCP complex

The mechanism by which Cdc25A is destabilized following DNA structure checkpoint activation is well understood (Figure 3). Human Cdc25A phosphorylation by Chk1 following S-phase DNA damage causes degradation by F-box protein β-TrCP associated with the Skp1-cullin-Fbox (SCF) complex [171,184]. Mutating the destruction box or KEN box (APC interaction motifs) of Cdc25A does not stabilize the protein following exposure to ionizing radiation [152]. This indicates that SCF-mediated degradation of Cdc25A after DNA damage is independent of the cell cycle regulated APC-mediated degradation which occurs at mitotic exit. β-TrCP recognizes a degron motif of DSG(X)4S where both serine residues are phosphorylated [185]. siRNA knockdown of β-TrCP causes stabilization of Cdc25A, and radiation-resistant DNA synthesis in cells exposed to ionizing radiation [171,184]. Human Cdc25A contains such a motif: DS82GFCLDS88. The S88A substitution does not stabilize the phosphatase, suggesting that S88 phosphorylation is not explicitly required for β-TrCP binding to Cdc25A [171]. Following ionizing radiation, Chk1 primes Cdc25A for destruction through phosphorylation of S76 [171,186]. The NimA-related NEK11 kinase targets Cdc25A S82 and S88, within the DSG degron sequence [187]. Depletion of NEK11 causes a marked decrease in S82 and S88 phosphorylation in vivo, and prevents Cdc25A degradation following IR. NEK11, itself thought to be a Chk1 substrate, can directly phosphorylate S82 and S88 in vitro.

Following ATM activation, Chk2 phosphorylates and activates the oncogene p53, and inhibits its negative regulator MDM2 after ionizing radiation [188-191]. p53 then induces the Cdk2-cyclin E inhibitor p21WAF [192]. p53 also activates Glycogen Synthase Kinase (GSK-3β), which phosphorylates human Cdc25A S76 following a priming phosphorylation of S80 which can be targeted in vitro by Plk3 [193,194]. Plk3 is unique among Polo kinase family members in causing Cdc25A stabilization when knocked down with siRNA [194]. In Plk3 null mice Cdc25A is stabilized following DNA damage [195]. Plk3 also phosphorylates and activates p53 following DNA damage and contributes to Chk2 activation [196,197]. In HeLa cells, Casein Kinase 1α (CK1α) sequentially phosphorylates both S79 and S82 following priming phosphorylation of S76 by Chk1 or GSK-3β [198]. The CK1ε isoform negatively regulates Cdc25A by S82 phosphorylation in HEK293 cells [199]. S82 phosphorylation in vitro and in vivo takes place in response to DNA damage, dependent on CK1α first phosphorylating S79 [198]. S79A substitution prevents S82 phosphorylation. S76 phosphorylation appears to be the first target in the cascade, since it does not require prior phosphorylation of S79, T80, S82 or S88. Only alanine substitutions of S76, S79 and S82 impair β-TrCP interaction with Cdc25A and stabilize the phosphatase suggesting that T80 and S88 are not critical for Cdc25A degradation. Additional sites (S107, S156, S192, S279 S293) are phosphorylated by Chk1 or Cds1 in vitro but mutating any of these sites does not eliminate β-TrCP binding [200]. Although an interaction between phosphorylated Xenopus Cdc25A and β-TrCP has not been demonstrated, Chk1 is required for Cdc25A degradation at the mid-blastula transition through phosphorylation of S73 [201]. Interestingly, Xenopus Cdc25A lacks the first serine in the DSG(X)4S degron motif found in human Cdc25A, instead possessing DDG. Xenopus Cdc25A S73 lies just upstream of the mutated degron and is analogous to S76 in human Cdc25A. The DAG motif of Xenopus Cdc25A lies within a larger PEST motif which regulates stability of the phosphatase through β-TrCP binding, independent of Chk1 [202]. In mouse embryonic stem cells, which lack a G1/S DNA damage response, Cdk2 kinase activity is not affected by Cdc25A degradation following exposure to ionizing radiation [203]. Cdc25A degradation is independent of Chk1 and Chk2 but instead dependent on GSK-3β. Like Cdc25A, mammalian Cdc25B is unstable following HU induced arrest [117]. Cdc25B binds β-TrCP strongly and contains the residues DAG rather than the DSG degron motif [202,204]. In contrast, Cdc25B accumulates following G2 DNA damage checkpoint arrest induced by a variety of agents [205]. This is reminiscent of the “stockpiling” phenomena noted earlier in S. pombe [70]. β-TrCP interaction with Cdc25B may also be required for mitotic exit [204]. A Cdc25B-DDA degron mutant which cannot bind β-TrCP accelerates mitotic entry slightly, but has a significant delay completing mitosis and progressing to G1. This mitotic delay is due to an extended metaphase in which Cdc25B-DDA shows a high proportion of lagging, misaligned and bridged chromosomes as well as mis-oriented spindles [204].

Figure 3.

Multi-step phosphorylation cascades involved in targeting human Cdc25A for degradation by the β-TrCP SCF complex following DNA damage in S-phase and G2.

5.2.1.2. Cdc25A inhibition by 14-3-3 binding

Cdc25A S76, S124, S178 and T507 match the consensus site for 14-3-3 binding RXXpS/T [177]. However, only the Chk1 dependent phosphorylation of S178 and T507 results in association with 14-3-3 [170]. Substitution of these residues to alanine results in a complete loss of 14-3-3 interaction in vivo. Phosphorylation of T507 in particular, and subsequent 14-3-3 binding, interferes with Cdk1-cyclin B association by blocking a cyclin B docking site. A recent study suggests that a ternary complex between Cdc25A, 14-3-3γ and Chk1 is formed following ionizing radiation [178]. The Chk1/14-3-3γ interaction requires auto-phosphorylation of Chk1 S296. Substituting Chk1 S296 to alanine precludes 14-3-3 binding and Cdc25A S76 phosphorylation and deactivates the DNA damage checkpoint [178]. 14-3-3 proteins preferably exist as thermostable homo- and heterodimers. Each isoform in a heterodimer binding a different protein provides a common mechanism for bringing enzymes and their substrate proteins into close proximity [179,180].

Following exposure to ionizing radiation during G1 phosphorylation of Cdc25A on S124 by Chk2 prevents entry to S-phase [173]. Chk2 cannot efficiently phosphorylate Cdc25A on S76 and so cannot induce Cdc25A degradation by the β-TrCP route [206]. The mechanism by which S124 phosphorylation induces Cdc25A degradation is not clear because it is not required for degradation via β-TrCP [173]. S124 conforms to a 14-3-3 phospho-serine binding site, but doesn’t bind 14-3-3 [170]. In contrast to the effects of Cdc25A phosphorylation sites discussed thus far, modification of Xenopus Cdc25A T504 by Chk1 negatively regulates interaction with Cdk1-cyclin A, Cdk1-cyclin B and Cdk2-cyclin E but in a 14-3-3 independent manner [207].

5.2.2. G2/M DNA damage checkpoint

The response to damage in G2 is dependent on the nature of the damage signal. Exposure to UV activates p38 MAP kinase and checkpoint arrest is independent of ATM and ATR since the arrest is not caffeine sensitive [208]. The primary target following UV irradiation is Cdc25B and although p38 can phosphorylate Cdc25C in vitro, UV exposure does not affect the Cdc25C/14-3-3 interaction [208]. Ionizing radiation activates ATM and ATR and results in Cdc25C phosphorylation by Chk1 and Chk2 [209].

5.2.2.1. Cdc25B inhibition following UV induced DNA damage

A number of conflicting reports have appeared relating to Cdc25B regulation following UV exposure. Some groups have reported that UV has no effect on Cdc25B protein levels [208,210], but others have shown that UV causes either MAP kinase mediated Cdc25B degradation or Cdc25B accumulation [205,208,211]. Cell line specific effects have no doubt contributed to these inconsistencies as human molecular biology relies heavily on transformed cell lines and mis-regulation of Cdc25B is a common phenomenon in tumors [212]. Cdc25B isolated from UV irradiated A2058 melanoma cells still retains a substantial portion of its Y15 phosphatase activity and is localized to the nucleus [213]. In HeLa cells Cdc25B is localized almost exclusively to the cytoplasm as detected by cell fractionation and immunofluorescence [120]. Variation in the apparatus used for UV irradiation could also have contributed to contradictory accounts of Cdc25B regulation. A recent re-examination of the effect of UV on Cdc25B showed that after 10 J/m2 exposure, Cdc25B levels did not decrease, although following 60 J/m2 Cdc25B was clearly downregulated [214]. Exposure of U2OS osteosarcoma cells to 10 J/m2 UV leads to Cdc25B nuclear export. Based on chemical inhibitor experiments Cdc25B downregulation is not mediated by ATM/ATR, p38 MAPK or JNK, but rather following 60 J/m2, by inhibiting Cdc25 translation. The eukaryotic initiation factor regulating Cdc25B expression, eIF2α, is phosphorylated and inhibited following UV exposure [215]. UV mediated DNA damage during G2 involves human Cdc25B S323 phosphorylation through the p38 kinase during interphase [208]. ATM/ATR inhibitor caffeine and the Chk1 inhibitor UNC-01 have no effect on UV mediated checkpoint arrest. Isoforms 14-3-3β and 14-3-3ε bind preferentially to Cdc25B phosphorylated S323, allowing its nuclear export [216]. Nuclear export of Cdc25B is abolished in cells expressing the Cdc25B S323A substitution, regardless of which 14-3-3 isoform is co-expressed [216]. Two amino-truncated Cdc25B isoforms localize to the nucleus in vivo and regulate recovery from G2/M checkpoint arrest but neither is required for mitotic entry [44]. Although one of these isoforms contains the DDG degron described above, both are more stable than the full length Cdc25B.

5.2.2.2. Inhibition of Cdc25C following DNA damage

Cdk1 activation is prevented by UV induced checkpoint activity coincident with the appearance of a phosphorylated form of Cdc25C [213]. In contrast with fission yeast Cdc25, which gradually accumulates in the nucleus during G2, human Cdc25C is primarily localized to the cytoplasm during interphase and only enters the nucleus at mitotic entry [217]. Thus, nuclear export of Cdc25C is not a requirement for G2 DNA damage response, since the phosphatase is already cytoplasmic at this time. However, exposure to ionizing radiation decreases the enzymatic activity of human Cdc25C [218]. Several research groups showed relatively early in the Cdc25 phosphorylation story that residue S216 is phosphorylated by Chk1 [219-221] and Chk2 in vitro. [209,222]. Phosphorylation of S216 results in 14-3-3 binding and nuclear export [219]. Residues surrounding S216, RSPS216MP, correspond to the canonical 14-3-3 consensus binding site RSXpSXP [177]. Cdc25C S216A mutants are unable to bind 14-3-3 [219]. This is likely due to close proximity of S216 to the NLS, leading to the obstruction of the import signal and trapping of the phosphatase in the cytoplasm [221,223]. A higher proportion of cells expressing Cdc25 S216A have nuclear abnormalities indicative of progression to mitosis prior to completion of DNA synthesis; this effect is exacerbated by addition of HU [217,219]. Such cells also have reduced ability to delay entry to mitosis following ionizing radiation exposure [224].

Although Cdc25C S216A is a relatively poor substrate for Chk1 compared to the wildtype protein, Chk1 can still execute some degree of phosphorylation on the mutant phosphatase [220]. This observation suggests the possibility that additional Cdc25C phosphorylation negatively regulates its enzymatic activity. Recent bioinformatics approaches to generate profiles from peptide binding arrays based on three diverse 14-3-3 binding sites have generated an improved 14-3-3 binding motif consensus [225]. This helped to identify two additional phosphorylated Cdc25C residues, Ser247 and Ser263, which interact with 14-3-3. Mutation of either residue to alanine reduces 14-3-3 binding, but neither of these mutant peptides was affected by Cdc25 S216 phosphorylation when expressed in cells. S263 was previously identified in an isolated report which showed phosphorylation of this residue induces Cdc25B nuclear export [226]. Cdc25C purified from cells treated with the topoisomerase II inhibitor etoposide is phosphorylated on S263, but S263A substitution results in enhanced nuclear localization. The kinase targeting this residue has yet to be determined experimentally. However, the residues surrounding S263 (KKTVpSLCD) conform to a Chk1 consensus site as Chk1 can tolerate a lysine (K) at the -3 position relative to the phosphorylated serine or threonine in vitro [227].

