Fact-checked by Grok 2 weeks ago

Cdc25

Cdc25 phosphatases constitute a family of dual-specificity protein phosphatases essential for eukaryotic progression, primarily functioning to activate cyclin-dependent kinases (CDKs) by removing inhibitory phosphate groups from specific residues, such as Thr14 and Tyr15 on CDK1. These enzymes were first identified in 1986 in the fission yeast by and colleagues as the product of the cdc25+ gene, a key positive regulator required for entry into . In mammals, three homologous isoforms—Cdc25A, Cdc25B, and Cdc25C—have evolved, each with distinct spatiotemporal expression patterns and regulatory mechanisms to orchestrate transitions through G1/S, , and G2/M checkpoints. The Cdc25 family shares a conserved catalytic domain but differs in their N-terminal regulatory regions, which confer substrate specificity and responsiveness to upstream signals like DNA damage checkpoints mediated by kinases such as Chk1 and Chk2. Cdc25A primarily drives G1/S progression and is rapidly degraded after to prevent unscheduled advancement, while Cdc25B initiates the G2/M transition by activating CDK1-cyclin B complexes at centrosomes. Cdc25C, in turn, amplifies mitotic entry and is tightly controlled during the DNA damage response to halt progression until repairs are complete. Dysregulation of these phosphatases, often through overexpression or mutation, disrupts checkpoint integrity and promotes genomic instability. Overexpression of Cdc25 isoforms is frequently observed in various human cancers, including , , and colorectal tumors, where it correlates with poor and resistance to by accelerating and evading DNA damage-induced arrest. Consequently, Cdc25 phosphatases have emerged as promising therapeutic targets, with small-molecule inhibitors under investigation to restore control in malignancies like . Their role extends beyond cancer to developmental processes and responses to genotoxic stress, underscoring their fundamental importance in cellular .

Introduction and Discovery

Historical Background

The was first identified in the fission yeast through the isolation of temperature-sensitive mutants that arrest in the of the , preventing progression to and resulting in elongated cells at the restrictive temperature. These key experiments, conducted by Fantes in 1979, demonstrated that cdc25 is essential for the G2/M transition, as mutant cells failed to initiate despite continued growth. In the , further characterization revealed cdc25's role as a positive regulator of . The was cloned by and Nurse in using complementation of temperature-sensitive mutants, showing that increased cdc25 expression advances mitotic entry at reduced cell sizes, establishing it as a dosage-dependent inducer. A homolog in budding yeast (Saccharomyces cerevisiae), known as MIH1, was cloned and characterized around the same period, linking it to activation of the Cdc28 (the budding yeast ortholog of Cdc2/Cdk1). Milestone work in 1990 by Moreno, Nurse, and highlighted the cyclic accumulation of the Cdc25 protein during the , peaking at G2/M to drive mitotic induction. The following year, Millar et al. from 's group demonstrated that Cdc25 possesses intrinsic dual-specificity activity, directly dephosphorylating inhibitory residues on Cdc2 to activate the for mitotic entry. The discovery extended to mammals in the early 1990s, with identification of multiple isoforms reflecting evolutionary conservation of Cdc25 function. Cdc25C, the first mammalian homolog, was cloned in 1990 by et al. as a nuclear protein predominantly expressed in and required for mitotic initiation. Cdc25A and Cdc25B were described in 1991 by Galaktionov and Beach as phosphatases with roles in early progression, distinct from the G2/M-specific Cdc25C. These findings solidified Cdc25 as a conserved family of regulators linking to activation.

Definition and Nomenclature

The Cdc25 phosphatases constitute a family of dual-specificity protein phosphatases within the protein tyrosine phosphatase (PTP) superfamily, distinguished by their capacity to dephosphorylate both phosphotyrosine and phosphothreonine residues on substrate proteins. These enzymes play a pivotal role in cell cycle regulation by activating cyclin-dependent kinase (CDK) complexes through the removal of inhibitory phosphate groups. Originally identified in the fission yeast Schizosaccharomyces pombe as the product of the cdc25 gene essential for mitotic entry, the nomenclature reflects species-specific conventions: lowercase "cdc25" denotes the genes and proteins in yeast orthologs. In humans and other mammals, the orthologous genes are designated with uppercase "CDC25," comprising three primary isoforms—CDC25A, CDC25B, and CDC25C—each encoded by distinct loci on separate chromosomes: CDC25A at 3p21.1, CDC25B at 20p13.1, and CDC25C at 5q31.2. Orthologs in non-mammalian species exhibit varied nomenclature; for instance, in , the two Cdc25 homologs are known as , which regulates mitotic divisions, and , which controls meiotic progression. This conserved family underscores Cdc25's fundamental function in promoting advancement across eukaryotes via CDK activation.

Structural Features

Domain Organization

The Cdc25 family of dual-specificity phosphatases exhibits a conserved modular comprising an N-terminal regulatory domain and a C-terminal catalytic domain, which together enable their roles in cell cycle control. The N-terminal regulatory domain varies in length across isoforms, typically spanning 200 to 400 residues, and serves as a platform for interactions with binding partners while harboring potential sites that influence protein stability and localization. This region displays low sequence conservation among isoforms (20-25% identity) and is characterized as intrinsically disordered in Cdc25A, Cdc25B, and Cdc25C, contributing to its flexibility in regulatory functions. In contrast, the C-terminal catalytic domain is highly conserved (approximately 60% pairwise identity between isoforms) and consists of 150 to 200 residues that fold into a compact protein tyrosine phosphatase (PTP)-like structure. This domain features key signature motifs, including the HCX5R loop that coordinates the catalytic cysteine residue essential for phosphotyrosine dephosphorylation. Early structural studies, including X-ray crystallography of the Cdc25A catalytic domain at 2.3 Å resolution in 1998 and subsequent analyses of Cdc25B around 2000, revealed an α/β fold with a central five-stranded β-sheet surrounded by α-helices, distinct from classical PTPs yet functionally analogous. NMR spectroscopy further confirmed the compact, stable nature of this domain in isolation. Isoform-specific variations in domain organization underscore their distinct temporal roles in the . Cdc25A possesses a relatively shorter N-terminal domain (approximately 313 residues) compared to Cdc25B (about 384 residues), potentially influencing its rapid turnover and broad activity. Cdc25C, with an N-terminal region of around 260 residues, includes localization signals (NLS) such as a bipartite basic motif involving residues 298-315, facilitating its accumulation during G2/M progression. Overall, the full-length proteins range in size from approximately 53 (Cdc25C, 473 residues) to 65 (Cdc25B, 580 residues), with Cdc25A at about 59 (524 residues), reflecting these domain length differences.

Catalytic Site

The catalytic site of Cdc25 phosphatases is characterized by a conserved geometry that deviates from classical protein phosphatases (PTPs), featuring a shallow pocket adapted for dual-specificity of phosphothreonine and phosphotyrosine residues. Central to this site is the signature motif HCX₅R, where the residue (e.g., Cys430 in Cdc25A, Cys473 in Cdc25B) serves as the , and the (e.g., Arg436 in Cdc25A, Arg479 in Cdc25B) stabilizes the negatively charged group through electrostatic interactions during . Unlike classical PTPs, which rely on a or aspartate in a WPD loop as the general acid, Cdc25 employs a conserved aspartate (e.g., Asp383 in Cdc25A) positioned to act as the general acid, protonating the in the first catalytic step and facilitating the second step as a general base. The reaction proceeds via a two-step mechanism typical of cysteine-based phosphatases. In the first step, the deprotonated thiolate of the catalytic performs a nucleophilic attack on the substrate's group, displacing the protein and forming a transient phosphoenzyme intermediate covalently bound to the . This is followed by a second step where a , activated by the conserved aspartate, hydrolyzes the phosphocysteine intermediate, releasing inorganic and regenerating the enzyme. This aspartate-based catalysis ensures efficient turnover without the structural constraints of a mobile WPD loop, contributing to the enzyme's specificity for protein substrates over small-molecule analogs. Cdc25 phosphatases exhibit high specificity for the inhibitory sites Thr14 and Tyr15 on cyclin-dependent kinases (CDKs), enabling targeted during progression. The shallow geometry facilitates direct access to these residues within the CDK segment. A 2024 cryo-EM structure of the CDK2-cyclin A-CDC25A complex at 2.7 Å resolution revealed a previously unrecognized C-terminal of CDC25A (residues 495-524) at the CDK2-cyclin A interface, stabilizing the complex and providing insights into substrate recognition. In vitro kinetic assays indicate moderate affinity and turnover rates for substrates, with catalytic rate constants (kₐₜ) around 1–2 s⁻¹ based on phosphoenzyme breakdown, underscoring efficient processing of physiological targets despite the enzyme's broad dual-specificity potential. These parameters were derived from studies using purified catalytic domains and mimics of CDK sites, highlighting the site's evolutionary optimization for regulation. Structural insights into the catalytic site were provided by the of the Cdc25B catalytic domain at 2.2 resolution, revealing a compact α/β fold with a shallow, open pocket that lacks deep substrate-binding grooves typical of classical PTPs. This structure, determined in an inhibitor-bound state, illustrates how the catalytic and adjacent residues form a flexible loop that accommodates bulky CDK substrates, while also exposing vulnerabilities for oxidative inactivation via formation.

