CDC25A Human

Cell Division Cycle 25A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CDC25A

CDC25A (Cell Division Cycle 25A) is a dual-specificity phosphatase encoded by the CDC25A gene in humans. It plays a pivotal role in cell cycle regulation by dephosphorylating cyclin-dependent kinases (CDKs), enabling their activation and progression through key cell cycle checkpoints. Overexpression of CDC25A is frequently observed in cancers and is associated with poor prognosis .

Gene Characteristics

  • Chromosomal Location: Chromosome 3 (3p21.1) .

  • Transcript Variants: Two isoforms exist due to alternative splicing .

Protein Architecture

  • Length: 524 amino acids .

  • Domains:

    • N-terminal regulatory domain: Contains phosphorylation sites, nuclear localization/export signals, and motifs for protein-protein interactions (e.g., RXL cyclin-binding motif) .

    • C-terminal catalytic domain: Features the conserved HCX5_5R phosphatase motif, enabling dual-specificity phosphatase activity .

Table 1: CDC25A Protein Overview

PropertyDetails
Molecular Weight63.2 kDa (recombinant form)
IsoformsTwo splice variants
Key MotifsRXL cyclin-binding motif (residues 11–15) , HCX5_5R phosphatase motif

Cell Cycle Regulation

CDC25A activates CDKs by removing inhibitory phosphates (Tyr15/Thr14 in CDK2 and CDK1; Tyr24 in CDK4/6), driving transitions at:

  • G1/S Phase: Activates CDK4/6-cyclin D and CDK2-cyclin E complexes .

  • S Phase: Sustains CDK2-cyclin A activity .

  • Mitosis: Promotes CDK1-cyclin B activation .

DNA Damage Response

  • Checkpoint kinases (e.g., CHK1) phosphorylate CDC25A, triggering its ubiquitination and proteasomal degradation to halt cell cycle progression .

  • Loss of CDC25A stabilizes inhibitory CDK phosphorylation, enforcing G1/S or G2/M arrest .

Oncogenic Properties

  • Overexpression: Linked to breast, esophageal, gastric, and lung cancers .

  • Prognostic Marker: High CDC25A expression correlates with tumor aggressiveness and reduced survival .

Table 2: CDC25A in Cancer Types

Cancer TypeAssociation with CDC25ASource
Breast CancerOverexpression in >50% of cases; poor prognosis
Head and Neck CancerAmplification drives proliferation
Thyroid CancerCorrelates with high-grade tumors

Transcriptional & Post-Translational Control

  • Upregulation: By oncogenes (e.g., c-Myc) and growth signals .

  • Downregulation: Via TGF-β signaling and miRNAs (e.g., miR-483-3p) .

Functional Studies

  • CDC25A Knockdown: Delays G1/S transition and reduces Rb phosphorylation .

  • CDC25A Overexpression: Bypasses DNA damage checkpoints, promoting genomic instability .

Targeting CDC25A in Cancer

  • Small-Molecule Inhibitors: Designed to block CDC25A’s phosphatase activity or disrupt CDK2-cyclin A binding .

  • miRNA-Based Strategies: miR-483-3p mimics suppress CDC25A, inducing cell cycle arrest .

Recombinant CDC25A Protein

  • Source: Escherichia coli-expressed, His-tagged (residues 1–524) .

  • Applications: Used in kinase assays and structural studies .