Regulation of Xenopus Cdc25C localization is similar to its human counterpart. In cultured Xenopus tissue Cdc25C is primarily in the cytoplasm while the Cdc25C S287A mutant, corresponding to S216 in human Cdc25C, is almost exclusively nuclear [228]. Cdc25C is phosphorylated on multiple sites by Chk1 but only S287 phosphorylation is required for 14-3-3 binding [229-231]. Although Xenopus Cdc25C can be made exclusively cytoplasmic by co-expression with 14-3-3ε, Cdc25C S287A is not affected. Nuclear export depends on the intrinsic 14-3-3ε nuclear export signal and re-import is prevented by blocking Cdc25C association with importin-α [228]. In egg extracts depleted of endogenous Cdc25C, expressing Cdc25C S287A accelerates mitotic entry relative to overexpression of the wildtype protein [229]. However, Cdc25C S287A has a less than two fold increase of in vitro Cdk1 Y15 dephosphorylation activity over wildtype Cdc25C when pre-incubated with 14-3-3ε. Thus, it seems likely that Xenopus Cdc25C phosphorylation and 14-3-3 binding regulates the phosphatase at the level of cellular localization, rather than inhibiting its activity per se.

Cdc25A is considered to regulate the G1/S transition in the “Traditional Model” of the human cell cycle but it also has a significant role in mitotic entry. As such, Cdc25A is an important target of the DNA damage checkpoint. In fact, mice lacking Cdc25B and Cdc25C do not have a G2/M checkpoint defect [106,107]. Phosphorylation by Chk1 causes degradation of Cdc25A following DNA damage during G2 ionizing radiation and exposure to the DNA intercalating agent adriamycin [172,181].

5.3. Chk1 regulation of Cdc25 orthologues in unperturbed cell cycles

Cdc25A, B and C are all phosphorylated by Chk1 in the absence of externally induced DNA damage. As Cdc25 phosphatases are such potent positive regulators of cell cycle transitions it is perhaps not surprising that the cell maintains their activity at a low level until their precise point of activation. Chk1 regulates human Cdc25A stability during unperturbed cell cycles. Phosphorylation of S82 and S88 can be detected using phospho-specific antibodies in unperturbed cells [184]. Depletion of the S82/S88 kinase NEK1 and S76 kinase CK1ε by siRNA, results in Cdc25A stabilization in the absence of DNA damage [187,199]. Cdc25A S124 phosphorylation by Chk1 also occurs in the absence of damage and destabilizes the phosphatase [168,169,200]. Inhibiting ATM/ATR, or a variety of upstream checkpoint components, also stabilizes in the absence of externally induced DNA damage which may indicate there is some basal level of spontaneous damage checkpoint signaling [232]. Cdc25A is also phosphorylated by Casein kinase 2β (CK2β) in a damage independent manner [233]. CK2 phosphorylates human Cdc25C T236 adjacent to the NLS in vitro [234]. A T236D mutation reduces β-importin binding, thus excluding Cdc25 from the nucleus [234]. CK2 phosphorylates Cdc25B on residues S186 and S187, just downstream from the KEN box, modestly increases its phosphatase activity, and potentially blocking APC mediated degradation [235].

Human Cdc25B is phosphorylated in vitro by Chk1 at S230 and S563 in the absence of DNA damage [236]. S230 phosphospecific antibodies show that Cdc25B modified on this residue is centrosome associated from S-phase until mitosis [236]. A population of Chk1 is localized to the centrosome during G2 and can prevent promiscuous Cdk1-cyclin B activation by Cdc25B [237]. S323 was previously identified as the major 14-3-3 binding site [238]. S151 and S230 account for the remainder of the interaction, but both need to be dephosphorylated before interaction with 14-3-3 is lost [239]. Human Cdc25B S563 resides in the extreme C-terminus of Cdc25B and is analogous to Cdc25A S504 and S. pombe Cdc25 T569. Cdc25B lacking the S323 phosphorylation site is almost exclusively nuclear. It is interesting that this residue is targeted by Chk1, but does not appear to bind 14-3-3 when phosphorylated. If the function of this phosphorylation is conserved between Cdc25A S504 and Cdc25B S563, phosphorylation may affect interaction with Cdk1-cyclin B [240].

Chk1 phosphorylation of human Cdc25C and nuclear export by 14-3-3 binding keeps the phosphatase cytoplasmic during unperturbed cell cycles [217]. Nuclear localization of Cdc25C(S216A) is enhanced, suggesting that part of the function of 14-3-3 binding is to obscure the NLS located adjacent to this residue [224]. Overexpression of Cdc25C(S216A) induces a higher degree of premature mitotic entry [217]. Xenopus Cdc25C is likewise phosphorylated on S287 by Chk1 during interphase [228,241]. Phospho-S287 is bound by 14-3-3ε and 14-3-3ζ obscuring the NLS and preventing nuclear import [228]. Xenopus Cdc25C is phosphorylated on T533 by Chk1, but not Chk2 [207]. Injecting Cdc25C T533A mRNA into Xenopus oocytes results in more rapid dephosphorylation of Cdk1 Y15 [207]. Again, this suggests that regulation of CDK-cyclin interaction with Cdc25 orthologues by C-terminal phosphorylation is a common mechanism for inhibiting cell cycle progression.

Although Cdc25A and Cdc25B are dispensable for embryonic development, Chk1 and ATR kinases are essential [242,243]. The Cdc25B/14-3-3 interaction is important for maintaining G2 arrest, and inhibiting germinal vesicle breakdown in Xenopus oocytes prior to progesterone exposure [244].

In S. pombe, Chk1 does not appear to negatively regulate Cdc25 in the absence of DNA damage. Loss of a negative regulator of Cdc25 is expected to cause the cell to divide at a reduced length. However, deletion of Chk1 does not cause a cell cycle phenotype [245]. In addition, expressing Cdc25 where all twelve putative Cds1/Chk1 phosphorylation sites are mutated to alanine does not cause acceleration of the cell cycle [60].

5.4. Cdk1 phosphorylation of Cdc25 precludes checkpoint mediated inhibition during mitosis

If Cdc25C is phosphorylated and inactivated during interphase via 14-3-3 binding, how is it then activated at mitotic entry? Cdk1-cyclin B phosphorylation of Cdc25C causes 14-3-3 dissociation and allows removal of interphase phosphorylations. Re-phosphorylation of these residues is simultaneously blocked. Cdk1-cyclin B thus potentiates Cdc25C for its pro-mitotic function and ensures that it remains active. The region surrounding S216 in human Cdc25C and S287 in Xenopus Cdc25C is a well conserved stretch in the N-terminal region of the two proteins; LYRSPS216MPE is identical between Human and Xenopus and contains S216 and S287, respectively.(Figure 4) In both organisms, the two serine residues upstream of the major phosphorylated 14-3-3 binding residue, S214 in human and S285 in Xenopus, is targeted by Cdk1-cyclin B [42,246-248]. In human cells phosphorylation of S214 precludes phosphorylation of S216 by Chk1 and 14-3-3 binding [42,246]. Substituting S214D prevents phosphorylation of S216 [246]. S216 is not phosphorylated during M-phase in vivo, and ionizing radiation in M-phase cells cannot induce its phosphorylation [246]. 14-3-3 is unable to bind Cdc25C immunoprecipitated from cells arrested in mitosis with nocodazole [219]. In Xenopus, Cdc25C S285 phosphorylation prevents the phosphorylation of S287 [247,249]. S285 can be phosphorylated by both Cdk1-cyclin B and Cdk1-cyclin A in vitro [146]. Until the mid-blastula transition, Xenopus Cdc25C is phosphorylated on S285 which precludes Chk1 mediated phosphorylation of S287 and subsequent 14-3-3 binding [247]. Human Cdc25C S214 is also phosphorylated in maturing human oocytes [250]. Removal of phospho-S287 bound 14-3-3 and phosphorylation of Xenopus Cdc25C S285 requires prior phosphorylation of Cdc25C T138 [249]. T138 is a substrate of Cdk1-cyclin B [146]. However, selective depletion of Cdk2 from Xenopus egg extracts using the N-terminus of the Cdk2 inhibitor p21, completely prevents removal of 14-3-3 from Cdc25C [251]. In Xenopus egg extracts Cdk2 is required for mitotic entry [81]. Other CDK phosphorylation sites on Xenopus Cdc25C such as T48 and T67 negatively regulate its activity in an S287 independent manner [249]. Phosphorylation of T138 is not sufficient to cause 14-3-3 dissociation [248]. How is 14-3-3 physically removed from phospho-S287? The pelleted fraction from ultracentrifuged interphase egg extracts contains a 14-3-3 dissociating activity, which was determined to be the intermediate filament component Keratin 8/18 [248]. Keratin is phosphorylated during mitosis, binds 14-3-3, and has a role in mitotic progression in hepatocytes [252]. Keratin may act as a “14-3-3 sink” during mitosis, stripping 14-3-3 from S287 [248]. Xenopus Cdc25C T138 corresponds with T130 in human Cdc25C. A phospho-T130 specific antibody shows that Cdc25C phosphorylated on this site localizes to the centrosome [145]. A localization for the de-inhibition of Cdc25C fits well with the putative centrosomal localization of Cdc25C activation.

PP1 phosphatase removes S287 phosphorylation once 14-3-3 has dissociated [251]. Binding of PP1 to Xenopus Cdc25C requires a docking motif “VXF”, amino acids 105-107, the loss of which prevents S287 dephosphorylation [251]. Phosphorylation of Xenopus Cdc25C S285 by Cdk1-cyclin B enhances recruitment of phosphatase PP1 to Cdc25C, inducing the dephosphorylation of S287 [249]. PP2A/B56δ dephosphorylates Xenopus Cdc25C T138 during interphase, mitotic exit and following replication arrest [248]. This maintains the phosphatase in a state where it can be inhibited by S287 phosphorylation. Inhibition of PP2A by okadaic acid prematurely induces mitotic entry in Xenopus egg extracts [253]. T138 is also dephosphorylated during replication arrest where B56δ is itself phosphorylated by Chk1, enhancing complex formation with PP2A [248]. PP2A mediated dephosphorylation of Cdc25 is assisted by the action of the Pin1 prolyl isomerase. Pin1 isomerizes the peptide bond between phospho-serine/threonine and proline placing their R-groups in a trans orientation [254]. This isomerization makes the phosphorylated residue a better substrate for the PP2A phosphatase [255]. In vitro, Pin1 decreases the catalytic activity of Cdc25C that has previously been phosphorylated by Cdk1 [256]. Pin1 is also involved in maintenance of replication checkpoint arrest in Xenopus encouraging the reversal of Cdk1 mediated Cdc25C phosphorylation [257]. In Humans, Cdc25C is deactivated in a similar manner. T130 on Cdc25C is dephosphorylated by PP2A [258]. Mitotic exit is delayed when PP2A is knocked down, suggesting that dephosphorylation of Cdc25C T130 in human cells is also important for the transition from M to G1 of the next cycle. A similar situation may exist where human Cdc25B1 S321 phosphorylation blocks phosphorylation of the major 14-3-3 binding residue S323 by p38 [246]. Expression of the phosphorylation-mimicking Cdc25B1 S321D prevents p38 mediated phosphorylation of S323 and abolishes binding by 14-3-3β and 14-3-3ε [246,259]. 14-3-3σ binding is unaffected by S321 phosphorylation [259]. 14-3-3σ preferentially interacts with Cdc25B3 S230 [216]. and is induced by p53 following activation by ATM/Chk2 [260].

Figure 4.

Cdk-cyclin mediated removal of inhibitory S216/S287 phosphorylation and 14-3-3 binding in human and Xenopus Cdc25C, respectively.

There are no Cdc2 phosphorylation motifs (S/TP) directly upstream of any of the twelve Cds1 in vitro phosphorylation sites in S. pombe. Large scale phosphoproteome analysis has detected S99, S178 and S359 phosphorylation of fission yeast Cdc25 in M-phase arrested cells [261]. Thus it appears that the vertebrate mechanism for reversing and preventing Chk1 phosphorylation of Cdc25C evolved relatively recently. However, parallels exist between some aspects of Cdc25 dephosphorylation between fission yeast and vertebrates. Treatment with okadaic acid or deletion of PP2A homologue Ppa2 causes premature mitotic entry in fission yeast [262,263]. Loss of ppa2 suppresses the cdc25-22ts mutation, a genetic interaction indicative of a negative regulator [263]. Loss of PP1 homologue Dis2 results in cell elongation, suggesting its role as a positive regulator of Cdc25 is conserved in S. pombe [264]. Temperature sensitive dis2 mutants have a defect in exit from mitosis, similar to the effect of siRNA inhibition of human PP2A [258,264].