Biochemical Function

Dephosphorylation of CDKs

Cdc25 phosphatases activate cyclin-dependent kinases (CDKs) by removing inhibitory phosphate groups from specific residues in the ATP-binding domain of CDK1 and CDK2. These residues include threonine 14 (Thr14) and tyrosine 15 (Tyr15), which are phosphorylated by kinases such as Wee1 and Myt1 to maintain CDK inactivity during interphase. All three Cdc25 isoforms (A, B, and C) dephosphorylate both Thr14 and Tyr15 on CDK1 and CDK2, with isoform-specific preferences; for example, Cdc25A shows a particular preference for Thr14 on CDK2. This dephosphorylation relieves steric hindrance in the active site, enabling ATP binding and full kinase activation. The activation primarily targets cyclin-bound CDK complexes critical for cell cycle progression. Cdc25C dephosphorylates the B-CDK1 complex, promoting entry into by enhancing CDK1's ability to phosphorylate substrates involved in breakdown and condensation. Similarly, Cdc25A acts on E-CDK2 and A-CDK2 complexes to facilitate the , where CDK2 drives initiation. These actions ensure timely progression through the by counterbalancing inhibitory phosphorylations. Cdc25-mediated activation follows a stepwise model characterized by sequential dephosphorylation, beginning with Tyr15 followed by Thr14, which exhibits positive to amplify the response. Initial removal of the Tyr15 partially activates the CDK, facilitating subsequent Thr14 dephosphorylation and full catalytic competence. This ordered process, observed in biochemical assays, ensures a sharp transition in CDK activity rather than gradual . Recent structural studies, including a 2024 cryo-EM of the CDK2-cyclin A-CDC25A , have elucidated the molecular basis of substrate recognition and dephosphorylation. In vitro studies from the 1990s demonstrated Cdc25's role using cell-free egg extracts, where recombinant Cdc25 reconstituted CDK activity by dephosphorylating inhibited B-CDK1 complexes, inducing mitotic events such as germinal vesicle breakdown. These assays confirmed that Cdc25 directly reverses inhibitory without requiring additional factors, establishing its essential function in CDK . A key regulatory feature is the loop where activated CDKs phosphorylate Cdc25 on multiple sites, enhancing its activity and subcellular localization. For instance, CDK1 phosphorylates Cdc25C on residues such as Thr48 and Ser214, increasing its catalytic efficiency toward CDK1 itself, thereby amplifying mitotic entry. This multisite creates an ultrasensitive switch for rapid advancement.

Substrate Specificity

Cdc25 phosphatases display isoform-specific preferences for their primary substrates, which are the inhibitory sites on cyclin-dependent kinases (CDKs). Cdc25A predominantly dephosphorylates CDK2 at Thr14 and Tyr15, facilitating activation of CDK2-cyclin E and CDK2-cyclin A complexes during the . In contrast, Cdc25B and Cdc25C primarily target CDK1 at the same sites, activating CDK1-cyclin B for mitotic entry, with Cdc25B initiating this at centrosomes in late and Cdc25C contributing in the during . These preferences arise from structural interactions, as demonstrated by kinetic assays showing Cdc25A's high catalytic efficiency (k_{cat}/K_m \approx 10^6 \, M^{-1} s^{-1}) toward the CDK2-pTpY-cyclin A complex, while Cdc25C exhibits negligible activity against it. Beyond core CDKs, Cdc25 isoforms show varying breadth in substrate recognition. Cdc25A exhibits the broadest activity, extending to early G1/S CDKs and potentially other phosphoproteins, whereas Cdc25B is more restricted to mitotic contexts and CDK1-specific activation, and Cdc25C is largely nuclear and focused on late /M targets. Experimental phosphoproteomics and studies from the have elucidated these differences, identifying a preference for substrates with basic residues (e.g., or ) adjacent to the phospho-Thr or phospho-Tyr sites, which enhance binding to the Cdc25 catalytic domain. For instance, substrate-trapping mutants (e.g., D424N in Cdc25A) and docking simulations with CDK2-pTpY-cyclin A confirmed that basic motifs near the phosphorylation site stabilize the enzyme-substrate complex, with the HCX5R catalytic motif playing a key role in . Evidence for secondary substrates beyond CDKs is limited and context-dependent, often involving low-affinity interactions.

Regulation Mechanisms

Post-Translational Modifications

Cdc25 phosphatases undergo extensive post-translational modifications, primarily , which modulate their activity, stability, and localization. Each isoform possesses multiple phosphorylation sites, with specific examples including at least 13 sites on Cdc25B by and 5 on Cdc25C by CDK1, and activating phosphorylations occurring on residues by kinases such as CDK1/2 and , enhancing catalytic efficiency during progression. For instance, CDK1 phosphorylates Cdc25C at multiple sites including Thr48, Thr67, Thr138, Ser205, and Ser285, promoting its activation at the G2/M transition. Inhibitory phosphorylations, conversely, are mediated by checkpoint kinases like CHK1 and CHK2; a key example is CHK1 phosphorylation of Cdc25C at Ser216, which creates a for 14-3-3 proteins and inhibits activity. studies from the 1990s and 2000s, such as those by Peng et al. (1997), mapped these sites and demonstrated that mutants lacking specific phosphorylation sites, like Ser/Thr-to-Ala substitutions on Cdc25B, abolish activation by at 13 sites (e.g., Ser50, Thr58). Ubiquitination represents another critical modification, particularly for Cdc25A, where SCF^β-TrCP ligase targets the protein for proteasomal following CHK1-mediated at Ser76. This process maintains Cdc25A at approximately 30 minutes during , preventing untimely CDK activation. Busino et al. (2003) showed that this pathway is essential for DNA damage responses, as β-TrCP depletion stabilizes Cdc25A and hyperactivates CDK2. Additional modifications include . of Cdc25A by ARD1 at residues increases its stability by inhibiting ubiquitination, extending beyond 15 minutes and reducing activity in response to genotoxic stress. Recent studies have also highlighted , such as N6-methyladenosine (m6A) modification enhancing Cdc25A mRNA stability to promote in cancers like esophageal (as of 2024). Temporal dynamics are evident in , where Cdc25C undergoes hyper at over 15 sites by CDK1, including 11 confirmed Ser/Thr-Pro motifs, sharpening the mitotic entry switch; mutants like Cdc25-13A delay this process and alter timing. In DNA damage scenarios, inhibitory phosphorylations dominate, such as CHK1/CHK2 targeting Cdc25A at Ser123, Ser178, Ser278, and Ser292, accelerating its degradation to enforce G2 arrest. These modifications, elucidated through 1990s-2000s studies like Hoffmann et al. (1994) on site mapping, ensure precise of Cdc25 function, with recent structural insights from cryo-EM (2024) revealing how N-terminal phosphorylation sites influence Cdc25A interactions with CDK2-cyclin A complexes.

Cellular Localization and Binding Partners

Cdc25 phosphatases exhibit dynamic subcellular localization that is critical for their spatiotemporal regulation during the , mediated by specific nuclear localization signals (NLS) and nuclear export signals (). In Cdc25C, a bipartite NLS located in the regulatory domain facilitates nuclear import, allowing the phosphatase to access nuclear (CDK) substrates. Conversely, Cdc25A and Cdc25B contain N-terminal NES motifs that promote cytoplasmic retention via CRM1-dependent export until activation signals trigger nuclear translocation. Mutation of the Cdc25A results in predominant nuclear accumulation, underscoring its role in shuttling dynamics. The three mammalian Cdc25 isoforms display distinct localization patterns aligned with their cell cycle roles. Cdc25A shuttles between the and throughout , enabling regulation of G1/S and S-phase progression. Cdc25B is primarily cytoplasmic during S and G2 phases but translocates to the at to initiate mitotic entry. In contrast, Cdc25C is predominantly nuclear, though it can be sequestered in the during via interactions that mask its NLS. These patterns were elucidated through studies tracking isoform-specific GFP fusions in synchronized cells. Key binding partners modulate Cdc25 localization to ensure precise timing of activity. The 14-3-3 proteins bind phosphorylated Cdc25C at Ser-216, occluding its NLS and sequestering it in the during to prevent premature mitotic activation. Polo-like kinase 1 () phosphorylates Cdc25C at Ser-198 within its , promoting nuclear import during and facilitating G2/M transition. Co-immunoprecipitation assays from the early 2000s confirmed these interactions and their impact on shuttling. Additionally, Cdc25 forms complexes with cyclin-CDK substrates through N-terminal RXL docking motifs, which bind the hydrophobic patch on cyclins to enhance substrate access and specificity.

