CDKN3 Human

Cyclin-Dependent Kinase Inhibitor 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Oncogenic Functions

  • Esophageal Squamous Cell Carcinoma (ESCC):

    • Upregulated in 68–80% of ESCC tissues and cell lines (EC-1, TE-1) .

    • Promotes proliferation, migration, and invasion via AKT pathway activation (↑p-AKT, ↑cyclin D1) .

    • Silencing CDKN3 reduces tumor growth in vitro (↓67% proliferation) and in vivo .

  • Cervical Cancer:

    • Overexpression correlates with poor survival (5-year survival: 19.2% vs. 68.2% in high vs. low expressors) .

    • siRNA-mediated knockdown decreases cell viability by 47–53% .

  • Low-Grade Glioma (LGG):

    • High CDKN3 expression linked to poor prognosis and immune microenvironment modulation (↑CD4+ T cells) .

Tumor-Suppressive Functions

  • Hepatocellular Carcinoma (HCC):

    • Reduced CDKN3 expression correlates with advanced tumor stages .

  • Chronic Myeloid Leukemia (CML):

    • Overexpression induces apoptosis by inhibiting CDK2/XIAP axis .

Mechanistic Insights and Signaling Pathways

AKT Pathway Activation:

  • CDKN3 upregulates p-AKT and cyclin D1, driving ESCC progression .

  • In vitro studies show siRNA knockdown reduces p-AKT by 60–70% .

DNA Damage Response (DDR):

  • CDKN3 interacts with RAD51, enhancing homologous recombination repair and chemoresistance in esophageal adenocarcinoma .

  • Cisplatin-resistant cells exhibit 2.5-fold higher CDKN3 expression .

Cell Cycle Dysregulation:

  • Mitotic Control: CDKN3 ensures proper CDC2 dephosphorylation (Thr-161) during mitotic exit, preventing centrosome amplification .

  • G1/S Transition: Loss of CDKN3 accelerates cell cycle progression (↑G0/G1 phase arrest) in ovarian cancer .

Clinical and Therapeutic Implications

Therapeutic Targeting:

  • siRNA Strategies: Silencing CDKN3 inhibits tumor growth in ESCC (↓50% invasion) and cervical cancer .

  • Drug Resistance: CDKN3 knockdown reverses cisplatin resistance in esophageal cancer models .

Cancer TypeCDKN3 RoleTherapeutic Target Potential
ESCCOncogenicsiRNA, AKT inhibitors
Cervical CancerOncogenicCDKN3-specific siRNAs
GlioblastomaTumor-suppressiveCDC2 pathway modulation

Contradictory Roles and Research Gaps

  • Tissue-Specific Effects: While CDKN3 acts oncogenically in ESCC and cervical cancer, it suppresses tumorigenesis in CML and HCC .

  • Mechanistic Ambiguities: The AKT pathway’s context-dependent activation and CDKN3’s interaction with non-cell cycle proteins (e.g., ARG1 in LGG) require further study .

Future Directions

  • Precision Medicine: Develop isoform-specific inhibitors targeting oncogenic CDKN3 splice variants .

  • Combination Therapies: Pair CDKN3 silencing with cisplatin or immune checkpoint inhibitors .

Product Specs

Introduction
Cyclin-dependent kinase inhibitor 3 (CDKN3) is a member of the dual specificity protein phosphatase family. It acts as a cyclin-dependent kinase inhibitor by interacting with and dephosphorylating CDK2 kinase, thereby preventing its activation. CDKN3 plays a crucial role in cell cycle regulation. Alterations in CDKN3, such as deletions, mutations, or overexpression, have been observed in various types of cancers.
Description
Recombinant human CDKN3, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 232 amino acids, with a 20 amino acid His tag at the N-terminus (1-212 a.a. of CDKN3), and has a molecular weight of 25.9 kDa. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The CDKN3 solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, 40% glycerol, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended periods, it is recommended to store the product frozen at -20°C. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
The purity of CDKN3 is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Cyclin-dependent kinase inhibitor 3, CDK2-associated dual-specificity phosphatase, Cyclin-dependent kinase interactor 1, Cyclin-dependent kinase-interacting protein 2, Kinase-associated phosphatase, KAP, CDI1, CIP2, KAP1, FLJ25787, MGC70625, CDKN3.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MKPPSSIQTS EFDSSDEEPI EDEQTPIHIS WLSLSRVNCS QFLGLCALPG CKFKDVRRNV QKDTEELKSC GIQDIFVFCT RGELSKYRVP NLLDLYQQCG IITHHHPIAD GGTPDIASCC EIMEELTTCL KNYRKTLIHC YGGLGRSCLV AACLLLYLSD TISPEQAIDS LRDLRGSGAI QTIKQYNYLH EFRDKLAAHL SSRDSQSRSV SR.

Q&A

What is the basic function of CDKN3 in human cells?

CDKN3 functions as a dual-specificity protein tyrosine phosphatase that primarily dephosphorylates CDK1/CDK2 and other proteins. It plays a crucial role in cell cycle regulation, with expression levels that fluctuate during the cell cycle, notably peaking during mitosis. While originally classified as a cyclin-dependent kinase inhibitor, recent research suggests its primary activity is as a phosphatase rather than a direct CDK inhibitor. This phosphatase activity is essential for proper cell cycle progression through regulation of CDK activity .