Advertisement

6. Cdc25 phosphorylation by MAP kinase cascades

In addition to regulation by DNA damage and DNA replication checkpoints, Cdc25 is the target of several MAPK cascades responding to stress and mitogenic signals. A detailed description of the variety of MAPK pathways is outside of the scope of this manuscript, but excellent reviews are available [265]. In general, MAP kinase cascades involve the sequential activation of three kinases; a MAP kinase kinase kinase (MAPKKK) phosphorylates a MAP kinase kinase (MAPKK), which phosphorylates a MAP kinase (MAPK). There are three such cascades which are salient to our discussion of Cdc25 regulation.

6.1. Raf/MEK/ERK

The ERK1/ERK2 MAPKs are activated by Raf MAPKKKs working on MEK1/MEK2 MAPKKs. This cascade is primarily activated by extracellular signaling through receptor tyrosine kinases in response to mitogenic signals [266]. During Cyto-Static Factor arrest in mature Xenopus oocytes metaphase II arrest is enforced by the combined activity of the MEK/ERK pathway upregulating ribosomal subunit S6 kinase/CamKII homologue p90rsk, and APC inhibition by the Emi2 kinase [267]. Calcium influx as a result of fertilization activates p90rsk (a Ca2+/calmodulin dependent kinase II homologue) which phosphorylates Emi2 [268]. This allows APC activation, cyclin B degradation, and progression through meiosis II. Cdc25C is phosphorylated on S287 by p90Rsk in vitro resulting in its inhibition via 14-3-3 binding [269,270]. Conversely, Xenopus Cdc25C S317, T318 and S319 are phosphorylated by p90rsk orthologue Rsk2 which serves to activate the phosphatase in mature oocyte extracts [271]. The Xenopus ERK homologue, p42, phosphorylates Cdc25C T48, T138 and S205 in vitro [272]. T138 and S205 are also Cdk1-cyclinB targets [146], but T48 is uniquely phosphorylated by p42. In HeLa cells after activation of MEK/ERK signaling by addition of 12-O-tetradecanoylphorbol-13- acetate (TPA), G2/M transition is inhibited by degradation of Cdc25B [273]. This is mediated through MEK dependent phosphorylation of Cdc25B S249 by CamKII, an activator of human Cdc25C [274]. Inhibition of CamKII results in a G2 arrest. Overexpression of CamKII can also arrest cells in G2, but with a high level of Cdk1/Cyclin B activity [275]. Another member of the CamKII family, C-TAK1, phosphorylates human Cdc25C on S216 resulting in inhibition via 14-3-3 binding and nuclear export [276]. Xenopus Cdc25C can be phosphorylated by CamKII in vitro [277]. Lastly, p14arf upregulation in response to anti-proliferative signals results in human Cdc25C downregulation through MEK/ERK MAPK signaling [278]. Human Cdc25C becomes phosphorylated on S216 and is subsequently ubiquitinated and degraded [278].

6.2. p38 and JNK MAPKs

The p38 and JNK kinases activate in response to extracellular stimuli such as heatshock, oxidative stress, ionizing radiation, UV and growth factor deprivation. Both are activated by MAPKKs of the MKK family which are themselves activated by a large variety of MAPKKKs. We have already discussed the function of p38 in response to UV irradiation. p38 also phosphorylates human Cdc25A on S124 and S76 in response to osmotic stress, destabilizing the protein [168] and a 42 C heatshock causes p38 and Chk2 to phosphorylate S76 and S178 of human Cdc25A, respectively [175]. MAPKAP kinase 2 (MK2) functions downstream of p38 and regulates G1 and S-phase cell cycle progression in response to UV [210]. Downstream of p38, it phosphorylates RxxS/T motifs and activation of MK2 correlates with increased binding of 14-3-3 to Cdc25B. p38 and MK2 kinase form a tight complex and are imported into the nucleus together, so previous work showing that p38 directly phosphorylates S216 on Cdc25C and S323 on Cdc25B may in fact have been inadvertently monitoring MK2 activity [279]. Cdc25A may also be an MK2 substrate as MK2 knockdowns ablate the G1 and S-phase checkpoints. JNK activity targets two serines within the region DAGLCMDS101PS103P of the DSG degron on human Cdc25B [280]. Simultaneous S101A and S103A substitution prevents β-TrCP binding and Cdc25B ubiquitination. JNK also phosphorylates Cdc25C on S168 inhibiting its phosphatase activity [281,282]. This residue is transiently phosphorylated in vivo on nuclear Cdc25C prior to and after mitotic entry [281]. S168 is phosphorylated following UV irradiation and osmotic shock [281,282].

6.3. The S. pombe stress activated MAP kinase pathway

In fission yeast, Cdc25 is phosphorylated by the CamKII homologue Srk1 in response to extracellular stress [283]. Srk1 is activated downstream of the Spc1 MAPK, Wis1 MAPKK, and Win1 or Wak1MAPKKKs [284]. This phosphorylation occurs on residues also targeted by Cds1 as Cdc25(9A) is not sensitive to Srk1 mediated inhibition [283]. Srk1 phosphorylation of Cdc25 results in its nuclear export, similar to the response to DNA damage and replication arrest.

Advertisement

7. Other kinases which phosphorylate Cdc25s

PKA prevents oocyte maturation by inhibition of Polo kinase mediated Cdc25 activation, and deactivating the Mos/MEK/ERK MAP kinase cascade which inhibits Myt1 [285]. Progesterone exposure in Xenopus oocytes down regulates adenylate cyclase, lowering cyclicAMP levels and consequently deactivating PKA. Prior to maturation, Xenopus Cdc25C is also inhibited by PKA phosphorylation of S287 and T318 in vitro [286]. Murine PKA phosphorylates Cdc25B S321, negatively regulating the phosphatase; S321A mutants cause enhanced germinal vesicle breakdown when injected into mouse oocytes [287].

Pim1 is a serine/threonine kinase induced by the SAT3 and SAT5 transcription factors following cytokine exposure thus linking pro-proliferative signals to the cell cycle control machinery [288]. Pim1 phosphorylates and activates Cdc25A and represses the Cdk4/6 inhibitor p21CIP to encourage the G1/S transition [289,290]. Pim1 is also able to phosphorylate and inhibit the CamKII homologue c-TAK and accelerate Cdc25C mediated mitotic entry [291] PAR-1/MARK (partitioning-defective 1/Microtubule affinity-Regulating Kinase) protein homologue pEG3 phosphorylates human Cdc25A S263, Cdc25B S169 and Cdc25C S216 [292,293]. Overexpressing pEg3 results in G2 arrest which can be reversed by co-expressing Cdc25B [292]. Anti-phospho PAR1 S169 antibodies stain spindle pole and centrosome in immunofluorescence experiments [293]. In C. elegans the first cell division is unequal which produces a larger anterior cell and a smaller posterior one. The next cell division is asynchronous with the anterior cell dividing prior to the posterior one [294]. Rapid anterior cell cycle timing is due to enrichment of Polo kinase and Cdc25.1 in the anterior cell. This is dependent on a network of polarity proteins, including Par1 [295].

Advertisement

8. Cdc25 and disease

Cdc25 orthologues are the subject of much attention as they are commonly upregulated in human tumors [296]. This is perhaps not surprising considering the role of Cdc25 inhibition in maintaining genomic stability and the regulation of these phosphatases by Rb, p53 and a number of other oncogenes. Cdc25A and Cdc25B themselves are oncogenes in humans [212]. Cdc25B is overexpressed in 32% of breast cancer tissue samples, and high Cdc25B levels correlate with high incidences of recurrence and decreased 10 year survival [212]. Overexpression of Cdc25A is similarly linked to poor clinical outcome [296,297]. Cells bearing oncogenic mutations of myc have elevated Cdc25A and Cdc25B levels [298]. Anti-Cdc25B autoantibody has been shown to be a predictor of poor prognosis in esophageal cancer patients [299]. Overexpression of Cdc25B has recently been shown to cause a variety of S-phase effects including increased Cdc45 recruitment to chromatin, impairment of replication fork progression DNA damage and chromosome instability [300].

An interesting link between Cdc25 and disease comes from the finding that the HIV-1 protein vpr causes G2/M arrest [301]. When expressed in S. pombe, vpr activates Srk1 kinase, resulting in Cdc25 phosphorylation and 14-3-3 mediated nuclear export [302,303]. The vpr protein also acts through upregulation of PP2A phosphatase acting on both Wee1 kinase and Cdc25, reversing activating Cdc2 phosphorylation [304].

Advertisement

9. Conclusion

It has been more than thirty five years since Cdc25 was first isolated as an elongated temperature-sensitive fission yeast mutant and twenty one years since its biochemical function was determined. The field of cell cycle research and the study of Cdc25 in particular are extremely active with numerous new manuscripts appearing each year. This research has revealed that Cdc25 is one of the most intricately regulated proteins in the cell. Cdc25 accepts input from numerous pathways and checkpoints monitoring whether conditions inside and outside the cell are permissive for cell cycle progression. When conditions warrant caution, Cdc25 is inhibited by phosphorylation leading to alterations in its catalytic activity, cellular localization, substrate recognition and stability. When the green light is given Cdc25 participates in an intricate series of interconnected positive feedback loops with the beating heart of cell cycle regulation, the Cyclin-CDK complex. When the cell loses control of Cdc25 regulation, the results are deadly.

Advertisement

Acknowledgement

Thank you to Silja Freitag for her critical reading of this manuscript. This work was supported by a grant from the Natural Sciences and Engineering Research Council of Canada to PGY.