Role in Cell Cycle Progression

G1/S Transition

Cdc25A plays a central role in the by dephosphorylating (CDK2) complexes bound to E or A, thereby activating these kinases to drive progression into . This activation enables CDK2- E/A to phosphorylate the (Rb) on multiple sites, resulting in Rb hyperphosphorylation and the release of bound transcription factors. Freed E2F then induces the transcription of S-phase genes, including those encoding machinery components like and , facilitating the initiation of . Cdc25A protein levels accumulate and peak during late G1 and early , coinciding with the need for CDK2 activation to commit cells to replication. Following entry, Cdc25A undergoes rapid ubiquitin-mediated proteasomal degradation, primarily through SCF^β-TrCP and other E3 ligases, which prevents excessive CDK activity and potential re-replication of the genome. This tightly controlled turnover ensures that Cdc25A function is temporally restricted to the G1/S boundary. In the context of DNA damage checkpoints, CHK2 kinase phosphorylates Cdc25A at specific serine residues (e.g., Ser123), promoting its ubiquitination and degradation, which inhibits CDK2 activation and enforces G1/S arrest to permit before replication. This mechanism integrates genotoxic stress signals to halt progression, highlighting Cdc25A's role as a key checkpoint effector. Overexpression of Cdc25A in mammalian cell lines, such as human fibroblasts, accelerates the by enhancing CDK2 activity and reducing the duration of , leading to premature S-phase entry. In , the Cdc25 homolog Mih1 dephosphorylates CDK orthologs (e.g., Cdc28 in ) to regulate transitions, with analogous functions in promoting phase-specific progression despite differences in primary timing.

G2/M Transition

Cdc25B initiates the G2/M transition through its localization to and subsequent nuclear import during , where it dephosphorylates and activates the CDK1-cyclin B complex, promoting centrosome separation and early events. This activation occurs specifically at centrosomes, highlighting Cdc25B's unique role in triggering mitotic entry independent of nuclear events initially. In contrast, Cdc25C contributes to the amplification and maintenance of CDK1 activity once is underway. The hyperphosphorylated form of Cdc25C, generated by multisite phosphorylation during mitotic entry, enhances its phosphatase activity and sustains CDK1-cyclin B activation through metaphase by continuously dephosphorylating inhibitory sites on CDK1. This persistent activity ensures the high CDK1 levels required for proper chromosome congression and metaphase maintenance. The spindle assembly checkpoint (SAC), involving BubR1 as a key component of the mitotic checkpoint complex, inhibits the anaphase-promoting complex/cyclosome (APC/C) until chromosomes achieve bipolar attachment and alignment at the metaphase plate. This checkpoint indirectly supports Cdc25C's role by preventing premature cyclin B degradation, thereby allowing sustained CDK1 activity facilitated by Cdc25C until alignment is complete. BubR1's pseudokinase domain contributes to APC/C inhibition, ensuring mitotic fidelity during this Cdc25C-dependent phase. Experimental evidence underscores these roles: microinjection of neutralizing anti-Cdc25 antibodies into immature starfish oocytes prevents Cdc25 activation, blocking cdc2 kinase (CDK1 homolog) activation and halting mitotic entry, demonstrating Cdc25's essential function in M-phase initiation. In human HeLa cells, Cdc25B overexpression induces premature mitosis from S or G2 phases, often bypassing DNA replication completion, whereas Cdc25C requires G2 phase and co-expression with cyclin B to promote entry, indicating distinct temporal contributions to the G2/M transition. A critical loop amplifies this process, as activated CDK1 phosphorylates Cdc25B and Cdc25C at multiple sites (e.g., S323 on Cdc25B and T48, T67 on Cdc25C), enhancing their activities and further activating CDK1, thereby committing cells irreversibly to . This ultrasensitive loop refines the abrupt switch from to M , with distributive ensuring rapid amplification. Post-translational modifications, such as these phosphorylations, tightly regulate the loop's timing.

Evolutionary Aspects

Conservation Across Species

The Cdc25 family of dual-specificity phosphatases demonstrates remarkable evolutionary across eukaryotes, reflecting their fundamental role in regulation. The catalytic domain, responsible for dephosphorylating cyclin-dependent kinases (CDKs) at inhibitory and residues, exhibits approximately 37% sequence identity between the human Cdc25 proteins and their ortholog in fission yeast (). This domain's core motifs, including the HCX5R signature, are highly preserved, enabling Cdc25 to activate CDKs universally and ensuring progression through the phases. The conservation extends to the functional mechanism, where Cdc25 counteracts inhibitory on conserved Thr14 and Tyr15 sites of CDKs, a feature shared from unicellular yeasts to multicellular metazoans. Orthologs of Cdc25 vary in number and similarity across species, highlighting both preservation and adaptation within eukaryotes. In S. pombe, a single Cdc25 isoform serves as the primary mitotic inducer, essential for dephosphorylating Cdc2 (the CDK homolog) to trigger mitosis. The budding yeast Saccharomyces cerevisiae possesses Mih1 as its Cdc25 ortholog, which shares lower sequence similarity but similarly regulates Cdc28p phosphorylation during the G2/M transition. In contrast, metazoans have undergone gene duplication, resulting in three isoforms in humans (Cdc25A, Cdc25B, and Cdc25C), each retaining the conserved catalytic core while diverging in regulatory domains. Functional homology is underscored by cross-species complementation, where orthologs from distant eukaryotes can substitute for one another. For instance, Cdc25C rescues the temperature-sensitive cdc25 mutation in S. pombe, restoring mitotic entry by dephosphorylating Cdc2 at Tyr15. string (a Cdc25 homolog) similarly complements S. pombe cdc25 mutants, confirming the interchangeability of these phosphatases despite sequence divergence. These assays from the early established the universality of Cdc25 function, linking yeast models to higher eukaryotes. Phylogenetically, Cdc25 phosphatases belong to the rhodanese superfamily and emerged alongside the eukaryotic lineage approximately 1.5 billion years ago, coinciding with the evolution of complex cell cycles. No orthologs exist in prokaryotes, emphasizing Cdc25's role as a eukaryotic innovation for precise temporal control of division.

Species-Specific Variations

In the fission yeast Schizosaccharomyces pombe, Cdc25 serves as the sole essential dual-specificity phosphatase for mitotic entry, featuring a notably long N-terminal regulatory domain that undergoes phosphorylation to control its activity and localization during the G2/M transition. In contrast, the budding yeast Saccharomyces cerevisiae lacks a direct ortholog but employs Mih1, a Cdc25 homolog, which functions redundantly with protein phosphatase 2C isoforms (Ptc2 and Ptc3) to dephosphorylate Cdc28 and terminate cell cycle delays, rendering Mih1 non-essential for viability. Among invertebrates, the fruit fly expresses multiple Cdc25 homologs, including , which is specifically required for meiotic progression in both male and female germlines rather than somatic mitoses, with expression confined to germ cells starting at embryogenesis. In the nematode , four distinct Cdc25 orthologs (Cdc-25.1 through Cdc-25.4) exhibit specialized roles in embryonic cell divisions; for instance, Cdc-25.2 promotes intestinal cell divisions post-16E stage by activating the CDK-1/CYB-1 complex, while Cdc-25.1 restricts in the intestinal lineage. Vertebrate Cdc25 isoforms show progressive diversification, with mammals expressing three canonical forms (Cdc25A, Cdc25B, and Cdc25C) that coordinate distinct cell cycle phases, compared to three isoforms (Cdc25A, Cdc25B, and Cdc25C) in amphibians like Xenopus laevis and multiple isoforms, including Cdc25a and Cdc25d, in teleost fish such as zebrafish during embryogenesis. Higher vertebrates display enhanced regulatory complexity, including an increased number of phosphorylation sites on Cdc25 proteins—such as multiple N-terminal motifs targeted by Chk1 and Chk2 kinases in mammals—to fine-tune stability and activity in response to DNA damage. In plants, Cdc25-like phosphatases, such as the Arabidopsis thaliana AtCdc25, represent the first identified green lineage dual-specificity orthologs but diverge functionally by primarily targeting phosphotyrosine on CDK-like kinases at non-canonical sites, lacking the broad substrate specificity of animal Cdc25 counterparts and instead contributing to stress responses rather than strict cell cycle induction. These species-specific adaptations were elucidated through post-2000 genome sequencing efforts, including the Drosophila (2000) and C. elegans (updated annotations) projects, alongside RNAi knockdown studies that revealed isoform-specific defects, such as delayed embryonic divisions in C. elegans upon Cdc-25.2 depletion.