Product Specs

Introduction
M-phase inducer phosphatase 1 (CDC25A), a member of the CDC25 phosphatase family, plays a crucial role in cell cycle progression from the G1 to the S phase. It activates cyclin-dependent kinase CDC2 by removing two phosphate groups, thereby promoting cell cycle advancement. In response to DNA damage, CDC25A undergoes degradation, preventing cells with chromosomal abnormalities from proceeding through cell division. While recognized as an oncogene, the precise role of CDC25A in oncogenesis remains to be fully elucidated.
Description
Recombinant human CDC25A, expressed in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 63.2 kDa. It consists of 560 amino acids (1-524 a.a.) and includes a 36 amino acid His tag at the N-terminus. Purification of CDC25A is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The CDC25A solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 5mM DTT, 20% glycerol, 0.2M NaCl, and 1mM EDTA.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability during frozen storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of CDC25A is determined by SDS-PAGE analysis to be greater than 80.0%.
Synonyms
M-phase inducer phosphatase 1, Dual specificity phosphatase Cdc25A, CDC25A, CDC25A2.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMELG PEPPHRRRLL FACSPPPASQ PVVKALFGAS AAGGLSPVTN LTVTMDQLQG LGSDYEQPLE VKNNSNLQRM GSSESTDSGF CLDSPGPLDS KENLENPMRR IHSLPQKLLG CSPALKRSHS DSLDHDIFQL IDPDENKENE AFEFKKPVRP VSRGCLHSHG LQEGKDLFTQ RQNSAPARML SSNERDSSEP GNFIPLFTPQ SPVTATLSDE DDGFVDLLDG ENLKNEEETP SCMASLWTAP LVMRTTNLDN RCKLFDSPSL CSSSTRSVLK RPERSQEESP PGSTKRRKSM SGASPKESTN PEKAHETLHQ SLSLASSPKG TIENILDNDP RDLIGDFSKG YLFHTVAGKH QDLKYISPEI MASVLNGKFA NLIKEFVIID CRYPYEYEGG HIKGAVNLHM EEEVEDFLLK KPIVPTDGKR VIVVFHCEFS SERGPRMCRY VRERDRLGNE YPKLHYPELY VLKGGYKEFF MKCQSYCEPP SYRPMHHEDF KEDLKKFRTK SRTWAGEKSK REMYSRLKKL.

Q&A

What is CDC25A and what is its primary function in human cells?

CDC25A is a dual-specificity phosphatase that plays a critical role in cell cycle regulation by removing inhibitory phosphates from cyclin-dependent kinases (CDKs), particularly CDK2 and CDK1. This activation promotes cell cycle progression, especially at the G1/S transition and during S phase. CDC25A belongs to the CDC25 family of phosphatases that positively regulate CDK activity .

The protein contains a conserved C-terminal catalytic domain responsible for its phosphatase activity and an N-terminal regulatory region with multiple phosphorylation sites that control its stability and function. A key structural element is the DSG (Asp-Ser-Gly) motif containing serine residues at positions 82 and 88, which is critical for both protein degradation and protein-protein interactions .

Research methodologies to study CDC25A function typically involve genetic manipulation (knockdown/knockout), expression of wildtype or mutant constructs, phosphatase activity assays, and analysis of cell cycle progression in various cellular models.

How is CDC25A regulated in response to DNA damage?

CDC25A undergoes rapid, ubiquitin- and proteasome-dependent degradation in response to DNA damage, representing a critical checkpoint mechanism. The degradation process follows these steps:

  • DNA damage activates ATM/ATR kinases, which then activate checkpoint kinases Chk1 and Chk2

  • Chk1/Chk2 phosphorylate CDC25A at multiple sites, particularly within the DSG motif (serines 82 and 88)

  • The SCFβ-TrCP (Skp1-cullin-β-TrCP) ubiquitin ligase complex recognizes the phosphorylated DSG motif

  • CDC25A becomes polyubiquitinated and is targeted for proteasomal degradation

This rapid degradation results in persistent inhibitory phosphorylation on CDK2, preventing S-phase entry and DNA replication when DNA is damaged. This response involves activated Chk1 protein kinase but operates independently of the p53 pathway, providing an immediate, post-translational checkpoint mechanism .

Experimental evidence shows that mutation of both serines 82 and 88 to alanine renders CDC25A fully resistant to degradation following DNA damage, while individual mutations confer partial resistance. The S88F mutation, a cancer-predisposing polymorphic variant in humans, also increases CDC25A stability .

What experimental methods are used to study CDC25A degradation?