References

  1. 1. HartwellL. H.CulottiJ.ReidB.1970Genetic control of the cell-division cycle in yeast. I. Detection of mutants. Proc Natl Acad Sci U S A 662352359
  2. 2. HartwellL. H.MortimerR. K.CulottiJ.CulottiM.1973Genetic control of the cell division cycle in yeast: V. Genetic analysis of cdc mutants. Genetics 742267286
  3. 3. NurseP.ThuriauxP.NasmythK.1976Genetic control of the cell division cycle in the fission yeast Schizosaccharomyces pombe. Mol Gen Genet 1462167178
  4. 4. NurseP.1975Genetic control of cell size at cell division in yeast. Nature 2565518547551
  5. 5. FantesP.1979Epistatic gene interactions in the control of division in fission yeast. Nature 2795712428430
  6. 6. Fantes PA.1981Isolation of cell size mutants of a fission yeast by a new selective method: characterization of mutants and implications for division control mechanisms. J Bacteriol 1462746754
  7. 7. ThuriauxP.NurseP.CarterB.1978Mutants altered in the control co-ordinating cell division with cell growth in the fission yeast Schizosaccharomyces pombe. Mol Gen Genet 1612215220
  8. 8. KishimotoT.KanataniH.1976Cytoplasmic factor responsible for germinal vesicle breakdown and meiotic maturation in starfish oocyte. Nature 2605549321322
  9. 9. MasuiY.MarkertC. L.1971Cytoplasmic control of nuclear behavior during meiotic maturation of frog oocytes. J Exp Zool 1772129145
  10. 10. Maller JL.1990Xenopus oocytes and the biochemistry of cell division. Biochemistry 291331573166
  11. 11. Smith LD, Ecker RE.1971The interaction of steroids with Rana pipiens Oocytes in the induction of maturation. Dev Biol 252232247
  12. 12. Murray AW, Solomon MJ, Kirschner MW.1989The role of cyclin synthesis and degradation in the control of maturation promoting factor activity. Nature 3396222280286
  13. 13. NewportJ.KirschnerM.1982A major developmental transition in early Xenopus embryos: II. Control of the onset of transcription. Cell 303687696
  14. 14. NewportJ.KirschnerM.1982A major developmental transition in early Xenopus embryos: I. characterization and timing of cellular changes at the midblastula stage. Cell 303675686
  15. 15. DunphyW. G.BrizuelaL.BeachD.NewportJ.1988The Xenopus cdc2 protein is a component of MPF, a cytoplasmic regulator of mitosis. Cell 543423431
  16. 16. EvansT.RosenthalE. T.YoungblomJ.DistelD.HuntT.1983Cyclin: a protein specified by maternal mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell 332389396
  17. 17. GautierJ.MinshullJ.LohkaM.GlotzerM.HuntT.MallerJ. L.1990Cyclin is a component of maturation-promoting factor from Xenopus. Cell 603487494
  18. 18. PatraD.DunphyW. G.1996Xe-9a Xenopus Suc1/Cks homolog, has multiple essential roles in cell cycle control. Genes Dev 10(12):1503-1515.
  19. 19. GautierJ.NorburyC.LohkaM.NurseP.MallerJ.1988Purified maturation-promoting factor contains the product of a Xenopus homolog of the fission yeast cell cycle control gene cdc2+. Cell 543433439
  20. 20. NurseP.BissettY.1981Gene required in G1 for commitment to cell cycle and in G2 for control of mitosis in fission yeast. Nature 2925823558560
  21. 21. BeachD.DurkaczB.NurseP.1982Functionally homologous cell cycle control genes in budding and fission yeast. Nature 3005894706709
  22. 22. HaganI.HaylesJ.NurseP.1988Cloning and sequencing of the cyclin-related cdc13+ gene and a cytological study of its role in fission yeast mitosis. J Cell Sci 91 ( Pt 4)(Pt 4):587-595.
  23. 23. BooherR.BeachD.1988Involvement of cdc13+ in mitotic control in Schizosaccharomyces pombe: possible interaction of the gene product with microtubules. EMBO J 7823212327
  24. 24. GautierJ.MJSolomonBooher.RNBazanJ. F.KirschnerM. W.1991Cdc25 is a Specific Tyrosine Phosphatase that Directly Activates 34cdc2Cell 67(1):197-211.
  25. 25. MillarJ. B.Mc GowanC. H.LenaersG.JonesR.RussellP.199180cdc25mitotic inducer is the tyrosine phosphatase that activates p34cdc2 kinase in fission yeast. EMBO J 10(13):4301-4309.
  26. 26. DoreeM.PeaucellierG.PicardA.1983Activity of the maturation-promoting factor and the extent of protein phosphorylation oscillate simultaneously during meiotic maturation of starfish oocytes. Dev Biol 992489501
  27. 27. MallerJ.WuM.GerhartJ. C.1977Changes in protein phosphorylation accompanying maturation of Xenopus laevis oocytes. Dev Biol 582295312
  28. 28. MazzeiG.GuerrierP.1982Changes in the pattern of protein phosphorylation during meiosis reinitiation in starfish oocytes. Dev Biol 912246256
  29. 29. WibletM.1974Protein kinase activities during maturation in Xenopus laevis oocytes. Biochem Biophys Res Commun 603991998
  30. 30. MargalitA.VlcekS.GruenbaumY.FoisnerR.2005Breaking and making of the nuclear envelope. J Cell Biochem 953454465
  31. 31. AbeS.NagasakaK.HirayamaY.Kozuka-HataH.OyamaM.AoyagiY.et al.2011The initial phase of chromosome condensation requires Cdk1-mediated phosphorylation of the CAP-D3 subunit of condensin II. Genes Dev 258863874
  32. 32. GlotzerM.2009The 3Ms of central spindle assembly: microtubules, motors and MAPs. Nat Rev Mol Cell Biol 101920
  33. 33. HoltL. J.TuchB. B.VillenJ.JohnsonA. D.GygiS. P.DOMorgan2009Global analysis of Cdk1 substrate phosphorylation sites provides insights into evolution. Science 325594816821686
  34. 34. Enserink JM, Kolodner RD.2010An overview of Cdk1controlled targets and processes. Cell Div 5:11.
  35. 35. RussellP.MorenoS.ReedS. I.1989Conservation of mitotic controls in fission and budding yeasts. Cell 572295303
  36. 36. NagataA.IgarashiM.JinnoS.SutoK.OkayamaH.1991An additional homolog of the fission yeast cdc25+ gene occurs in humans and is highly expressed in some cancer cells. New Biol 310959968
  37. 37. GalaktionovK.BeachD.1991Specific activation of cdc25 tyrosine phosphatases by B-type cyclins: evidence for multiple roles of mitotic cyclins. Cell 67611811194
  38. 38. SadhuK.ReedS. I.RichardsonH.RussellP.1990Human homolog of fission yeast cdc25 mitotic inducer is predominantly expressed in G2. Proc Natl Acad Sci U S A 871351395143
  39. 39. WickramasingheD.BeckerS.ErnstM. K.ResnickJ. L.CentanniJ. M.TessarolloL.et al.1995Two CDC25 homologues are differentially expressed during mouse development. Development 121720472056
  40. 40. OkazakiK.HayashidaK.IwashitaJ.HaranoM.FurunoN.SagataN.1996Isolation of a cDNA encoding the X enopus homologue of mammalian Cdc25A that can induce meiotic maturation of oocytes. Gene 178(1-2):111-114.
  41. 41. Forrest AR, McCormack AK, DeSouza CP, Sinnamon JM, Tonks ID, Hayward NK, et al.1999Multiple splicing variants of cdc25B regulate G2/M progression. Biochem Biophys Res Commun 2602510515
  42. 42. BonnetJ.MayonoveP.MorrisM. C.2008Differential phosphorylation of Cdc25C phosphatase in mitosis. Biochem Biophys Res Commun 3703483488
  43. 43. WegenerS.HampeW.HerrmannD.SchallerH. C.2000Alternative splicing in the regulatory region of the human phosphatases CDC25A and CDC25C. Eur J Cell Biol 7911810815
  44. 44. JullienD.BuglerB.DozierC.CazalesM.DucommunB.2011Identification of N-terminally truncated stable nuclear isoforms of CDC25B that are specifically involved in G2/M checkpoint recovery. Cancer Res 71519681977
  45. 45. KumagaiA.DunphyW. G.1992Regulation of the cdc25 protein during the cell cycle in Xenopus extracts. Cell 70139151
  46. 46. RimeH.HuchonD.De SmedtV.ThibierC.GalaktionovK.JessusC.et al.1994Microinjection of Cdc25 protein phosphatase into Xenopus prophase oocyte activates MPF and arrests meiosis at metaphase I. Biol Cell 8211122
  47. 47. AshcroftN. R.MEKosinskiWickramasinghe. D.DonovanP. J.GoldenA.1998The four cdc25 genes from the nematode Caenorhabditis elegans. Gene 214(1-2):59-66.
  48. 48. AlpheyL.JimenezJ.White-CooperH.DawsonI.NurseP.GloverD. M.1992twine, a cdc25 homolog that functions in the male and female germline of Drosophila. Cell 696977988
  49. 49. Edgar BA, O’Farrell PH.1989Genetic control of cell division patterns in the Drosophila embryo. Cell 571177187
  50. 50. KumagaiA.DunphyW. G.1991The cdc25 protein controls tyrosine dephosphorylation of the cdc2 protein in a cell-free system. Cell 645903914
  51. 51. KhadarooB.RobbensS.FerrazC.DerelleE.EychenieS.CookeR.et al.2004The first green lineage cdc25 dual-specificity phosphatase. Cell Cycle 34513518
  52. 52. LandrieuI.daCosta. M.De VeylderL.DewitteF.VandepoeleK.HassanS.et al.2004A small CDC25 dual-specificity tyrosine-phosphatase isoform in Arabidopsis thaliana. Proc Natl Acad Sci U S A 101361338013385
  53. 53. SorrellD. A.ChrimesD.DickinsonJ. R.RogersH. J.FrancisD.2005The Arabidopsis CDC25 induces a short cell length when overexpressed in fission yeast: evidence for cell cycle function. New Phytol 1652425428
  54. 54. Booher RN, Alfa CE, Hyams JS, Beach DH.1989The fission yeast cdc2/cdc13/suc1 protein kinase: regulation of catalytic activity and nuclear localization. Cell 583485497
  55. 55. BooherR.BeachD.1987Interaction between cdc13+ and cdc2+ in the control of mitosis in fission yeast; dissociation of the G1 and G2 roles of the cdc2+ protein kinase. EMBO J 61134413447
  56. 56. Martin-CastellanosC.MABlanco dePrada. J. M.MorenoS.2000The puc1 cyclin regulates the G1 phase of the fission yeast cell cycle in response to cell size. Mol Biol Cell 112543554
  57. 57. DurkaczB.CarrA.NurseP.1986Transcription of the cdc2 cell cycle control gene of the fission yeast Schizosaccharomyces pombe. EMBO J 52369373
  58. 58. CreanorJ.MitchisonJ. M.1996The kinetics of the B cyclin 56cdc13and the phosphatase p80cdc25 during the cell cycle of the fission yeast Schizosaccharomyces pombe. J Cell Sci 109 ( Pt 6)(Pt 6):1647-1653.
  59. 59. DucommunB.DraettaG.YoungP.BeachD.1990Fission yeast cdc25 is a cell-cycle regulated protein. Biochem Biophys Res Commun 1671301309
  60. 60. FrazerC.YoungP. G.2011Redundant mechanisms prevent mitotic entry following replication arrest in the absence of Cdc25 hyper-phosphorylation in fission yeast. PLoS One 6(6):e21348.
  61. 61. DenHaese. G. J.WalworthN.CarrA. M.GouldK. L.1995The Wee1 protein kinase regulates T14 phosphorylation of fission yeast Cdc2. Mol Biol Cell 64371385
  62. 62. GouldK. L.NurseP.1989Tyrosine phosphorylation of the fission yeast cdc2+ protein kinase regulates entry into mitosis. Nature 34262453945
  63. 63. LundgrenK.WalworthN.BooherR.DembskiM.KirschnerM.BeachD.1991Mik1 and Wee1 Cooperate in the Inhibitory Tyrosine Phosphorylation of Cdc2. Cell 64611111122
  64. 64. MSLeeEnoch. T.Piwnica-WormsH.1994Mik1+ Encodes a Tyrosine Kinase that Phosphorylates 34cdc2on Tyrosine 15. J Biol Chem 269(48):30530-30537.
  65. 65. RussellP.NurseP.1987Negative regulation of mitosis by wee1+, a gene encoding a protein kinase homolog. Cell 494559567