Experimental Studies

Genetic Knockout Models

Genetic knockout models have elucidated the non-redundant and overlapping functions of Cdc25 phosphatases in control across species, revealing their essentiality for , , and . In mice, homozygous of Cdc25A leads to embryonic around embryonic day 6.5 (E6.5), characterized by defects, widespread , and failure of peri-implantation . This phenotype underscores Cdc25A's critical role in early embryonic cell divisions, consistent with its broad expression during embryogenesis. Homozygous Cdc25B knockout mice are viable and develop normally but exhibit female-specific sterility due to arrest at I of in oocytes, preventing resumption of and activation of (MPF). Male Cdc25B-null mice are fertile, indicating isoform-specific requirements for female . In contrast, Cdc25C knockout mice are fully viable, fertile, and show no overt developmental abnormalities or defects in , T- and B-cell development, or checkpoint responses. This lack of suggests significant functional with Cdc25A and Cdc25B in somatic cells. Mice with double knockout of Cdc25B and Cdc25C are also viable at expected Mendelian ratios, with normal embryonic development, cell cycle progression, and DNA damage checkpoint responses in embryonic fibroblasts. These findings highlight compensatory mechanisms by Cdc25A, as triple inactivation via conditional approaches results in synthetic lethality and mitotic defects. In fission yeast (Schizosaccharomyces pombe), temperature-sensitive cdc25 mutants arrest uniformly in G2 phase at restrictive temperatures, forming elongated cells unable to enter mitosis due to persistent inhibitory phosphorylation of Cdc2 (CDK homolog). This G2 arrest is rescued by co-inactivation of wee1, which hyperactivates Cdc2, demonstrating cdc25's role as a positive regulator opposing Wee1-mediated inhibition. Such mutants were instrumental in defining the G2/M transition and size control in eukaryotic cell cycles during the 1980s and 1990s. These models, generated primarily through in the 1990s and early 2000s, illustrate isoform compensation and context-specific essentiality, informing broader understanding of Cdc25 functions beyond simple redundancy.

In Vitro and Cell-Based Assays

assays for Cdc25 activity primarily utilize recombinant proteins expressed in bacterial or insect cell systems to measure of model substrates. A standard colorimetric method employs p-nitrophenyl phosphate (pNPP) as the substrate, where Cdc25 catalyzes its to p-nitrophenol, detectable by at 405 nm; this approach quantifies activity across all human Cdc25 isoforms (A, B, and C) under controlled conditions of and . Alternatively, radioactive assays use 32P-labeled (CDK) substrates, such as phosphorylated CDK2 or CDK1, followed by separation and autoradiographic detection of dephosphorylated products to assess specific dual-specificity function. These assays enable calculation of inhibitory concentration 50% () values for potential Cdc25 inhibitors by plotting dose-response curves, where inhibitor potency is determined from the concentration reducing substrate by half, often in the nanomolar range for selective compounds. Cell-based assays provide insights into Cdc25 function within cellular contexts, often using human embryonic kidney (HEK293) or cells for their robust efficiency. Transient overexpression of Cdc25 isoforms, achieved via , activates downstream CDKs, which is monitored through histone H1 kinase assays measuring of as a proxy for CDK2 or CDK1 activity; for instance, Cdc25B overexpression in HEK293 cells accelerates G2/M progression by enhancing B1-CDK1 activation at centrosomes. Conversely, small interfering RNA (siRNA)-mediated knockdown demonstrates loss-of-function effects, where depletion of Cdc25A or Cdc25B in HEK293 cells delays mitotic entry and reduces A-CDK2 activity, as evidenced by diminished in multiple replicates; combined knockdown of Cdc25A and Cdc25B fully blocks , underscoring their redundant yet essential roles. These assays highlight Cdc25's integration into regulatory networks without requiring whole-organism models. High-throughput screening (HTS) efforts in the early 2000s have facilitated Cdc25 discovery by adapting fluorescence polarization assays to detect binding or enzymatic inhibition. In one seminal screen of the National Cancer Institute's chemical library against recombinant Cdc25B, fluorescence polarization measured displacement of a fluorescently labeled analog, identifying NSC95397 as a potent, mixed-mode with values of 32 nM for Cdc25A, 96 nM for Cdc25B, and 40 nM for Cdc25C; this compound exhibited 125- to 180-fold selectivity over related phosphatases like VHR and PTP1B, and it blocked G2/M transition in cell lines. Such screens have prioritized quinone-based scaffolds like NSC95397 for further development, establishing benchmarks for selectivity and cellular efficacy. Xenopus egg extracts serve as a powerful cell-free system to reconstitute Cdc25-dependent mitotic cycles, mimicking physiological oscillations in phosphatase activity. Interphase or CSF-arrested extracts are supplemented with cyclin B and monitored for Cdc2/cyclin B activation via histone H1 kinase assays, revealing that Cdc25C dephosphorylates inhibitory sites on Cdc2 to drive mitotic entry; for example, calcium-induced activation of CaMKII phosphorylates Cdc25C at S287, inhibiting its activity and delaying mitosis until dephosphorylation restores the cycle. This model has elucidated feedback loops, such as Cdc25 auto-amplification, and checkpoint overrides, providing quantitative insights into timing (e.g., S287 phosphorylation peaks at 75-90 minutes post-interphase entry). Recent advances leverage CRISPR-Cas9 editing for isoform-specific interrogation of Cdc25 in human pluripotent s (PSCs), including induced PSCs, to dissect dependencies. Genome-wide CRISPR interference screens in human and PSCs have identified species-specific vulnerabilities in regulators, including components of the CDK1/CDK2 regulatory network involving CDC25. These approaches enable precise, high-fidelity studies of Cdc25 redundancy across isoforms in stem cell contexts.

Implications in Human Disease

Association with Cancer

The Cdc25 family of phosphatases, particularly Cdc25A, Cdc25B, and Cdc25C, is frequently overexpressed in various human cancers, with reported frequencies ranging from 20% to 60% across tumor types such as , colorectal, , and hepatocellular carcinomas. This overexpression often correlates with advanced disease stages, increased tumor aggressiveness, and poor patient prognosis, as evidenced by higher relapse rates and reduced overall survival in affected cohorts. Mechanistically, Cdc25A overexpression promotes unchecked progression through the G1/S checkpoint by dephosphorylating and activating E-CDK2 complexes, thereby bypassing (Rb) protein-mediated arrest and facilitating uncontrolled proliferation. In contrast, elevated Cdc25B and Cdc25C levels accelerate the G2/M transition by activating B-CDK1, leading to premature mitotic entry, replication stress, and genomic instability that contributes to oncogenesis. Genomic alterations in Cdc25 genes are less common than overexpression but include missense mutations observed in approximately 3-4% of cases across diverse cancers such as , colon, , , stomach, and uterine; however, promoter hypomethylation is a prevalent epigenetic mechanism driving upregulation, particularly in , , and tumors. In p53-mutant tumors, which comprise over 50% of cancers, Cdc25A hyperactivity is exacerbated due to the loss of p53-mediated transcriptional repression, resulting in abrogation of DNA damage checkpoints and evasion of arrest following genotoxic stress. As of 2025, analyses of (TCGA) datasets reveal consistent upregulation of Cdc25 family members in 14 cancer types, including colorectal and breast invasive carcinoma, associating this pattern with adverse clinical outcomes. Supporting functional comes from xenograft models, where knockdown of Cdc25A significantly reduces tumor growth and in nude mice implanted with liver, cervical, or cells, underscoring its oncogenic role.