Researchers employ several methods to investigate CDC25A degradation:

  • Protein stability assays:

    • Cycloheximide chase experiments to track protein half-life by blocking new protein synthesis

    • Western blotting with CDC25A-specific antibodies at defined time intervals

    • Pulse-chase experiments with radioisotope labeling

  • DNA damage induction:

    • Chemical agents such as etoposide (topoisomerase II inhibitor)

    • Physical methods including ultraviolet light and ionizing radiation

    • Time-course analysis following damage

  • Mutation analysis:

    • Site-directed mutagenesis of key regulatory residues (e.g., S82A, S88F)

    • Expression of mutant constructs in cell lines

    • Analysis of mutant protein stability compared to wildtype

  • Ubiquitination analysis:

    • Immunoprecipitation under denaturing conditions

    • Detection of polyubiquitinated CDC25A species

    • In vitro ubiquitination assays with purified components

In published research, these approaches have demonstrated that substitution of either serine 82 or 88 to alanine individually was sufficient to confer partial resistance to degradation following etoposide treatment, while mutation of both residues rendered CDC25A fully resistant to SCFβ-TrCP-mediated degradation .

What evidence links CDC25A to hepatocellular carcinoma (HCC) progression?

Multiple lines of evidence establish CDC25A as a significant factor in HCC progression and prognosis:

  • Expression analysis:

    • Reverse transcription-PCR showed CDC25A mRNA overexpression in 69% (9/13) of HCC samples

    • Immunohistochemistry revealed high CDC25A protein expression in 56% (33/59) of HCC specimens

    • Western blot analysis confirmed elevated CDC25A protein levels in tumor tissues

  • Clinical correlations:

    • High CDC25A expression significantly associated with dedifferentiated phenotype (p=0.001)

    • CDC25A overexpression correlated with portal vein invasion (p=0.031)

    • CDC25A levels showed strong correlation with proliferating cell nuclear antigen (PCNA) labeling index (p=0.005)

  • Prognostic significance:

    • Univariate analysis showed high CDC25A expression as a significant predictor of shorter disease-free survival (p=0.004)

    • Multivariate analysis confirmed CDC25A as an independent prognostic marker (risk ratio for cancer relapse: 2.98, p=0.029)

Notably, CDC25B, another member of the CDC25 family, did not correlate with any clinicopathological features in HCC, highlighting the specific role of CDC25A in this malignancy .

CDC25 IsoformmRNA Overexpression in HCCAssociation with Clinical FeaturesPrognostic Value
CDC25A69% (9/13)Dedifferentiation, portal vein invasionIndependent marker for disease-free survival
CDC25B31% (4/13)None identifiedNot significant

How does the S88F polymorphism in CDC25A contribute to cancer risk?

The S88F (serine-to-phenylalanine at position 88) polymorphism in CDC25A represents a cancer-predisposing variant in humans through multiple mechanisms:

  • Increased protein stability:

    • The S88F mutation renders CDC25A partially resistant to degradation following DNA damage

    • The half-life of CDC25A S88F is significantly longer than wildtype protein

    • This leads to sustained CDC25A activity when it should be downregulated

  • Disrupted protein-protein interactions:

    • The S88F mutant fails to interact with ASK1 (Apoptosis Signal-regulating Kinase 1)

    • This prevents CDC25A from suppressing ASK1-mediated apoptotic pathways

    • Consequently, cells expressing CDC25A S88F show activation of downstream stress-activated kinases (SEK, MKK3, JNK) and increased apoptosis

  • Developmental consequences:

    • Homozygous S88F mutation causes early embryonic lethality in mice

    • Genotyping of embryos at different developmental stages revealed:

Age+/++/--/-Expected -/-
Pups <22 days64 (23%)213 (77%)025%
e12.52 (11%)17 (89%)025%
e3.518 (40%)24 (53%)3 (7%)25%
e2.513 (37%)20 (57%)2 (6%)25%
e1.547 (51%)46 (49%)025%

Homozygous mutant embryos showed morphological features of apoptotic cell death, with shrunken appearance and bleb-like protuberances .

Interestingly, the S88F mutant protein retains normal phosphatase activity, suggesting that the cancer-predisposing effect relates to altered protein stability and disrupted interaction networks rather than changes in enzymatic function .

How does CDC25A regulate cell cycle progression in embryonic development?