  66. 66. ParkerL. L.Atherton-FesslerS.Piwnica-WormsH.1992107wee1is a Dual-Specificity Kinase that Phosphorylates P34cdc2 on Tyrosine 15. Proc Natl Acad Sci U S A 89(7):2917-2921.
  67. 67. FeatherstoneC.RussellP.1991Fission yeast 107wee1mitotic inhibitor is a tyrosine/serine kinase. Nature 349:808-811.
  68. 68. ChuaG.LingnerC.FrazerC.YoungP. G.2002The sal3(+) gene encodes an importin-beta implicated in the nuclear import of Cdc25 in Schizosaccharomyces pombe. Genetics 1622689703
  69. 69. RupesI.BAWebbMak. A.YoungP. G.2001G2/M arrest caused by actin disruption is a manifestation of the cell size checkpoint in fission yeast. Mol Biol Cell 121238923903
  70. 70. KovelmanR.RussellP.1996Stockpiling of Cdc25 during a DNA replication checkpoint arrest in Schizosaccharomyces pombe. Mol Cell Biol 1618693
  71. 71. MorenoS.NurseP.1990Substrates for 34cdc2in vivo veritas? Cell 61(4):549-551.
  72. 72. ShenoyS.ChoiJ. K.BagrodiaS.CopelandT. D.MallerJ. L.ShallowayD.1989Purified maturation promoting factor phosphorylates 60cat the sites phosphorylated during fibroblast mitosis. Cell 57(5):763-774.
  73. 73. Wolfe BA, Gould KL.2004Fission yeast Clp1p phosphatase affects G2/M transition and mitotic exit through Cdc25p inactivation. EMBO J 234919929
  74. 74. MulvihillD. P.PetersenJ.OhkuraH.GloverD. M.HaganI. M.1999Plo1 kinase recruitment to the spindle pole body and its role in cell division in Schizosaccharomyces pombe. Mol Biol Cell 10827712785
  75. 75. CharlesJ. F.JaspersenS. L.Tinker-KulbergR. L.HwangL.SzidonA.DOMorgan1998The Polo-related kinase Cdc5 activates and is destroyed by the mitotic cyclin destruction machinery in S. cerevisiae. Curr Biol 89497507
  76. 76. KatsunoY.SuzukiA.SugimuraK.OkumuraK.ZineldeenD. H.ShimadaM.et al.2009Cyclin A-Cdk1 regulates the origin firing program in mammalian cells. Proc Natl Acad Sci U S A 106931843189
  77. 77. Elledge SJ, Spottswood MR.1991A new human 34protein kinase, CDK2, identified by complementation of a cdc28 mutation in Saccharomyces cerevisiae, is a homolog of Xenopus Eg1. EMBO J 10(9):2653-2659.
  78. 78. GuY.RosenblattJ.DOMorgan1992Cell cycle regulation of CDK2 activity by phosphorylation of Thr160 and Tyr15. EMBO J 111139954005
  79. 79. ElledgeS. J.RichmanR.HallF. L.WilliamsR. T.LodgsonN.HarperJ. W.1992CDK2 encodes a 33-kDa cyclin A-associated protein kinase and is expressed before CDC2 in the cell cycle. Proc Natl Acad Sci U S A 89729072911
  80. 80. RosenblattJ.GuY.DOMorgan1992Human cyclin-dependent kinase 2 is activated during the S and G2 phases of the cell cycle and associates with cyclin A. Proc Natl Acad Sci U S A 89728242828
  81. 81. Guadagno TM, Newport JW.1996Cdk2 kinase is required for entry into mitosis as a positive regulator of Cdc2-cyclin B kinase activity. Cell 8417382
  82. 82. BatesS.BonettaL.MacAllan. D.ParryD.HolderA.DicksonC.et al.1994CDK6 (PLSTIRE) and CDK4 (PSK-J3) are a distinct subset of the cyclin-dependent kinases that associate with cyclin D1. Oncogene 917179
  83. 83. Fisher RP, Morgan DO.1994A novel cyclin associates with MO15/CDK7 to form the CDK-activating kinase. Cell 784713724
  84. 84. Weinberg RA.1995The retinoblastoma protein and cell cycle control. Cell 813323330
  85. 85. DysonN.1998The regulation of E2F by pRB-family proteins. Genes Dev 121522452262
  86. 86. VidalA.KoffA.2000Cell-cycle inhibitors: three families united by a common cause. Gene 247(1-2):1-15.
  87. 87. ChenX.PrywesR.1999Serum-induced expression of the cdc25A gene by relief of E2F-mediated repression. Mol Cell Biol 19746954702
  88. 88. MalumbresM.PellicerA.1998RAS pathways to cell cycle control and cell transformation. Front Biosci 3:d887912
  89. 89. Sherr CJ, Roberts JM.1999CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 131215011512
  90. 90. Lundberg AS, Weinberg RA.1998Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol Cell Biol 182753761
  91. 91. BlombergI.HoffmannI.1999Ectopic expression of Cdc25A accelerates the G(1)/S transition and leads to premature activation of cyclin E- and cyclin A-dependent kinases. Mol Cell Biol 19961836194
  92. 92. Sherr CJ, Roberts JM.2004Living with or without cyclins and cyclin-dependent kinases. Genes Dev 182226992711
  93. 93. RNBooherHolman. P. S.FattaeyA.1997Human Myt1 is a cell cycle-regulated kinase that inhibits Cdc2 but not Cdk2 activity. J Biol Chem 272352230022306
  94. 94. SebastianB.KakizukaA.HunterT.1993Cdc25M2 activation of cyclin-dependent kinases by dephosphorylation of threonine-14 and tyrosine-15. Proc Natl Acad Sci U S A 90835213524
  95. 95. HoffmannI.DraettaG.KarsentiE.1994Activation of the phosphatase activity of human cdc25A by a cdk2-cyclin E dependent phosphorylation at the G1/S transition. EMBO J 131843024310
  96. 96. Gabrielli BG, Clark JM, McCormack AK, Ellem KA.1997Hyperphosphorylation of the N-terminal domain of Cdc25 regulates activity toward cyclin B1/Cdc2 but not cyclin A/Cdk2. J Biol Chem 272452860728614
  97. 97. LammerC.WagererS.SaffrichR.MertensD.AnsorgeW.HoffmannI.1998The cdc25B phosphatase is essential for the G2/M phase transition in human cells. J Cell Sci 111 ( Pt 16)(Pt 16):2445-2453.
  98. 98. SexlV.DiehlJ. A.SherrC. J.AshmunR.BeachD.RousselM. F.1999A rate limiting function of cdc25A for S phase entry inversely correlates with tyrosine dephosphorylation of Cdk2. Oncogene 183573582
  99. 99. Ducruet AP, Lazo JS.2003Regulation of Cdc25A half-life in interphase by cyclin-dependent kinase 2 activity. J Biol Chem 278343183831842
  100. 100. PaganoM.PepperkokR.VerdeF.AnsorgeW.DraettaG.1992Cyclin A is required at two points in the human cell cycle. EMBO J 113961971
  101. 101. PetersenB. O.LukasJ.SorensenC. S.BartekJ.HelinK.1999Phosphorylation of mammalian CDC6 by cyclin A/CDK2 regulates its subcellular localization. EMBO J 182396410
  102. 102. CoverleyD.PelizonC.TrewickS.LaskeyR. A.2000Chromatin-bound Cdc6 persists in S and G2 phases in human cells, while soluble Cdc6 is destroyed in a cyclin A-cdk2 dependent process. J Cell Sci 113 ( Pt 11)(Pt 11):1929-1938.
  103. 103. Alexandrow MG, Hamlin JL.2005Chromatin decondensation in S-phase involves recruitment of Cdk2 by Cdc45 and histone H1 phosphorylation. J Cell Biol 1686875886
  104. 104. Garner-HamrickP. A.FisherC.1998Antisense phosphorothioate oligonucleotides specifically down-regulate cdc25B causing S-phase delay and persistent antiproliferative effects. Int J Cancer 765720728
  105. 105. LindqvistA.KallstromH.LundgrenA.BarsoumE.RosenthalC. K.2005Cdc25B cooperates with Cdc25A to induce mitosis but has a unique role in activating cyclin B1-Cdk1 at the centrosome. J Cell Biol 17113545
  106. 106. MSChenHurov. J.WhiteL. S.Woodford-ThomasT.Piwnica-WormsH.2001Absence of apparent phenotype in mice lacking Cdc25C protein phosphatase. Mol Cell Biol 211238533861
  107. 107. LincolnA. J.WickramasingheD.SteinP.SchultzR. M.MEPalko DeMiguel. M. P.et al.2002Cdc25b phosphatase is required for resumption of meiosis during oocyte maturation. Nat Genet 304446449
  108. 108. FergusonA. M.WhiteL. S.DonovanP. J.Piwnica-WormsH.2005Normal cell cycle and checkpoint responses in mice and cells lacking Cdc25B and Cdc25C protein phosphatases. Mol Cell Biol 25728532860
  109. 109. FurunoN.denElzen. N.PinesJ.1999Human cyclin A is required for mitosis until mid prophase. J Cell Biol 147295306
  110. 110. MitraJ.EndersG. H.2004Cyclin A/Cdk2 complexes regulate activation of Cdk1 and Cdc25 phosphatases in human cells. Oncogene 231933613367
  111. 111. MinshullJ.GolsteynR.HillC. S.HuntT.1990The A- and B-type cyclin associated cdc2 kinases in Xenopus turn on and off at different times in the cell cycle. EMBO J 9928652875
  112. 112. CansC.DucommunB.BaldinV.1999Proteasome-dependent degradation of human CDC25B phosphatase. Mol Biol Rep 26(1-2):53-57.
  113. 113. BaldinV.CansC.KnibiehlerM.DucommunB.1997Phosphorylation of human CDC25B phosphatase by CDK1-cyclin A triggers its proteasome-dependent degradation. J Biol Chem 272523273132734
  114. 114. GabrielliB. G.MSLeeWalker. D. H.Piwnica-WormsH.MallerJ. L.1992Cdc25 regulates the phosphorylation and activity of the Xenopus cdk2 protein kinase complex. J Biol Chem 267251804018046
  115. 115. MalumbresM.BarbacidM.2005Mammalian cyclin-dependent kinases. Trends Biochem Sci 3011630641
  116. 116. De BoerL.OakesV.BeamishH.GilesN.StevensF.Somodevilla-TorresM.et al.2008Cyclin A/cdk2 coordinates centrosomal and nuclear mitotic events. Oncogene 273142614268
  117. 117. NishijimaH.NishitaniH.SekiT.NishimotoT.1997A dual-specificity phosphatase Cdc25B is an unstable protein and triggers 34cdc2)/cyclin B activation in hamster BHK21 cells arrested with hydroxyurea. J Cell Biol 138(5):1105-1116.
  118. 118. TimofeevO.CizmeciogluO.SetteleF.KempfT.HoffmannI.2010Cdc25 phosphatases are required for timely assembly of CDK1-cyclin B at the G2/M transition. J Biol Chem 285221697816990
  119. 119. BaillyE.DoreeM.NurseP.BornensM.198934cdc2is located in both nucleus and cytoplasm; part is centrosomally associated at G2/M and enters vesicles at anaphase. EMBO J 8(13):3985-3995.
  120. 120. Gabrielli BG, De Souza CP, Tonks ID, Clark JM, Hayward NK, Ellem KA.1996Cytoplasmic accumulation of cdc25B phosphatase in mitosis triggers centrosomal microtubule nucleation in HeLa cells. J Cell Sci 109 ( Pt 5)(Pt 5):1081-1093.
  121. 121. PinesJ.HunterT.1991Human cyclins A and B1 are differentially located in the cell and undergo cell cycle-dependent nuclear transport. J Cell Biol 1151117
  122. 122. HirotaT.KunitokuN.SasayamaT.MarumotoT.ZhangD.NittaM.et al.2003Aurora-A and an interacting activator, the LIM protein Ajuba, are required for mitotic commitment in human cells. Cell 1145585598
  123. 123. BarrA. R.GergelyF.2007Aurora-A: the maker and breaker of spindle poles. J Cell Sci 120(Pt 17):2987-2996.
  124. 124. DutertreS.CazalesM.QuarantaM.FromentC.TrabutV.DozierC.et al.2004Phosphorylation of CDC25B by Aurora-A at the centrosome contributes to the G2M transition. J Cell Sci 117(Pt 12):2523-2531.
  125. 125. JackmanM.LindonC.NiggE. A.PinesJ.2003Active cyclin B1-Cdk1 first appears on centrosomes in prophase. Nat Cell Biol 52143148
  126. 126. PinesJ.HunterT.1994The differential localization of human cyclins A and B is due to a cytoplasmic retention signal in cyclin B. EMBO J 131637723781
  127. 127. HagtingA.KarlssonC.CluteP.JackmanM.PinesJ.1998MPF localization is controlled by nuclear export. EMBO J 171441274138
  128. 128. ToyoshimaF.MoriguchiT.WadaA.FukudaM.NishidaE.1998Nuclear export of cyclin B1 and its possible role in the DNA damage-induced G2 checkpoint. EMBO J 171027282735
  129. 129. YangJ.BardesE. S.JDMooreBrennan. J.MAPowersKornbluth. S.1998Control of cyclin B1 localization through regulated binding of the nuclear export factor CRM1. Genes Dev 121421312143