Role in Other Pathologies

Cdc25 phosphatases have been implicated in several non-cancer pathologies through dysregulation of cell cycle control and related signaling pathways. In developmental disorders, particularly microcephalic osteodysplastic primordial dwarfism type II (MOPD II), Cdc25B plays a key role in centrosomal regulation of mitotic entry. Mutations or disruptions in upstream regulators like pericentrin (PCNT) lead to improper Chk1-mediated phosphorylation of Cdc25B, resulting in delayed cyclin B-Cdk1 activation and spindle misalignment, which contribute to reduced brain size and proportionate dwarfism. This pathway disruption highlights Cdc25B's involvement in the severe growth failure characteristic of MOPD II, as evidenced by associations in disease databases linking Cdc25B to the disorder. In neurodegenerative diseases such as (AD), Cdc25A activation in postmitotic neurons promotes aberrant re-entry, leading to neuronal death. Studies have shown elevated Cdc25A levels and activity in degenerating neurons from AD brains, where it co-localizes with neurofibrillary tangles and neuritic plaques, facilitating of substrates that exacerbate pathology. Cdc25A overexpression in neuronal models induces progression and , with its tyrosine enhancing Cdk activity that indirectly contributes to hyperphosphorylation through dysregulated networks, including interactions with Cdk5-p25. Furthermore, Cdc25A inhibitors have demonstrated neuroprotective effects in AD models by preventing this aberrant activation. Cdc25C dysregulation has been observed in cardiovascular pathologies, including models of cardiac remodeling and failure. In mice with cardiomyocyte-specific GSK-3 knockout, increased inhibitory phosphorylation of Cdc25C at Ser-216 correlates with elevated Cdk1 and B1 levels, leading to and maladaptive . This suggests that impaired Cdc25C function disrupts G2/M checkpoint control, promoting aberrant cardiomyocyte and contributing to pathological remodeling in response to stress. In infectious diseases, viruses such as human papillomavirus (HPV) hijack Cdc25A to facilitate host cell transformation and viral replication. The HPV-16 E7 oncoprotein maintains elevated Cdc25A levels by preventing its ubiquitin-proteasome degradation, thereby sustaining Cdk2 activity and overriding during differentiation. This stabilization allows HPV to reprogram quiescent for , supporting viral genome amplification and contributing to persistent infection. Regarding and , Cdc25A regulates T-cell activation and , with implications for diseases like (RA). In + T cells, Cdc25A expression is suppressed by PD-1 signaling to inhibit via p27 and p15 upregulation, and its dysregulation promotes hyperproliferative responses in autoimmune contexts. Preclinical studies indicate that targeting Cdc25A could modulate T-cell effector functions in RA, as miR-34a-3p-mediated downregulation of Cdc25A reduces synovial fibroblast driven by inflammatory signals. This positions Cdc25A as a potential regulator of immune cell hyperactivity in inflammatory arthritides.

Therapeutic Potential

Cdc25 Inhibitors

Cdc25 phosphatases have been recognized as promising therapeutic targets since the mid-1990s, when initial high-throughput screens identified the first small-molecule inhibitors, such as the unselective alkaloid natural products known as dnacins. Development efforts intensified in the , focusing on optimizing potency, selectivity, and pharmacokinetic properties through structure-activity relationship () studies and rational design based on the enzyme's catalytic domain. These advances led to diverse classes of inhibitors, primarily small molecules, aimed at disrupting Cdc25 function to halt aberrant progression in cancer cells. Inhibitor classes include quinone-based compounds, which act as irreversible oxidants of the catalytic residue in the . A representative example is NSC663284, a potent derivative with values of approximately 29 nM for Cdc25A, 95 nM for Cdc25B, and 89 nM for Cdc25C, demonstrating broad activity across isoforms by forming a covalent with the essential Cys residue. mimetics represent another class, designed to mimic substrates and target the catalytic site directly; for instance, a pentapeptide achieves an of 1.1 μM against Cdc25A by competing for the phosphatase's active pocket. Mechanisms of inhibition vary, with competitive inhibitors binding directly to the to block access. BN82685, a quinone-based compound, exemplifies this approach, exhibiting IC50 values in the low nanomolar range (e.g., 201 nM for Cdc25C) and preventing dephosphorylation of cyclin-dependent kinases. In contrast, allosteric inhibitors disrupt regulatory domains without occupying the catalytic site; allosteric inhibitors derived from computational screening impair Cdc25B activity through conformational changes, demonstrating potential isoform specificity. Challenges in selectivity arise from the structural similarities among the dual-specificity Cdc25 isoforms (A, B, and C), complicating isoform-specific targeting. Cdc25A-specific inhibitors like ARQ-501 address this by preferentially inhibiting Cdc25A ( ~1 μM), minimizing off-target effects on other phosphatases. Pan-Cdc25 inhibitors, such as the natural product derived from , indirectly suppress all isoforms through modulation (antiproliferative values in the 10-20 μM range), leveraging broad suppression for antitumor effects. In preclinical models, Cdc25 inhibitors have shown efficacy by inducing G2/M phase arrest in tumor cells, preventing activation and mitotic entry. Notably, compounds like NSC663284 exhibit synergy with DNA-damaging agents, enhancing through prolonged checkpoint activation and increased in cancer cell lines, without notable effects in normal cells. As of 2025, novel quinoid-based CDC25 inhibitors have demonstrated antiproliferative effects in lung adenocarcinoma models and are advancing toward applications.

Clinical Developments

Early clinical efforts targeting Cdc25 phosphatases focused on small-molecule inhibitors, with ARQ-501 (also known as beta-lapachone) representing one of the few compounds to advance into trials. Investigated for its effects on including indirect modulation of Cdc25 pathways, ARQ-501 underwent phase I testing in patients with advanced, chemotherapy-resistant solid tumors during the mid-2000s, demonstrating preliminary antitumor activity but exhibiting dose-limiting toxicities such as . A subsequent phase II trial combined ARQ-501 with in treatment-naïve patients with unresectable pancreatic , where the regimen showed modest with partial responses in a subset of participants, though overall was limited, and concerns persisted. Another phase II study evaluated ARQ-501 monotherapy in advanced of the head and neck, reporting stable disease in approximately 20% of patients but no complete responses, leading to halted further development due to insufficient relative to . As of 2025, no Cdc25-specific inhibitors have reached late-stage clinical development or regulatory approval, with most candidates remaining in preclinical or early-phase exploration. Recent preclinical data highlight promising candidates like novel quinoid-based Cdc25 inhibitors, which have shown antiproliferative effects in models and are advancing toward applications, potentially entering phase I trials soon. Preclinical studies show that CDC25A inhibition sensitizes cells to and , suggesting potential for combination regimens. Allosteric inhibitors derived from computational screening remain in preclinical validation as of 2025, with potential for future clinical evaluation in hematologic malignancies. Combination therapies pairing Cdc25 inhibitors with immunotherapies or chemotherapeutics hold potential for enhanced efficacy, as evidenced by preclinical models in non-small cell lung cancer (NSCLC) from the 2020s. For instance, Cdc25 inhibition synergizes with PD-1 blockade to promote antitumor immunity by altering in NSCLC xenografts, prompting early-phase clinical exploration in ongoing basket trials for advanced solid tumors. Similarly, combinations with standard chemotherapeutics like have demonstrated sensitization of resistant cells , with translational studies supporting patient selection via Cdc25 overexpression. Key challenges in Cdc25-targeted clinical development include off-target effects on other phosphatases and kinases, leading to non-specific toxicity, as observed with early inhibitors like ARQ-501. Isoform selectivity remains a hurdle, as pan-Cdc25 inhibitors often fail to distinguish between Cdc25A, B, and C, complicating therapeutic windows in proliferative diseases. Biomarkers such as elevated Cdc25A expression in tumor tissue are emerging for patient stratification, enabling identification of responsive subsets in cancers like and , where high Cdc25 levels correlate with poor prognosis and resistance to therapy. Recent updates as of 2025 underscore renewed interest, with two preclinical-to-phase I transitions for isoform-specific Cdc25B inhibitors in models showing promise in overcoming platinum resistance, though full phase II initiation awaits further safety profiling.