CDC25A plays a crucial role in regulating cell cycle timing in early embryonic development through a carefully balanced mechanism:

  • Embryonic cell cycle regulation:

    • CDC25A acts as a key regulator of CDK1 activity in early mouse embryos

    • CHK1 kinase restrains CDK1 activity by promoting CDC25A phosphatase degradation

    • This regulation is essential for proper timing of cell divisions in early embryogenesis

  • Experimental evidence:

    • In mouse embryos, CDC25A-EGFP fusion protein levels decrease prior to nuclear envelope breakdown

    • In CHK1 knockout embryos, CDC25A-EGFP levels remain stable, leading to premature cell division

    • Partial inhibition of CDK1 with RO3306 delays mitosis and affects CDC25A dynamics

  • Developmental consequences:

    • CDC25A knockout embryos show delayed timing of first and second mitotic divisions

    • CHK1 inhibition does not accelerate mitotic timing in CDC25A knockout embryos

    • These findings establish CDC25A as essential for regulating cell cycle progression in zygotes and early embryos

This regulatory mechanism appears to be particularly important during early embryogenesis, when cell cycles are rapid and lack robust checkpoint controls. The CHK1-CDC25A-CDK1 pathway ensures proper coordination of DNA replication and mitosis during this critical developmental period .

How does CDC25A interact with the ASK1-mediated apoptotic pathway?

CDC25A interacts with the ASK1 (Apoptosis Signal-regulating Kinase 1) pathway, revealing a function beyond its canonical role in cell cycle regulation:

  • Direct physical interaction:

    • CDC25A physically binds to ASK1 protein

    • This interaction can be detected by co-immunoprecipitation assays

    • Wildtype CDC25A co-immunoprecipitates with ASK1 from cell lysates, while the S88F mutant fails to do so

  • Functional consequences:

    • When expressed at elevated levels, CDC25A suppresses stress-induced activation of ASK1

    • This suppression inhibits downstream stress-activated protein kinases

    • Experimental evidence shows that cells transfected with wildtype CDC25A show reduced levels of activated SEK, MKK3, JNK, and caspase 3 following DNA damage

    • In contrast, cells expressing CDC25A S88F mutant show unaffected levels of these activated proteins

  • Apoptotic regulation:

    • Through ASK1 suppression, CDC25A inhibits stress-induced apoptosis

    • Cells expressing the S88F mutant undergo rapid apoptotic cell death

    • This is visualized by Annexin V staining and morphological changes consistent with apoptosis

    • Mouse embryos homozygous for the S88F mutation show features of apoptotic cell death

This interaction represents a critical link between cell cycle regulation and stress response pathways, with the DSG motif, particularly serine 88, playing a dual role in both CDC25A degradation and ASK1 interaction .

How does CDC25A function in the DNA damage checkpoint response?

CDC25A serves as a critical effector in the rapid DNA damage checkpoint response through a multi-step mechanism:

  • Checkpoint activation:

    • DNA damage (UV light, ionizing radiation) activates ATM/ATR kinases

    • These kinases activate Chk1/Chk2, which then phosphorylate CDC25A

    • Phosphorylation creates binding sites for the SCFβ-TrCP ubiquitin ligase complex

  • CDC25A degradation:

    • Ubiquitinated CDC25A undergoes rapid proteasomal degradation

    • This degradation occurs within minutes of DNA damage detection

    • The process depends on phosphorylation of the DSG motif (serines 82 and 88)

  • CDK inhibition:

    • Reduced CDC25A levels prevent removal of inhibitory phosphates from CDK2

    • Persisting inhibitory tyrosine phosphorylation on CDK2 blocks S-phase entry

    • This prevents DNA replication in the presence of damaged DNA

  • Checkpoint bypass:

    • Overexpression of CDC25A can override this checkpoint mechanism

    • This inappropriate CDK activation leads to enhanced DNA damage

    • Decreased cell survival results from checkpoint failure

Notably, this CDC25A-dependent checkpoint response operates independently of the p53 pathway, providing an immediate, post-translational mechanism for cell cycle arrest before the p53-dependent transcriptional response is activated .

The rapid degradation of CDC25A represents one of the earliest cellular responses to DNA damage, highlighting its central role in maintaining genomic integrity.

What methodological approaches can distinguish between CDC25 family members in experimental settings?