  130. 130. BorgneA.OstvoldA. C.FlamentS.MeijerL.1999Intra-M phase-promoting factor phosphorylation of cyclin B at the prophase/metaphase transition. J Biol Chem 274171197711986
  131. 131. Toyoshima-MorimotoF.TaniguchiE.ShinyaN.IwamatsuA.NishidaE.2001Polo-like kinase 1 phosphorylates cyclin B1 and targets it to the nucleus during prophase. Nature 4106825215220
  132. 132. Okano-UchidaT.OkumuraE.IwashitaM.YoshidaH.TachibanaK.KishimotoT.2003Distinct regulators for Plk1 activation in starfish meiotic and early embryonic cycles. EMBO J 222056335642
  133. 133. NakajimaH.Toyoshima-MorimotoF.TaniguchiE.NishidaE.2003Identification of a consensus motif for Plk (Polo-like kinase) phosphorylation reveals Myt1 as a Plk1 substrate. J Biol Chem 278282527725280
  134. 134. WatanabeN.AraiH.NishiharaY.TaniguchiM.WatanabeN.HunterT.et al.2004M-phase kinases induce phospho-dependent ubiquitination of somatic Wee1 by SCFbeta-TrCP. Proc Natl Acad Sci U S A 1011344194424
  135. 135. BaldinV.PelpelK.CazalesM.CansC.DucommunB.2002Nuclear localization of CDC25B1 and serine 146 integrity are required for induction of mitosis. J Biol Chem 277383517635182
  136. 136. LobjoisV.JullienD.BoucheJ. P.DucommunB.2009The polo-like kinase 1 regulates CDC25B-dependent mitosis entry. Biochim Biophys Acta 17933462468
  137. 137. LobjoisV.FromentC.BraudE.GrimalF.Burlet-SchiltzO.DucommunB.et al.2011Study of the docking-dependent PLK1 phosphorylation of the CDC25B phosphatase. Biochem Biophys Res Commun 41018790
  138. 138. QianY. W.EriksonE.TaiebF. E.MallerJ. L.2001The polo-like kinase Plx1 is required for activation of the phosphatase Cdc25C and cyclin B-Cdc2 in Xenopus oocytes. Mol Biol Cell 12617911799
  139. 139. RoshakA. K.CapperE. A.ImburgiaC.FornwaldJ.ScottG.MarshallL. A.2000The human polo-like kinase, PLK, regulates cdc2/cyclin B through phosphorylation and activation of the cdc25C phosphatase. Cell Signal 126405411
  140. 140. Toyoshima-MorimotoF.TaniguchiE.NishidaE.2002Plk1 promotes nuclear translocation of human Cdc25C during prophase. EMBO Rep 34341348
  141. 141. Bahassi elM.HenniganR. F.MyerD. L.StambrookP. J.2004Cdc25C phosphorylation on serine 191 by Plk3 promotes its nuclear translocation. Oncogene 231526582663
  142. 142. BonniS.GanuelasM. L.PetrinacS.HudsonJ. W.2008Human Plk4 phosphorylates Cdc25C. Cell Cycle 74545547
  143. 143. StrausfeldU.FernandezA.CaponyJ. P.GirardF.LautredouN.DerancourtJ.et al.1994Activation of 34cdc2protein kinase by microinjection of human cdc25C into mammalian cells. Requirement for prior phosphorylation of cdc25C by p34cdc2 on sites phosphorylated at mitosis. J Biol Chem 269(8):5989-6000.
  144. 144. HondaR.OhbaY.NagataA.OkayamaH.YasudaH.1993Dephosphorylation of human 34cdc2kinase on both Thr-14 and Tyr-15 by human cdc25B phosphatase. FEBS Lett 318(3):331-334.
  145. 145. FranckhauserC.MamaevaD.Heron-MilhavetL.FernandezA.LambN. J.2010Distinct pools of cdc25C are phosphorylated on specific TP sites and differentially localized in human mitotic cells. PLoS One 5(7):e11798.
  146. 146. IzumiT.MallerJ. L.1993Elimination of cdc2 phosphorylation sites in the cdc25 phosphatase blocks initiation of M-phase. Mol Biol Cell 41213371350
  147. 147. PinesJ.HunterT.CyclinsA.inB.theHuman.CellCycle.In: Marsh J, editor. Regulation of the Eukaryotic Cell New York: Wiley; 1992187208
  148. 148. MailandN.PodtelejnikovA. V.GrothA.MannM.BartekJ.LukasJ.2002Regulation of G(2)/M events by Cdc25A through phosphorylation-dependent modulation of its stability. EMBO J 212159115920
  149. 149. TumurbaatarI.CizmeciogluO.HoffmannI.GrummtI.VoitR.2011Human Cdc14B promotes progression through mitosis by dephosphorylating Cdc25 and regulating Cdk1/cyclin B activity. PLoS One 6(2):e14711.
  150. 150. BarfordD.2011Structural insights into anaphase-promoting complex function and mechanism. Philos Trans R Soc Lond B Biol Sci 366158436053624
  151. 151. EstebanV.MDVazquez-NovelleCalvo. E.BuenoA.SacristanM. P.2006Human Cdc14A reverses CDK1 phosphorylation of Cdc25A on serines 115 and 320. Cell Cycle 52428942898
  152. 152. DonzelliM.SquatritoM.GanothD.HershkoA.PaganoM.DraettaG. F.2002Dual mode of degradation of Cdc25 A phosphatase. EMBO J 211848754884
  153. 153. KiefferI.LorenzoC.DozierC.SchmittE.DucommunB.2007Differential mitotic degradation of the CDC25B phosphatase variants. Oncogene 265778477858
  154. 154. RhindN.FurnariB.RussellP.1997Cdc2 tyrosine phosphorylation is required for the DNA damage checkpoint in fission yeast. Genes Dev 114504511
  155. 155. BoddyM. N.FurnariB.MondesertO.RussellP.1998Replication checkpoint enforced by kinases Cds1 and Chk1. Science 2805365909912
  156. 156. ZengY.ForbesK. C.WuZ.MorenoS.Piwnica-WormsH.EnochT.1998Replication checkpoint requires phosphorylation of the phosphatase Cdc25 by Cds1 or Chk1. Nature 3956701507510
  157. 157. FurnariB.BlasinaA.BoddyM. N.Mc GowanC. H.RussellP.1999Cdc25 inhibited in vivo and in vitro by checkpoint kinases Cds1 and Chk1. Mol Biol Cell 104833845
  158. 158. BranzeiD.FoianiM.2010Maintaining genome stability at the replication fork. Nat Rev Mol Cell Biol 113208219
  159. 159. MeloJ.ToczyskiD.2002A unified view of the DNA-damage checkpoint. Curr Opin Cell Biol 142237245
  160. 160. Lopez-GironaA.FurnariB.MondesertO.RussellP.1999Nuclear localization of Cdc25 is regulated by DNA damage and a 14-3-3 protein. Nature 3976715172175
  161. 161. FordJ. C.al-KhodairyF.FotouE.SheldrickK. S.GriffithsD. J.CarrA. M.1994protein homologs required for the DNA damage checkpoint in fission yeast. Science 2655171533535
  162. 162. ZengY.Piwnica-WormsH.1999DNA damage and replication checkpoints in fission yeast require nuclear exclusion of the Cdc25 phosphatase via 14-3-3 binding. Mol Cell Biol 191174107419
  163. 163. DaiY.GrantS.2010New insights into checkpoint kinase 1 in the DNA damage response signaling network. Clin Cancer Res 162376383
  164. 164. NiidaH.NakanishiM.2006DNA damage checkpoints in mammals. Mutagenesis 21139
  165. 165. MinellaA. C.GrimJ. E.WelckerM.ClurmanB. E.200753and SCFFbw7 cooperatively restrain cyclin E-associated genome instability. Oncogene 26(48):6948-6953.
  166. 166. TeradaY.TatsukaM.JinnoS.OkayamaH.1995Requirement for tyrosine phosphorylation of Cdk4 in G1 arrest induced by ultraviolet irradiation. Nature 3766538358362
  167. 167. MailandN.FalckJ.LukasC.SyljuasenR. G.WelckerM.BartekJ.et al.2000Rapid destruction of human Cdc25A in response to DNA damage. Science 288547014251429
  168. 168. GoloudinaA.YamaguchiH.ChervyakovaD. B.AppellaE.FornaceA. J.Jr BulavinD. V.2003Regulation of human Cdc25A stability by Serine 75 phosphorylation is not sufficient to activate a S phase checkpoint. Cell Cycle 25473478
  169. 169. HassepassI.VoitR.HoffmannI.2003Phosphorylation at serine 75 is required for UV-mediated degradation of human Cdc25A phosphatase at the S-phase checkpoint. J Biol Chem 278322982429829
  170. 170. MSChenRyan.CEPiwnica-WormsH.2003Chk1 kinase negatively regulates mitotic function of Cdc25A phosphatase through 14-3-3 binding. Mol Cell Biol 232174887497
  171. 171. JinJ.ShiroganeT.XuL.NalepaG.QinJ.ElledgeS. J.et al.2003SCFbeta-TRCP links Chk1 signaling to degradation of the Cdc25A protein phosphatase. Genes Dev 172430623074
  172. 172. ZhaoH.WatkinsJ. L.Piwnica-WormsH.2002Disruption of the checkpoint kinase 1/cell division cycle 25A pathway abrogates ionizing radiation-induced S and G2 checkpoints. Proc Natl Acad Sci U S A 99231479514800
  173. 173. FalckJ.MailandN.SyljuasenR. G.BartekJ.LukasJ.2001The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 4106830842847
  174. 174. ShreeramS.HeeW. K.BulavinD. V.2008Cdc25A serine 123 phosphorylation couples centrosome duplication with DNA replication and regulates tumorigenesis. Mol Cell Biol 282474427450
  175. 175. MadlenerS.RosnerM.KriegerS.GiessriglB.GridlingM.VoT. P.et al.2009Short 42 degrees C heat shock induces phosphorylation and degradation of Cdc25A which depends on 38MAPKChk2 and 14.3.3. Hum Mol Genet 18(11):1990-2000.
  176. 176. FalckJ.PetriniJ. H.WilliamsB. R.LukasJ.BartekJ.2002The DNA damage-dependent intra-S phase checkpoint is regulated by parallel pathways. Nat Genet 303290294
  177. 177. YaffeM. B.RittingerK.VoliniaS.CaronP. R.AitkenA.LeffersH.et al.1997The structural basis for 14-3-3:phosphopeptide binding specificity. Cell 917961971
  178. 178. KasaharaK.GotoH.EnomotoM.TomonoY.KiyonoT.InagakiM.2010gamma mediates Cdc25A proteolysis to block premature mitotic entry after DNA damage. EMBO J 291628022812
  179. 179. AitkenA.2002Functional specificity in 14-3-3 isoform interactions through dimer formation and phosphorylation. Chromosome location of mammalian isoforms and variants. Plant Mol Biol 5069931010
  180. 180. AitkenA.BaxterH.DuboisT.ClokieS.MackieS.MitchellK.et al.2002Specificity of 14-3-3 isoform dimer interactions and phosphorylation. Biochem Soc Trans 30351360
  181. 181. XiaoZ.ChenZ.GunasekeraA. H.SowinT. J.RosenbergS. H.FesikS.et al.2003Chk1 mediates S and G2 arrests through Cdc25A degradation in response to DNA-damaging agents. J Biol Chem 278242176721773
  182. 182. CostanzoV.RobertsonK.YingC. Y.KimE.AvvedimentoE.GottesmanM.et al.2000Reconstitution of an ATM-dependent checkpoint that inhibits chromosomal DNA replication following DNA damage. Mol Cell 63649659
  183. 183. MolinariM.MercurioC.DominguezJ.GoubinF.DraettaG. F.2000Human Cdc25 A inactivation in response to S phase inhibition and its role in preventing premature mitosis. EMBO Rep 117179
  184. 184. BusinoL.DonzelliM.ChiesaM.GuardavaccaroD.GanothD.DorrelloN. V.et al.2003Degradation of Cdc25A by beta-TrCP during S phase and in response to DNA damage. Nature 42669628791
  185. 185. WinstonJ. T.StrackP.Beer-RomeroP.ChuC. Y.ElledgeS. J.HarperJ. W.1999The SCFbeta-TRCP-ubiquitin ligase complex associates specifically with phosphorylated destruction motifs in IkappaBalpha and beta-catenin and stimulates IkappaBalpha ubiquitination in vitro. Genes Dev 133270283
  186. 186. DonzelliM.BusinoL.ChiesaM.GanothD.HershkoA.DraettaG. F.2004Hierarchical order of phosphorylation events commits Cdc25A to betaTrCP-dependent degradation. Cell Cycle 34469471
  187. 187. MelixetianM.KleinD. K.SorensenC. S.HelinK.2009NEK11 regulates CDC25A degradation and the IR-induced G2/M checkpoint. Nat Cell Biol 111012471253
  188. 188. ShiehS. Y.AhnJ.TamaiK.TayaY.PrivesC.2000The human homologs of checkpoint kinases Chk1 and Cds1 (Chk2) phosphorylate 53at multiple DNA damage-inducible sites. Genes Dev 14(3):289-300.
  189. 189. KhosraviR.MayaR.GottliebT.OrenM.ShilohY.ShkedyD.1999Rapid ATM-dependent phosphorylation of MDM2 precedes 53accumulation in response to DNA damage. Proc Natl Acad Sci U S A 96(26):14973-14977.