References

  1. [1]
    Potential of CDC25 phosphatases in cancer research and treatment
    Jan 18, 2024 · Discovery of potent and selective Cdc25 phosphatase inhibitors via rapid assembly and in situ screening of Quinonoid-focused libraries.
  2. [2]
  3. [3]
    Therapeutic Targeting the Cell Division Cycle 25 (CDC25 ... - MDPI
    CDC25 in Cell Cycle Regulation. CDC25 phosphatases function as cell cycle regulators: they activate CDKs by dephosphorylating two residues within the ATP ...<|control11|><|separator|>
  4. [4]
    Phosphatases and Kinases Regulating CDC25 Activity in the Cell ...
    CDC25A phosphatase is a prominent stimulator of cell cycle progression and it plays a very basic role in the development of thyroid neoplasms [38]. CDC25B.
  5. [5]
    The role of CDC25C in cell cycle regulation and clinical cancer ...
    Jun 3, 2020 · As an important cell cycle regulatory protein, Cell Division Cycle 25C (CDC25C) participates in regulating G2/M progression and in mediating DNA damage repair.
  6. [6]
    A Comprehensive Overview of the Developments of Cdc25 ... - NIH
    Apr 7, 2022 · Dysidiolide 53 was the first naturally discovered Cdc25A phosphatase inhibitor (IC50 9.4 µM) with antitumor activity [61]. The γ- ...
  7. [7]
    Regulation of mitosis by cyclic accumulation of p80 cdc25 mitotic ...
    Apr 5, 1990 · In fission yeast (Schizosaccharomyces pombe), cell size at mitosis is determined by expression of the cdc25+ and nim1+ inducer genes and of the ...
  8. [8]
    A Comprehensive Overview of the Developments of Cdc25 ... - MDPI
    Cdc25 phosphatases have been considered promising targets for anticancer development due to the correlation of their overexpression with a wide variety of ...<|control11|><|separator|>
  9. [9]
    Entry - *157680 - CELL DIVISION CYCLE 25C; CDC25C - OMIM
    The cell division cycle 25 (CDC25) family of proteins are highly conserved dual specificity phosphatases that activate cyclin-dependent kinase (CDK) complexes.Missing: definition nomenclature
  10. [10]
    994 - Gene ResultCDC25B cell division cycle 25B [ (human)] - NCBI
    Sep 5, 2025 · CDC25B is a member of the CDC25 family of phosphatases. CDC25B activates the cyclin dependent kinase CDC2 by removing two phosphate groups.
  11. [11]
    995 - Gene ResultCDC25C cell division cycle 25C [ (human)] - NCBI
    Sep 9, 2025 · Cdc25C is located in the cytoplasm at defined dense structures, which according to immunofluorescence analysis, electron microscopy as well as ...
  12. [12]
    twine, a cdc25 homolog that functions in the male and ... - PubMed
    twine is the second homolog of the fission yeast gene cdc25 to be found in Drosophila. Both string and twine cDNAs can rescue a temperature-sensitive cdc25 ...
  13. [13]
    CDC25A (Cell division cycle 25A)
    Aug 1, 2015 · CDC25A is a highly expressed gene in a variety of human cancers including breast, esophageal, gastric, lung, thyroid, head and neck cancers and ...Missing: definition | Show results with:definition
  14. [14]
    Revisiting phosphoregulation of Cdc25C during M-phase induction
    Dec 15, 2024 · intrinsically disordered regulatory domain of Cdc25C, the sud- den ... Cdc25 evolved through four major conceptual steps. The first.
  15. [15]
    Therapeutic Targeting the Cell Division Cycle 25 (CDC25 ... - NIH
    CDC25s seem to have a role in the development of several human malignancies, including acute myeloid leukemia (AML); and CDC25 inhibition is therefore ...
  16. [16]
    Crystal structure of the catalytic domain of the human cell ... - PubMed
    The 2.3 A structure of the human Cdc25A catalytic domain reveals a small alpha/beta domain with a fold unlike previously described phosphatase structures.Missing: 2000 NMR
  17. [17]
    CDC25A - M-phase inducer phosphatase 1 - Homo sapiens (Human)
    This entry describes 2 isoforms produced by Alternative splicing. P30304-1. This isoform has been chosen as the canonical sequence. All positional information ...Missing: definition | Show results with:definition
  18. [18]
    CDC25B - M-phase inducer phosphatase 2 - Homo sapiens (Human)
    Chromosome 20. Organism-specific databases. AGR · HGNC:1726 · HGNC · HGNC:1726 ... Gene name, Length. G8JLH2, G8JLH2_HUMAN, CDC25B, 489. B4DIG0, B4DIG0_HUMAN ...
  19. [19]
    CDC25C - M-phase inducer phosphatase 3 - Homo sapiens (Human)
    Functions as a dosage-dependent inducer in mitotic control. Tyrosine protein phosphatase required for progression of the cell cycle.
  20. [20]
    Cytoplasmic Localization of Human cdc25C during Interphase ... - NIH
    cdc25C induces mitosis by activating the cdc2-cyclin B complex. The intracellular localization of cyclin B1 is regulated in a cell cycle-specific manner, ...
  21. [21]
    Identification of an essential acidic residue in Cdc25 protein ...
    The reaction mechanism of protein tyrosine phosphatases (PTPases) and dual-specificity protein phosphatases is thought to involve a catalytic aspartic acid ...Missing: no aspartate
  22. [22]
    The catalytic mechanism of Cdc25A phosphatase - PubMed - NIH
    Cdc25 phosphatases are dual specificity phosphatases that dephosphorylate and activate cyclin-dependent kinases (CDKs), thereby effecting the progression ...
  23. [23]
    Kinetic analysis of the catalytic domain of human cdc25B - PubMed
    Nov 1, 1996 · In the present work, we have analyzed the kinetic properties of the Cdc25B catalytic domain using the artificial substrate 3-O-methylfluorescein phosphate ( ...Missing: parameters CDK
  24. [24]
    Crystal structure of the catalytic subunit of Cdc25B required for G2/M ...
    Oct 29, 1999 · The overall folding and structure of the domain is similar to that found for Cdc25A. An important difference between the two is that the Cdc25B ...Missing: 2001 | Show results with:2001
  25. [25]
    Cdc25 Phosphatases Are Required for Timely Assembly of CDK1 ...
    Our data suggest that complex assembly and dephosphorylation of Cdk1 at G 2 /M is tightly coupled and regulated by Cdc25 phosphatases.
  26. [26]
  27. [27]
    A small CDC25 dual-specificity tyrosine-phosphatase isoform in ...
    Sep 7, 2004 · We identify a CDC25-related protein (Arath;CDC25) of A. thaliana, constituted by a sole catalytic domain. The protein has a tyrosine-phosphatase ...
  28. [28]
    Specificity of Natural and Artificial Substrates for Human Cdc25A
    Phospho-peptide substrates exhibit extremely low second-order rate constants and a flat specificity profile toward Cdc25A and dN25A (kcat/Km = 1 to 10 M−1 s−1).
  29. [29]
    Specificity of natural and artificial substrates for human Cdc25A
    Feb 1, 2001 · Phospho-peptide substrates exhibit extremely low second-order rate constants and a flat specificity profile toward Cdc25A and dN25A (k(cat)/K(m) ...
  30. [30]
    CDC25A phosphatase controls meiosis I progression in mouse ...
    May 1, 2008 · CDC25A has many non-CDK substrates, including components of the MAPK pathway (Kar et al., 2002, Nemoto et al., 2004, Wang et al., 2005, Xia ...
  31. [31]
    Regulating mammalian checkpoints through Cdc25 inactivation
    This review focuses on our understanding of the biochemical mechanisms that specifically inactivate Cdc25 (cell division cycle 25) phosphatases to achieve this.
  32. [32]
  33. [33]
    SCFβ-TRCP links Chk1 signaling to degradation of the Cdc25A ...
    SCF β-TRCP promotes Chk1-dependent Cdc25A ubiquitination in vitro, and this involves serine 76, a known Chk1 phosphorylation site.
  34. [34]
    Acetylation and deacetylation of Cdc25A constitutes a novel ...
    We have characterized its acetylation and deacetylation and its biological effects on Cdc25A stability and function. When acetylated, its half-life ...
  35. [35]
    ATM/ATR checkpoint activation downregulates CDC25C to prevent ...
    Jul 27, 2012 · CHK1/CHK2-dependent CDC25C phosphorylation at Ser216 destabilizes CDC25C by promoting its nuclear export and targeting it to the proteasome.
  36. [36]
    Multisite phosphoregulation of Cdc25 activity refines the mitotic ...
    Activated Cdk1 phosphorylates Cdc25 and other substrates, further activating Cdc25 to form a positive feedback loop that drives the abrupt G2/mitosis switch.
  37. [37]
  38. [38]
    Cdc25A Localisation and Shuttling - PubMed
    Feb 1, 2005 · A nuclear localisation signal (NLS) is also characterised, mutation of which leads to cytoplasmic localisation of Cdc25A. Our results imply that ...
  39. [39]
    Localization of human Cdc25C is regulated both by nuclear export ...
    Apr 5, 2001 · The interactions between Cdc2 and Cdc25C are negatively regulated throughout interphase and in response to G2 checkpoint activation. This is ...Missing: SUMOylation | Show results with:SUMOylation