Researchers employ several specialized techniques to distinguish between CDC25A, CDC25B, and CDC25C functions:

  • Gene-specific manipulation:

    • Isoform-specific siRNA or shRNA with validated target specificity

    • CRISPR-Cas9 knockout of individual CDC25 genes

    • Selective complementation with one family member in knockout backgrounds

  • Expression analysis:

    • Quantitative RT-PCR with isoform-specific primers

    • Western blotting with validated antibodies specific to each family member

    • Immunohistochemical analysis using isoform-specific antibodies

  • Functional differentiation:

    • Cell cycle phase-specific functions (CDC25A active throughout the cell cycle, CDC25B/C primarily in G2/M)

    • Differential responses to cellular stresses

    • Substrate preference analysis using recombinant proteins

  • In vivo validation:

    • Examination of distinct phenotypes in knockout/knockdown models

    • Tissue-specific expression patterns

    • Differential association with disease states

Research on hepatocellular carcinoma illustrates the value of these approaches. Analysis of both CDC25A and CDC25B expression in the same tumor samples revealed that while CDC25A overexpression strongly correlated with tumor dedifferentiation, portal vein invasion, and poor prognosis, CDC25B expression showed no significant correlation with any clinicopathological features .

This methodological rigor is essential for establishing the specific roles of different CDC25 family members in both normal physiology and disease states.

How might targeting CDC25A be exploited for cancer therapy?

CDC25A represents a promising therapeutic target for cancer treatment through several strategic approaches:

  • Direct inhibition strategies:

    • Small molecule inhibitors targeting the catalytic pocket

    • Allosteric modulators affecting phosphatase activity

    • Compounds that disrupt critical protein-protein interactions

  • Degradation-enhancing approaches:

    • Compounds that promote phosphorylation of the DSG motif

    • Molecules that enhance SCFβ-TrCP binding to CDC25A

    • Proteolysis-targeting chimeras (PROTACs) specific for CDC25A

  • Synthetic lethality:

    • Combining CDC25A inhibition with DNA-damaging agents

    • Exploiting the finding that CDC25A overexpression bypasses DNA damage checkpoints

    • This bypass leads to enhanced DNA damage and decreased cell survival

  • Tissue-specific targeting:

    • Delivery systems targeting tumors with CDC25A overexpression

    • Hepatocellular carcinoma represents a promising candidate given the strong correlation between CDC25A overexpression and poor prognosis

  • Biomarker-guided therapy:

    • Using CDC25A expression as a predictive biomarker for response

    • Targeting tumors with specific CDC25A mutations or expression patterns

    • Monitoring treatment response through CDC25A-dependent pathways

The identified role of CDC25A as an independent prognostic marker in HCC (risk ratio for cancer relapse: 2.98) suggests that therapies targeting this phosphatase could have particular value in aggressive tumors with high CDC25A expression . Additionally, understanding the ASK1-CDC25A interaction provides potential for developing compounds that mimic the S88F mutation's effect on ASK1 binding, potentially sensitizing cancer cells to apoptosis .

Product Science Overview

Gene and Protein Information
  • Gene Symbol: CDC25A
  • Cytogenetic Location: 3p21.31
  • Genomic Coordinates (GRCh38): 3:48,157,146-48,188,417
Function and Mechanism

CDC25A is essential for the progression from the G1 phase to the S phase of the cell cycle. It achieves this by removing inhibitory phosphate groups from CDK2-cyclin E and CDK2-cyclin A complexes . Additionally, CDC25A has a role during the G2-M transition by activating CDK1-cyclin B complexes, which are thought to initiate chromosome condensation .

Regulation and Degradation

CDC25A is specifically degraded in response to DNA damage, which prevents cells with chromosomal abnormalities from progressing through cell division . This degradation is part of the DNA damage checkpoint mechanism and involves the ubiquitin-proteasome pathway . The rapid degradation of CDC25A in response to genotoxic stress helps maintain genome integrity by halting cell cycle progression and allowing time for DNA repair .

Clinical Significance

Overexpression of CDC25A has been observed in various human cancers, including breast cancer . This overexpression can bypass the cell cycle arrest mechanisms, leading to enhanced DNA damage and decreased cell survival . As a result, CDC25A is considered a potential target for cancer therapy.

Subcellular Localization

CDC25A is primarily localized to the nucleoplasm but can also be found in the Golgi apparatus . It is an intracellular protein with no predicted secreted forms .

Tissue Expression

CDC25A shows tissue-enhanced expression in bone marrow, lymphoid tissue, and testis . It is also expressed in various other tissues, including the cerebral cortex, cerebellum, and hypothalamus .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.