  190. 190. MayaR.BalassM.KimS. T.ShkedyD.LealJ. F.ShifmanO.et al.2001ATM-dependent phosphorylation of Mdm2 on serine 395: role in 53activation by DNA damage. Genes Dev 15(9):1067-1077.
  191. 191. TritarelliA.OricchioE.CiciarelloM.MangiacasaleR.PalenaA.LaviaP.et al.200453localization at centrosomes during mitosis and postmitotic checkpoint are ATM-dependent and require serine 15 phosphorylation. Mol Biol Cell 15:3751.
  192. 192. el -DeiryW. S.HarperJ. W.O’ConnorP. M.VelculescuV. E.CECanmanJackman. J.et al.1994WAF1/CIP1 is induced in 53G1 arrest and apoptosis. Cancer Res 54(5):1169-1174.
  193. 193. WatcharasitP.BijurG. N.ZmijewskiJ. W.SongL.ZmijewskaA.ChenX.et al.2002Direct, activating interaction between glycogen synthase kinase-3beta and 53after DNA damage. Proc Natl Acad Sci U S A 99(12):7951-7955.
  194. 194. KangT.WeiY.HonakerY.YamaguchiH.AppellaE.HungM. C.et al.2008GSK-3 beta targets Cdc25A for ubiquitin-mediated proteolysis, and GSK-3 beta inactivation correlates with Cdc25A overproduction in human cancers. Cancer Cell 1313647
  195. 195. MyerD. L.RobbinsS. B.YinM.BoivinG. P.LiuY.GreisK. D.et al.2011Absence of polo-like kinase 3 in mice stabilizes Cdc25A after DNA damage but is not sufficient to produce tumors. Mutat Res 714(1-2):1-10.
  196. 196. XieS.WuH.WangQ.CogswellJ. P.HusainI.ConnC.et al.2001Plk3 functionally links DNA damage to cell cycle arrest and apoptosis at least in part via the 53pathway. J Biol Chem 276(46):43305-43312.
  197. 197. Bahassi elM.MyerD. L.Mc KenneyR. J.HenniganR. F.StambrookP. J.2006Priming phosphorylation of Chk2 by polo-like kinase 3 (Plk3) mediates its full activation by ATM and a downstream checkpoint in response to DNA damage. Mutat Res 596(1-2):166-176.
  198. 198. HonakerY.Piwnica-WormsH.2010Casein kinase 1 functions as both penultimate and ultimate kinase in regulating Cdc25A destruction. Oncogene 292333243334
  199. 199. PiaoS.LeeS. J.XuY.GwakJ.OhS.ParkB. J.et al.2011CK1epsilon targets Cdc25A for ubiquitin-mediated proteolysis under normal conditions and in response to checkpoint activation. Cell Cycle 103531537
  200. 200. SorensenC. S.SyljuasenR. G.FalckJ.SchroederT.RonnstrandL.KhannaK. K.et al.2003Chk1 regulates the S phase checkpoint by coupling the physiological turnover and ionizing radiation-induced accelerated proteolysis of Cdc25A. Cancer Cell 33247258
  201. 201. ShimutaK.NakajoN.UtoK.HayanoY.OkazakiK.SagataN.2002Chk1 is activated transiently and targets Cdc25A for degradation at the Xenopus midblastula transition. EMBO J 211436943703
  202. 202. KanemoriY.UtoK.SagataN.2005Beta-TrCP recognizes a previously undescribed nonphosphorylated destruction motif in Cdc25A and Cdc25B phosphatases. Proc Natl Acad Sci U S A 1021862796284
  203. 203. KoledovaZ.KafkovaL. R.KramerA.DivokyV.2010DNA damage-induced degradation of Cdc25A does not lead to inhibition of Cdk2 activity in mouse embryonic stem cells. Stem Cells 283450461
  204. 204. ThomasY.CouxO.BaldinV.2010betaTrCP-dependent degradation of CDC25B phosphatase at the metaphase-anaphase transition is a pre-requisite for correct mitotic exit. Cell Cycle 92143384350
  205. 205. BansalP.LazoJ. S.2007Induction of Cdc25B regulates cell cycle resumption after genotoxic stress. Cancer Res 67733563363
  206. 206. JinJ.AngX. L.YeX.LivingstoneM.HarperJ. W.2008Differential roles for checkpoint kinases in DNA damage-dependent degradation of the Cdc25A protein phosphatase. J Biol Chem 283281932219328
  207. 207. UtoK.InoueD.ShimutaK.NakajoN.SagataN.2004Chk1, but not Chk2, inhibits Cdc25 phosphatases by a novel common mechanism. EMBO J 231633863396
  208. 208. BulavinD. V.HigashimotoY.PopoffI. J.GaardeW. A.BasrurV.PotapovaO.et al.2001Initiation of a G2/M checkpoint after ultraviolet radiation requires 38kinase. Nature 411(6833):102-107.
  209. 209. MatsuokaS.HuangM.ElledgeS. J.1998Linkage of ATM to cell cycle regulation by the Chk2 protein kinase. Science 282539518931897
  210. 210. MankeI. A.NguyenA.LimD.StewartM. Q.EliaA. E.YaffeM. B.2005MAPKAP kinase-2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase progression in response to UV irradiation. Mol Cell 1713748
  211. 211. UchidaS.YoshiokaK.KizuR.NakagamaH.MatsunagaT.IshizakaY.et al.2009Stress-activated mitogen-activated protein kinases c-Jun NH2-terminal kinase and 38target Cdc25B for degradation. Cancer Res 69(16):6438-6444.
  212. 212. GalaktionovK.LeeA. K.EcksteinJ.DraettaG.MecklerJ.LodaM.et al.1995CDC25 phosphatases as potential human oncogenes. Science 269523015751577
  213. 213. Gabrielli BG, Clark JM, McCormack AK, Ellem KA.1997Ultraviolet light-induced G2 phase cell cycle checkpoint blocks cdc25-dependent progression into mitosis. Oncogene 157749758
  214. 214. LemaireM.DucommunB.NebredaA. R.2010UV-induced downregulation of the CDC25B protein in human cells. FEBS Lett 584611991204
  215. 215. DengJ.HardingH. P.RaughtB.GingrasA. C.BerlangaJ. J.ScheunerD.et al.2002Activation of GCN2 in UV-irradiated cells inhibits translation. Curr Biol 121512791286
  216. 216. UchidaS.KumaA.OhtsuboM.ShimuraM.HirataM.NakagamaH.et al.2004Binding of 14-3-3beta but not 14-3-3sigma controls the cytoplasmic localization of CDC25B: binding site preferences of 14-3-3 subtypes and the subcellular localization of CDC25B. J Cell Sci 117(Pt 14):3011-3020.
  217. 217. DalalS. N.SchweitzerC. M.GanJ.De CaprioJ. A.1999Cytoplasmic localization of human cdc25C during interphase requires an intact 14-3-3 binding site. Mol Cell Biol 19644654479
  218. 218. BarthH.HoffmannI.KinzelV.1996Radiation with 1 Gy prevents the activation of the mitotic inducers mitosis-promoting factor (MPF) and cdc25-C in HeLa cells. Cancer Res 561022682272
  219. 219. PengC. Y.GravesP. R.ThomaR. S.WuZ.ASShaw-WormsPiwnica.H.1997Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216. Science 277533115011505
  220. 220. SanchezY.WongC.ThomaR. S.RichmanR.WuZ.Piwnica-WormsH.et al.1997Conservation of the Chk1 checkpoint pathway in mammals: linkage of DNA damage to Cdk regulation through Cdc25. Science 277533114971501
  221. 221. OggS.GabrielliB.Piwnica-WormsH.1994Purification of a serine kinase that associates with and phosphorylates human Cdc25C on serine 216. J Biol Chem 269483046130469
  222. 222. BrownA. L.LeeC. H.SchwarzJ. K.MitikuN.Piwnica-WormsH.ChungJ. H.1999A human Cds1-related kinase that functions downstream of ATM protein in the cellular response to DNA damage. Proc Natl Acad Sci U S A 96737453750
  223. 223. MorrisM. C.HeitzA.MeryJ.HeitzF.DivitaG.2000An essential phosphorylation-site domain of human cdc25C interacts with both 14-3-3 and cyclins. J Biol Chem 275372884928857
  224. 224. GravesP. R.LovlyC. M.UyG. L.Piwnica-WormsH.2001Localization of human Cdc25C is regulated both by nuclear export and 14-3-3 protein binding. Oncogene 201518391851
  225. 225. ChanP. M.NgY. W.ManserE.2011A robust protocol to map binding sites of the 14-3-3 interactome: Cdc25C requires phosphorylation of both S216 and S263 to bind 14-3-3. Mol Cell Proteomics 10(3):M110.005157.
  226. 226. Esmenjaud-MailhatC.LobjoisV.FromentC.GolsteynR. M.MonsarratB.DucommunB.2007Phosphorylation of CDC25C at S263 controls its intracellular localisation. FEBS Lett 5812139793985
  227. 227. O’NeillT.GiarrataniL.ChenP.IyerL.LeeC. H.BobiakM.et al.2002Determination of substrate motifs for human Chk1 and hCds1/Chk2 by the oriented peptide library approach. J Biol Chem 277181610216115
  228. 228. KumagaiA.DunphyW. G.1999Binding of 14-3-3 proteins and nuclear export control the intracellular localization of the mitotic inducer Cdc25. Genes Dev 13910671072
  229. 229. KumagaiA.YakowecP. S.DunphyW. G.1998proteins act as negative regulators of the mitotic inducer Cdc25 in Xenopus egg extracts. Mol Biol Cell 92345354
  230. 230. KumagaiA.GuoZ.EmamiK. H.WangS. X.DunphyW. G.1998The Xenopus Chk1 protein kinase mediates a caffeine-sensitive pathway of checkpoint control in cell-free extracts. J Cell Biol 142615591569
  231. 231. HutchinsJ. R.DikovskayaD.ClarkeP. R.2002Dephosphorylation of the inhibitory phosphorylation site S287 in Xenopus Cdc25C by protein phosphatase-2A is inhibited by 14-3-3 binding. FEBS Lett 528(1-3):267-271.
  232. 232. SorensenC. S.SyljuasenR. G.LukasJ.BartekJ.2004ATR, Claspin and the Rad9-Rad1-Hus1 complex regulate Chk1 and Cdc25A in the absence of DNA damage. Cell Cycle 37941945
  233. 233. KreutzerJ.GuerraB.2007The regulatory beta-subunit of protein kinase CK2 accelerates the degradation of CDC25A phosphatase through the checkpoint kinase Chk1. Int J Oncol 31512511259
  234. 234. SchwindlingS. L.NollA.MontenarhM.GotzC.2004Mutation of a CK2 phosphorylation site in cdc25C impairs importin alpha/beta binding and results in cytoplasmic retention. Oncogene 232341554165
  235. 235. Theis-FebvreN.FilholO.FromentC.CazalesM.CochetC.MonsarratB.et al.2003Protein kinase CK2 regulates CDC25B phosphatase activity. Oncogene 222220232
  236. 236. SchmittE.BoutrosR.FromentC.MonsarratB.DucommunB.DozierC.2006CHK1 phosphorylates CDC25B during the cell cycle in the absence of DNA damage. J Cell Sci 119(Pt 20):4269-4275.
  237. 237. KramerA.MailandN.LukasC.SyljuasenR. G.WilkinsonC. J.NiggE. A.et al.2004Centrosome-associated Chk1 prevents premature activation of cyclin-B-Cdk1 kinase. Nat Cell Biol 69884891
  238. 238. ForrestA.GabrielliB.2001Cdc25B activity is regulated by 14-3-3. Oncogene 203243934401
  239. 239. GilesN.ForrestA.GabrielliB.2003acts as an intramolecular bridge to regulate cdc25B localization and activity. J Biol Chem 278312858028587
  240. 240. DavezacN.BaldinV.GabrielliB.ForrestA.Theis-FebvreN.YashidaM.et al.2000Regulation of CDC25B phosphatases subcellular localization. Oncogene 191821792185
  241. 241. Stanford JS, Ruderman JV.2005Changes in regulatory phosphorylation of Cdc25C Ser287 and Wee1 Ser549 during normal cell cycle progression and checkpoint arrests. Mol Biol Cell 161257495760
  242. 242. BrownE. J.BaltimoreD.2003Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance. Genes Dev 175615628
  243. 243. LiuQ.GuntukuS.CuiX. S.MatsuokaS.CortezD.TamaiK.et al.2000Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev 141214481459
  244. 244. YangJ.WinklerK.YoshidaM.KornbluthS.1999Maintenance of G2 arrest in the Xenopus oocyte: a role for 14-3-3-mediated inhibition of Cdc25 nuclear import. EMBO J 18821742183
  245. 245. BrondelloJ. M.BoddyM. N.FurnariB.RussellP.1999Basis for the checkpoint signal specificity that regulates Chk1 and Cds1 protein kinases. Mol Cell Biol 19642624269
  246. 246. BulavinD. V.HigashimotoY.DemidenkoZ. N.MeekS.GravesP.PhillipsC.et al.2003Dual phosphorylation controls Cdc25 phosphatases and mitotic entry. Nat Cell Biol 56545551