  40. [40]
    Binding of 14-3-3 proteins and nuclear export control the ...
    In Xenopus Cdc25C, the binding site for 14-3-3 around. Ser-287 is immediately adjacent to a putative bipartite nuclear localization sequence (NLS) at residues ...
  41. [41]
    Characterisation of Cdc25B localisation and nuclear export during ...
    Oct 1, 2004 · Cdc25B starts to appear in S-phase cells and accumulates until prophase, after which the protein disappears.Missing: localization | Show results with:localization
  42. [42]
    Cytoplasmic localization of human cdc25C during interphase ...
    These results suggest that 14-3-3 proteins may negatively regulate cdc25C function by sequestering cdc25C in the cytoplasm. Publication types. Research Support, ...
  43. [43]
    Plk1 promotes nuclear translocation of human Cdc25C ... - PubMed
    These results suggest that Plk1 phosphorylates Cdc25C on Ser198 and regulates nuclear translocation of Cdc25C during prophase.Missing: activation | Show results with:activation
  44. [44]
    Binding of 14-3-3 proteins and nuclear export control the ... - NIH
    In Xenopus Cdc25C, the binding site for 14-3-3 around Ser-287 is immediately adjacent to a putative bipartite nuclear localization sequence (NLS) at residues ...
  45. [45]
    Cryo-EM structure of the CDK2-cyclin A-CDC25A complex - PMC
    This structure reveals the overall binding mode of CDC25A with CDK2-cyclin A, detailing interactions between the CDC25A catalytic domain and the N- and C- ...
  46. [46]
    Phosphorylation at serine 75 is required for UV-mediated ... - PubMed
    The human Cdc25A phosphatase plays a pivotal role at the G1/S transition by activating cyclin E and A/Cdk2 complexes through dephosphorylation.
  47. [47]
    Cdc25A phosphatase: a key cell cycle protein that regulates neuron ...
    Mar 23, 2017 · Cdc25A is required for activation of the apoptotic cell cycle pathway and neuron death in response to nerve growth factor (NGF) deprivation and Aβ treatment.
  48. [48]
    Crystal Structure of the Catalytic Domain of the Human Cell Cycle ...
    The human Cdc25s are 470–530 residues in length and are composed of two domains: a variable N-terminal region, suspected to serve regulatory functions (Peng et ...
  49. [49]
    Human Cdc25 A inactivation in response to S phase inhibition and ...
    The Cdc25 A phosphatase is required for the G1-S transition of the cell cycle and is overexpressed in human cancers. We found that it is ubiquitylated and ...
  50. [50]
    A novel CDC25A/DYRK2 regulatory switch modulates cell cycle and ...
    Aug 6, 2021 · The cell division cycle 25A (CDC25A) phosphatase is a key regulator of cell cycle progression that acts on the phosphorylation status of ...
  51. [51]
    Pathways governing G1/S transition and their response to DNA ...
    Feb 16, 2001 · DNA damage induced kinase cascades involving ATR/ATM and Chk1/Chk2 target Cdc25A for rapid ubiquitin/proteasome-mediated proteolysis. This ...
  52. [52]
    G1 Phase Cell Cycle Checkpoint - an overview | ScienceDirect Topics
    If DNA damage occurs prior to or in the S phase, Chk2 phosphorylates Cdc25A and targets it for degradation, while G2/M transition is regulated by Chk1 and ...<|separator|>
  53. [53]
    Ectopic expression of Cdc25A accelerates the G(1)/S transition and ...
    Ectopic expression of Cdc25A accelerates the G(1)/S-phase transition, indicating that Cdc25A controls an event(s) that is rate limiting for entry into S phase.
  54. [54]
    Redundant Regulation of Cdk1 Tyrosine Dephosphorylation in ...
    Mutation of cdc25 in Schizosaccharomyces pombe blocks Cdk1 dephosphorylation and causes cell cycle arrest. In contrast, deletion of MIH1, the cdc25 homolog in ...
  55. [55]
    The Chk1-mediated S-phase Checkpoint Targets Initiation Factor ...
    Cdc25A and Cdk2 Are Not Targets of the BPDE-induced S-phase Checkpoint—Chk1 mediates the S-phase checkpoint induced by low doses of BPDE (<100 nm) (4, 9). The ...
  56. [56]
    Human homolog of fission yeast cdc25 mitotic inducer is ...
    This human gene encodes a protein with a predicted molecular mass of 53,000 daltons whose C-terminal domain shares about 37% sequence identity with the fission ...Missing: percentage | Show results with:percentage
  57. [57]
    Universal control mechanism regulating onset of M-phase - Nature
    ### Summary: Universal Control Mechanism for Cdc25 and Cell Cycle Conservation Across Species
  58. [58]
    PomBase - protein coding gene - cdc25 (SPAC24H6.05) - PomBase
    Cdc25 is a protein tyrosine phosphatase (M phase inducer) that modulates the probability of entering mitosis and has phosphoprotein phosphatase activity.
  59. [59]
    MIH1 | SGD - Saccharomyces Genome Database
    MIH1 is a protein tyrosine phosphatase involved in cell cycle control, specifically the G2/M transition, and protein transport. It regulates Cdc28p and Vps26p.
  60. [60]
    Complementation of the Mitotic Activator, p80 cdc25 , by a Human ...
    In this study, it is shown that dephosphorylation of Tyr15 triggered activation of the pp34-cyclin complex from fission yeast, that a human protein-tyrosine ...
  61. [61]
    The rhodanese/Cdc25 phosphatase superfamily - EMBO Press
    The catalysis occurs via a double displacement mechanism involving the transient formation of a persulfide‐containing intermediate (Rhod‐S), in which the ...
  62. [62]
    cdc25 - M-phase inducer phosphatase | UniProtKB - UniProt
    Tyrosine protein phosphatase which functions as a dosage-dependent inducer of mitotic and meiotic progression.
  63. [63]
    PomBase - protein coding gene - cdc25 (SPAC24H6.05) - PomBase
    ... identified through quantitative phosphoproteomics of fission yeast. Koch A ... gene in fission yeast. Grenon M et al. Mol Gen Genet 1999 Jan;260(6):522 ...
  64. [64]
    MIH1 - M-phase inducer phosphatase | UniProtKB - UniProt
    Function. function. Terminates the cell cycle delay. Reverses the CDC28 phosphorylation catalyzed by SWE1. Catalytic activity.
  65. [65]
    The Drosophila cdc25 homolog twine is required for meiosis
    Oct 1, 1992 · We have identified a second Drosophila cdc25 homolog. Expression of this second homolog, twine, was only detected in the germ line starting at ...Missing: germline | Show results with:germline
  66. [66]
    cdc-25.2, a C. elegans ortholog of cdc25, is required to promote ...
    Mar 15, 2010 · Previous phylogenetic analysis suggests that the four C. elegans CDC25 proteins are more closely related to each other than they are to those ...
  67. [67]
    CDC-25.2, a C. elegans ortholog of cdc25, is essential for the ... - PMC
    CDC-25.2 regulates intestinal cell divisions and binucleations by counteracting WEE-1.3 and by activating the CDK-1/CYB-1 complex.
  68. [68]
    CDC-25.1 stability is regulated by distinct domains to restrict cell ...
    Apr 1, 2008 · Cdc25 phosphatases are key positive cell cycle regulators that coordinate cell divisions with growth and morphogenesis in many organisms.Missing: isoforms | Show results with:isoforms
  69. [69]
    Multiple isoforms of CDC25 oppose ATM activity to maintain cell ...
    Unlike mammals, zebrafish express a single canonical CDC25, designated cdc25a, during embryogenesis (19). In zebrafish embryos, overexpression of cdc25a drives ...
  70. [70]
    Cdc25 and Wee1: analogous opposites? | Cell Division | Full Text
    May 4, 2007 · This mutant form of Cdc25 was considerably less potent than wild-type Cdc25 in inducing entry into M-phase in Xenopus oocytes [21]. These ...<|control11|><|separator|>
  71. [71]
    Chk1, but not Chk2, inhibits Cdc25 phosphatases by ... - EMBO Press
    Thus, inhibition of interactions with Cdk–cyclin complexes may be a general mechanism by which Chk1 (but not Chk2) inhibits virtually all Cdc25 phosphatases.Missing: aspartate | Show results with:aspartate
  72. [72]
    Cell cycle controls stress response and longevity in C. elegans | Aging
    Sep 25, 2016 · Intriguingly, knockdown of cdc-25 in adult C. elegans has been shown to promote stress tolerance and longevity [29].
  73. [73]
    Response of small intestinal epithelial cells to acute disruption of ...
    Mar 24, 2009 · At E6.5, 12 resorbed deciduas and 1 phenotypically normal Cdc25A null embryo were found (data not shown). Apoptosis was not observed in this ...Results · Cdc25a Is Essential For... · Cdc25a Is Disrupted In Adult...
  74. [74]
    In vivo roles of CDC25 phosphatases: Biological insight into the anti ...
    A number of studies indicated that CDC25 phosphatases are critical substrates of Chk1 and Chk2.
  75. [75]
    Pharmacophore-guided discovery of CDC25 inhibitors causing cell ...
    Feb 4, 2019 · CDC25 phosphatases play a key role in cell cycle transitions and are important targets for cancer therapy. Here, we set out to discover ...
  76. [76]
    Assaying Cdc25 Phosphatase Activity - Springer Nature Experiments
    To determine the activity of all human Cdc25 phosphatases, two different methods are described. For assaying phosphatase activities of recombinant Cdc25 ...
  77. [77]
    Cdc25B cooperates with Cdc25A to induce mitosis but has a ... - NIH
    The original discovery of Cdc25 in yeast led to the identification of three mammalian Cdc25 isoforms: A, B, and C. Subsequently, all three isoforms were ...
  78. [78]
    Small molecule tools for functional interrogation of protein tyrosine ...
    Jul 20, 2012 · High-throughput screening of the NCI's chemical substances against Cdc25B led to the identification of compound 38 (NSC 95397; Fig. 12), which ...
  79. [79]
    Regulation of Cdc2/Cyclin B Activation in Xenopus Egg Extracts via ...
    Activation of Cdc2/cyclin B kinase and entry into mitosis requires dephosphorylation of inhibitory sites on Cdc2 by Cdc25 phosphatase.Peptide Synthesis And... · Xenopus Egg Extracts · Results
  80. [80]
    Overexpression of cdc25A and cdc25B is frequent in primary non ...
    Sep 15, 1998 · cdc25A was overexpressed in 60% (24 of 40) of the tumors and cdc25B in 45% (18 of 40) of the tumors, whereas cdc25C was not overexpressed in any ...Missing: review | Show results with:review
  81. [81]
    CDC25 phosphatases in cancer cells: key players? Good targets?
    Aug 6, 2025 · We will focus on the roles of CDC25 phosphatases in both normal and abnormal cell proliferation, provide a critical assessment of the current ...
  82. [82]
    Overexpression of CDC25B Phosphatase as a Novel Marker of Poor ...
    On the other hand, CDC25A mRNA was overexpressed in 9 of 10 colorectal cancer cases, and immunohistochemistry for CDC25A showed high expression in 52 of 111 ...
  83. [83]
    Cdc25A Regulates Matrix Metalloprotease 1 through Foxo1 ... - NIH
    Cdc25A is a cell cycle-activating phosphatase, and its overexpression in breast cancers has been shown to correlate with poor prognosis.
  84. [84]
    Overexpression of Cyclin E1 or Cdc25A leads to replication stress ...
    Oct 7, 2020 · Cyclin E1 or Cdc25A-induced replication stress leads to mitotic segregation defects and genomic instability.
  85. [85]
    CDC25B induces cellular senescence and correlates with tumor ...
    With its role in checkpoint control, Cdc25B overexpression might contribute to tumorigenic pathway by rapidly pushing cells into mitosis with incompletely ...
  86. [86]
    CDC25A inhibition sensitizes melanoma cells to doxorubicin ... - PMC
    Apr 11, 2025 · In most cancers, diverse genetic mutations, and epigenetic alterations of CDC25 family were observed. Furthermore, we selected melanoma as a ...
  87. [87]
    Dual regulation of Cdc25A by Chk1 and p53-ATF3 in DNA ... - PubMed
    Feb 13, 2009 · We show that activation of tumor suppressor p53 by DNA damage results in inhibition of Cdc25A transcription as a result of activation of transcriptional ...Missing: knockout | Show results with:knockout
  88. [88]
    Targeting mutant p53 for cancer therapy: direct and indirect strategies
    Sep 28, 2021 · Tumors lacking p53 are particularly susceptible to G2 checkpoint abrogation when exposed to DNA damage. ... cancer cells with disrupted p53 ...Targeting P53 For Cancer... · Making Mutant P53 Functional... · Mutp53 And Micrornas
  89. [89]
    Cell division cycle 25 C (CDC25C) mediates cell-cycle progression ...
    Sep 29, 2025 · CDC25C, a phosphatase that activates the CDK1–Cyclin B1 complex at the G₂/M checkpoint, is implicated in several cancers but its role in glioma ...
  90. [90]
    Silencing CDC25A inhibits the proliferation of liver cancer cells ... - NIH
    Silencing CDC25A significantly inhibited the proliferation of liver cancer cells in vitro and in vivo, potentially via an interaction with IL-6.Missing: conditional knockout
  91. [91]
    Oncogenic role of ALX3 in cervical cancer cells through KDM2B ...
    Jul 16, 2021 · This study suggested that the ALX3 increased CDC25A expression through KDM2B-mediated demethylation of H3K4me3, which induced proliferation and cell cycle ...Missing: hypomethylation | Show results with:hypomethylation
  92. [92]
    YBX1 regulates tumor growth via CDC25a pathway in human lung ...
    Dec 13, 2016 · These results demonstrated that YBX1 activate the growth of the xenografted human lung adenocarcinoma partly through CDC25a pathway in vivo.
  93. [93]
    Microcephalin and pericentrin regulate mitotic entry via centrosome ...
    During unperturbed cell cycles, activation of cyclin B–Cdk1 at the centrosome is mediated by Cdc25B (De Souza et al., 2000; Krämer et al., 2004), which itself ...
  94. [94]
    CDC25B - DISEASES - JensenLab
    Primary microcephaly, Seckel syndrome, and microcephalic osteodysplastic primordial dwarfism type II (MOPD II) are disorders exhibiting marked microcephaly ...
  95. [95]
    Role and regulation of Cdc25A phosphatase in neuron death ...
    Dec 12, 2016 · Thus these experiments indicate that Cdc25A is upstream of and required for activation of cell-cycle-related pathways involved in neuronal death ...Missing: hyperphosphorylation | Show results with:hyperphosphorylation
  96. [96]
    Small-Molecule Cdc25A Inhibitors Protect Neuronal Cells from ...
    Apr 5, 2023 · Previously, we have found that Cdc25A is activated in models of neurodegenerative diseases, including AD and PD. In the present study, we have ...
  97. [97]
    Loss of Adult Cardiac Myocyte GSK-3 Leads to Mitotic Catastrophe ...
    Mar 14, 2016 · Results indicate increased protein expression of cyclin-dependent kinase 1 (CDK1), Cyclin B1, and CDC25C pSer-216 in DKO hearts compared with ...
  98. [98]
    Human Papillomavirus Type 16 E7 Maintains Elevated Levels of the ...
    Cdc25A stability is controlled by the ubiquitin-proteasome pathway during cell cycle progression and terminal differentiation. Oncogene 19:2447–2454. [DOI] ...
  99. [99]
    Metabolic profiling of human CD4+ cells following treatment with ...
    The autoimmune process in rheumatoid arthritis depends on activation of immune cells, which utilize intracellular kinases to respond to external stimuli ...
  100. [100]
    MicroRNA‑34a‑3p inhibits proliferation of rheumatoid arthritis ...
    Jul 23, 2019 · In the event of RA, abnormal activation of macrophages can promote FLS to produce pro-inflammatory cytokines and matrix metalloproteinase (MMPs) ...
  101. [101]
    Is Cdc25 a Druggable Target? - PMC - NIH
    While the discovery of the first Cdc25 inhibitors dates back to 1994 – a class of relatively unselective alkaloid natural products, the dnacins [31] – the ...
  102. [102]
    Medicinal chemistry insights into novel CDC25 inhibitors
    Sep 1, 2020 · This review is mainly focused on the clinical applications and synthetic methods of 34 small-molecule drugs newly approved by the FDA in 2020.
  103. [103]
    Inhibition of human tumor cell growth in vivo by an orally ... - PubMed
    BN82685 inhibits recombinant CDC25A, B, and C phosphatases in vitro. It inhibits the growth of human tumor cell lines with an IC(50) in the submicromolar range.
  104. [104]
    A phase II study of ARQ 501 in patients with advanced squamous ...
    Conclusions: ARQ 501 is well tolerated in patients with advanced, recurrent or persistent squamous cell carcinoma of the head and neck. Enrollment is ongoing ...
  105. [105]
    A novel quinoid-based CDC25 phosphatase inhibitor against ...
    Cell division cycle 25 (CDC25) phosphatase is a key cell cycle regulator whose overexpression is linked to tumor malignancy and poor prognosis.<|control11|><|separator|>
  106. [106]
    CDC25A inhibition sensitizes melanoma cells to doxorubicin and ...
    Apr 11, 2025 · Overexpression of cdc25A and cdc25B is frequent in primary non-small cell lung cancer but is not associated with overexpression of c-myc.
  107. [107]
    CDC25 Inhibition in Acute Myeloid Leukemia–A Study of Patient ...
    To conclude, the antiproliferative effects of CDC25 inhibition differ among patients and patient subsets, identified based on genetic abnormalities and AML cell ...Cdc25 Inhibition In Acute... · 2. Results · 2.1. Cdc25 Inhibition Has...
  108. [108]
    Potential of CDC25 phosphatases in cancer research and treatment
    Jan 19, 2024 · This comprehensive review explores the confluence of precision medicine and CDC25 phosphatases in the context of cancer research.