  247. 247. BulavinD. V.DemidenkoZ. N.PhillipsC.MoodyS. A.FornaceA. J.Jr 2003Phosphorylation of Xenopus Cdc25C at Ser285 interferes with ability to activate a DNA damage replication checkpoint in pre-midblastula embryos. Cell Cycle 23263266
  248. 248. MargolisS. S.PerryJ. A.ForesterC. M.NuttL. K.GuoY.MJJardimet.al2006Role for the 2AB56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis. Cell 127(4):759-773.
  249. 249. MargolisS. S.PerryJ. A.WeitzelD. H.FreelC. D.YoshidaM.HaysteadT. A.et al.2006A role for 1in the Cdc2/Cyclin B-mediated positive feedback activation of Cdc25. Mol Biol Cell 17(4):1779-1789.
  250. 250. CunatS.AnahoryT.BerthenetC.HedonB.FranckhauserC.FernandezA.et al.2008The cell cycle control protein cdc25C is present, and phosphorylated on serine 214 in the transition from germinal vesicle to metaphase II in human oocyte meiosis. Mol Reprod Dev 75711761184
  251. 251. MargolisS. S.WalshS.WeiserD. C.YoshidaM.ShenolikarS.KornbluthS.20031control of M phase entry exerted through 14-3-3-regulated Cdc25 dephosphorylation. EMBO J 22(21):5734-5745.
  252. 252. KuN. O.MichieS.ResurreccionE. Z.BroomeR. L.OmaryM. B.2002Keratin binding to 14-3-3 proteins modulates keratin filaments and hepatocyte mitotic progression. Proc Natl Acad Sci U S A 99743734378
  253. 253. ClarkeP. R.HoffmannI.DraettaG.KarsentiE.1993Dephosphorylation of cdc25-C by a type-2A protein phosphatase: specific regulation during the cell cycle in Xenopus egg extracts. Mol Biol Cell 44397411
  254. 254. CrenshawD. G.YangJ.MeansA. R.KornbluthS.1998The mitotic peptidyl-prolyl isomerase, Pin1, interacts with Cdc25 and Plx1. EMBO J 17513151327
  255. 255. ZhouX. Z.KopsO.WernerA.LuP. J.ShenM.StollerG.et al.2000Pin1-dependent prolyl isomerization regulates dephosphorylation of Cdc25C and tau proteins. Mol Cell 64873883
  256. 256. Stukenberg PT, Kirschner MW.2001Pin1 acts catalytically to promote a conformational change in Cdc25. Mol Cell 7510711083
  257. 257. WinklerK. E.SwensonK. I.KornbluthS.MeansA. R.2000Requirement of the prolyl isomerase Pin1 for the replication checkpoint. Science 287545816441647
  258. 258. ForesterC. M.MaddoxJ.LouisJ. V.GorisJ.VirshupD. M.2007Control of mitotic exit by 2Aregulation of Cdc25C and Cdk1. Proc Natl Acad Sci U S A 104(50):19867-19872.
  259. 259. AstutiP.BoutrosR.DucommunB.GabrielliB.2010Mitotic phosphorylation of Cdc25B Ser321 disrupts 14-3-3 binding to the high affinity Ser323 site. J Biol Chem 285453436434370
  260. 260. HermekingH.LengauerC.PolyakK.HeT. C.ZhangL.ThiagalingamS.et al.1997sigma is a 53inhibitor of G2/M progression. Mol Cell 1(1):3-11.
  261. 261. Wilson-GradyJ. T.VillénJ.GygiS. P.2008Phosphoproteome analysis of fission yeast. Journal of Proteome Research 7310881097
  262. 262. KinoshitaN.OhkuraH.YanagidaM.1990Distinct, essential roles of type 1 and 2A protein phosphatases in the control of the fission yeast cell division cycle. Cell 632405415
  263. 263. KinoshitaN.YamanoH.NiwaH.YoshidaT.YanagidaM.1993Negative regulation of mitosis by the fission yeast protein phosphatase ppa2. Genes Dev 7610591071
  264. 264. OhkuraH.AdachiY.KinoshitaN.NiwaO.TodaT.YanagidaM.1988Cold-sensitive and caffeine-supersensitive mutants of the Schizosaccharomyces pombe dis genes implicated in sister chromatid separation during mitosis. EMBO J 7514651473
  265. 265. CargnelloM.RouxP. P.2011Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev 7515083
  266. 266. ShaulY. D.SegerR.2007The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta 1773812131226
  267. 267. LiuJ.GrimisonB.MallerJ. L.2007New insight into metaphase arrest by cytostatic factor: from establishment to release. Oncogene 26912861289
  268. 268. RauhN. R.SchmidtA.BormannJ.NiggE. A.MayerT. U.2005Calcium triggers exit from meiosis II by targeting the APC/C inhibitor XErp1 for degradation. Nature 437706110481052
  269. 269. ChunJ.ASChauMaingat. F. G.EdmondsS. D.OstergaardH. L.ShibuyaE. K.2005Phosphorylation of Cdc25C by 90Rskcontributes to a G2 cell cycle arrest in Xenopus cycling egg extracts. Cell Cycle 4(1):148-154.
  270. 270. HutchinsJ. R.DikovskayaD.ClarkeP. R.2003Regulation of Cdc2/cyclin B activation in Xenopus egg extracts via inhibitory phosphorylation of Cdc25C phosphatase by Ca(2+)/calmodulin-dependent protein [corrected]. kinase II. Mol Biol Cell 141040034014
  271. 271. WangR.JungS. Y.WuC. F.QinJ.KobayashiR.GallickG. E.et al.2010Direct roles of the signaling kinase RSK2 in Cdc25C activation during Xenopus oocyte maturation. Proc Natl Acad Sci U S A 107461988519890
  272. 272. WangR.HeG.Nelman-GonzalezM.AshornC. L.GallickG. E.StukenbergP. T.et al.2007Regulation of Cdc25C by ERK-MAP kinases during the G2/M transition. Cell 128611191132
  273. 273. AstutiP.PikeT.WidbergC.PayneE.HardingA.HancockJ.et al.2009MAPK pathway activation delays G2/M progression by destabilizing Cdc25B. J Biol Chem 284493378133788
  274. 274. PatelR.HoltM.PhilipovaR.MossS.SchulmanH.HidakaH.et al.1999Calcium/calmodulin-dependent phosphorylation and activation of human Cdc25-C at the G2/M phase transition in HeLa cells. J Biol Chem 2741279587968
  275. 275. Planas-Silva MD, Means AR.1992Expression of a constitutive form of calcium/calmodulin dependent protein kinase II leads to arrest of the cell cycle in G2. EMBO J 112507517
  276. 276. PengC. Y.GravesP. R.OggS.ThomaR. S.ByrnesM.J,3rdWu. Z.et al.1998C-TAK1 protein kinase phosphorylates human Cdc25C on serine 216 and promotes 14-3-3 protein binding. Cell Growth Differ 93197208
  277. 277. IzumiT.MallerJ. L.1995Phosphorylation and activation of the Xenopus Cdc25 phosphatase in the absence of Cdc2 and Cdk2 kinase activity. Mol Biol Cell 62215226
  278. 278. EyminB.ClaverieP.SalonC.BrambillaC.BrambillaE.GazzeriS.200614ARFtriggers G2 arrest through ERK-mediated Cdc25C phosphorylation, ubiquitination and proteasomal degradation. Cell Cycle 5(7):759-765.
  279. 279. KotlyarovA.YannoniY.FritzS.LaassK.TelliezJ. B.PitmanD.et al.2002Distinct cellular functions of MK2. Mol Cell Biol 221348274835
  280. 280. UchidaS.WatanabeN.KudoY.YoshiokaK.MatsunagaT.IshizakaY.et al.2011SCFbeta(TrCP) mediates stress-activated MAPK-induced Cdc25B degradation. J Cell Sci 124(Pt 16):2816-2825.
  281. 281. GutierrezG. J.TsujiT.CrossJ. V.DavisR. J.TempletonD. J.JiangW.et al.2010JNK-mediated phosphorylation of Cdc25C regulates cell cycle entry and G(2)/M DNA damage checkpoint. J Biol Chem 285191421714228
  282. 282. GossV. L.CrossJ. V.MaQianK.MolaY.TempletonP. W.D. J.2003SAPK/JNK regulates cdc2/cyclin B kinase through phosphorylation and inhibition of cdc25c. Cell Signal 157709718
  283. 283. Lopez-AvilesS.GrandeM.GonzalezM.HelgesenA. L.AlemanyV.Sanchez-PirisM.et al.2005Inactivation of the Cdc25 phosphatase by the stress-activated Srk1 kinase in fission yeast. Mol Cell 1714959
  284. 284. SmithD. A.BAMorganQuinn. J.2010Stress signalling to fungal stress-activated protein kinase pathways. FEMS Microbiol Lett 306118
  285. 285. Stanford JS, Lieberman SL, Wong VL, Ruderman JV.2003Regulation of the G2/M transition in oocytes of xenopus tropicalis. Dev Biol 2602438448
  286. 286. Duckworth BC, Weaver JS, Ruderman JV.2002G2 arrest in Xenopus oocytes depends on phosphorylation of cdc25 by protein kinase A. Proc Natl Acad Sci U S A 99261679416799
  287. 287. PirinoG.WescottM. P.DonovanP. J.2009Protein kinase A regulates resumption of meiosis by phosphorylation of Cdc25B in mammalian oocytes. Cell Cycle 84665670
  288. 288. BachmannM.MoroyT.2005The serine/threonine kinase Pim-1. Int J Biochem Cell Biol 374726730
  289. 289. MochizukiT.KitanakaC.NoguchiK.MuramatsuT.AsaiA.KuchinoY.1999Physical and functional interactions between Pim-1 kinase and Cdc25A phosphatase. Implications for the Pim-1-mediated activation of the c-Myc signaling pathway. J Biol Chem 274261865918666
  290. 290. WangZ.BhattacharyaN.MixterP. F.WeiW.SedivyJ.MagnusonN. S.2002Phosphorylation of the cell cycle inhibitor 21Cip1WAF1 by Pim-1 kinase. Biochim Biophys Acta 1593(1):45-55.
  291. 291. BachmannM.HennemannH.XingP. X.HoffmannI.MoroyT.2004The oncogenic serine/threonine kinase Pim-1 phosphorylates and inhibits the activity of Cdc25C-associated kinase 1 (C-TAK1): a novel role for Pim-1 at the G2/M cell cycle checkpoint. J Biol Chem 279464831948328
  292. 292. DavezacN.BaldinV.BlotJ.DucommunB.TassanJ. P.2002Human pEg3 kinase associates with and phosphorylates CDC25B phosphatase: a potential role for pEg3 in cell cycle regulation. Oncogene 215076307641
  293. 293. MireyG.ChartrainI.FromentC.QuarantaM.BoucheJ. P.MonsarratB.et al.2005CDC25B phosphorylated by pEg3 localizes to the centrosome and the spindle poles at mitosis. Cell Cycle 46806811
  294. 294. NanceJ.2005PAR proteins and the establishment of cell polarity during C. elegans development.Bioessays 27126135
  295. 295. RiversD. M.MorenoS.AbrahamM.AhringerJ.2008PAR proteins direct asymmetry of the cell cycle regulators Polo-like kinase and Cdc25. J Cell Biol 1805877885
  296. 296. KristjansdottirK.RudolphJ.2004Cdc25 phosphatases and cancer. Chem Biol 11810431051
  297. 297. LofflerH.SyljuasenR. G.BartkovaJ.WormJ.LukasJ.BartekJ.2003Distinct modes of deregulation of the proto-oncogenic Cdc25A phosphatase in human breast cancer cell lines. Oncogene 225080638071
  298. 298. GalaktionovK.ChenX.BeachD.1996Cdc25 cell-cycle phosphatase as a target of c-myc. Nature 3826591511517
  299. 299. DongJ.ZengB. H.XuL. H.WangJ. Y.LiM. Z.MSZenget.al2010Anti-CDC25B autoantibody predicts poor prognosis in patients with advanced esophageal squamous cell carcinoma. J Transl Med 8:81.
  300. 300. BuglerB.SchmittE.AressyB.DucommunB.2010Unscheduled expression of CDC25B in S-phase leads to replicative stress and DNA damage. Mol Cancer 9:29.
  301. 301. PlanellesV.BenichouS.2009Vpr and its interactions with cellular proteins. Curr Top Microbiol Immunol 339177200
  302. 302. HuardS.ElderR. T.LiangD.LiG.ZhaoR. Y.2008Human immunodeficiency virus type 1 Vpr induces cell cycle G2 arrest through Srk1/MK2-mediated phosphorylation of Cdc25. J Virol 82629042917
  303. 303. MatsudaN.TanakaH.YamazakiS.SuzukiJ.TanakaK.YamadaT.et al.2006HIV-1 Vpr induces G2 cell cycle arrest in fission yeast associated with Rad24/14-3-3-dependent, Chk1/Cds1independent Wee1 upregulation. Microbes Infect 8(12-13):2736-2744.
  304. 304. ElderR. T.YuM.ChenM.ZhuX.YanagidaM.ZhaoY.2001HIV-1 Vpr induces cell cycle G2 arrest in fission yeast (Schizosaccharomyces pombe) through a pathway involving regulatory and catalytic subunits of 2Aand acting on both Wee1 and Cdc25. Virology 287(2):359-370.
  305. 305. FrazerC.YoungP. G.2012Carboxy-terminal phosphorylation sites in Cdc25 contribute to enforcement of the DNA damage and replication checkpoints in fission yeast. Curr Genetics, DOIs00294-012-0379-1

Written By

C. Frazer and P.G. Young

Submitted: 21 November 2011 Published: 06 September 2012