How does CDKN3 expression vary across normal and cancerous tissues?

CDKN3 is frequently overexpressed in multiple cancer types compared to normal tissues. In esophageal squamous cell carcinoma (ESCC), significant upregulation was observed in cancer cell lines (EC-1, EC-7, Eca-109, and TE-1) compared to normal esophageal epidermal cells (Het1A) . Similarly, elevated expression has been documented in neuroblastoma and hepatocellular carcinoma. This overexpression pattern contrasts with typical tumor suppressor genes, suggesting CDKN3 may function as an oncogene in certain contexts. The higher expression in cancer cells may be partially explained by the increased proportion of mitotic cells in rapidly dividing tumors, as CDKN3 levels naturally peak during mitosis .

What alternative splicing patterns are observed for CDKN3?

CDKN3 undergoes alternative splicing, generating multiple transcript variants. The two predominant transcripts are:

  • Full-length CDKN3 transcript encoding the complete functional protein

  • A major variant that skips exon 2, expressed in both normal and cancer cells

How does CDKN3 affect cancer patient survival?

The survival difference is particularly pronounced, as shown in the SEQC dataset where:

Similar negative correlations between CDKN3 expression and patient outcomes have been observed in hepatocellular carcinoma and other cancer types, establishing CDKN3 as a potential prognostic biomarker .

What mechanisms explain CDKN3's role in promoting cancer progression?

CDKN3 promotes cancer progression through multiple mechanisms:

  • Cell cycle regulation: CDKN3 affects cell cycle progression, particularly at the G0/G1 checkpoint. Knockdown experiments in ESCC cells demonstrated that CDKN3 silencing reduces cell proliferation and promotes G0/G1 phase arrest .

  • AKT pathway activation: In ESCC, CDKN3 activates the AKT signaling pathway, a major oncogenic driver. CDKN3 knockdown downregulates AKT pathway-related proteins, suggesting CDKN3 functions upstream of this pathway to promote cell proliferation, invasion, and migration .

  • MYCN interaction: In neuroblastoma, CDKN3 is regulated by MYCN, an oncogenic transcription factor. MYCN binds to the E-box element (CAACTG) in the CDKN3 promoter, directly regulating its expression. This creates a potential oncogenic mechanism in MYCN-amplified neuroblastomas .

  • Cell differentiation inhibition: CDKN3 knockdown promotes neuroblastoma cell differentiation, suggesting it may help maintain the undifferentiated state characteristic of aggressive neuroblastomas .

Is CDKN3 a tumor suppressor or oncogene?

  • Consistent overexpression across multiple cancer types

  • Correlation between high expression and poor patient outcomes

  • Functional studies showing promotion of proliferation, invasion, and migration

  • Rarity of CDKN3 mutations and copy number alterations in human cancers

The current consensus suggests that CDKN3 predominantly functions as an oncogene in most cancer contexts. The previous view of CDKN3 as a tumor suppressor has been questioned, with recent evidence indicating its overexpression in cancer is likely due to the high proportion of mitotic cells in rapidly growing tumors rather than dominant-negative inhibition by aberrant transcripts .

What are effective approaches for CDKN3 knockdown in experimental models?

For effective CDKN3 knockdown in experimental models, siRNA-mediated silencing has been validated in multiple studies. Specifically:

  • siRNA design: Two siRNAs targeting different regions of CDKN3 were evaluated in ESCC cell lines, with siCDKN3-1 demonstrating superior knockdown efficiency compared to siCDKN3-2 . Researchers should design multiple siRNAs and validate their efficiency.

  • Verification methods: Successful knockdown should be confirmed at both mRNA and protein levels using:

    • qPCR for transcript quantification

    • Western blot for protein expression analysis

  • Functional validation: After CDKN3 knockdown, researchers should examine:

    • Cell proliferation (using colony formation assays)

    • Cell cycle distribution (flow cytometry)

    • Migration and invasion capabilities (transwell assays)

When designing knockdown experiments, researchers should include appropriate negative controls and consider potential off-target effects by using multiple siRNA sequences or other gene silencing methods such as CRISPR-Cas9 for validation .

How can researchers effectively analyze CDKN3's impact on cell cycle regulation?

To analyze CDKN3's impact on cell cycle regulation, researchers should employ a multi-faceted approach:

  • Cell cycle distribution analysis:

    • Flow cytometry with propidium iodide staining to determine the percentage of cells in G0/G1, S, and G2/M phases

    • Compare cell cycle profiles between CDKN3-knockdown cells and controls

    • Studies have shown CDKN3 knockdown increases the percentage of cells in G0/G1 phase

  • Cell cycle protein expression:

    • Western blot analysis of cell cycle regulators (cyclins, CDKs)

    • Phosphorylation status of CDK1/CDK2 (direct targets of CDKN3 phosphatase activity)

    • Expression of checkpoint proteins (p21, p27)

  • Synchronized cell experiments:

    • Synchronize cells using methods such as double thymidine block

    • Analyze CDKN3 expression and activity across different cell cycle phases

    • Monitor the timing of cell cycle progression after synchronization

  • Live-cell imaging:

    • Real-time monitoring of cell cycle progression using fluorescent reporters

    • Measure mitotic duration and timing in CDKN3-manipulated cells

This comprehensive approach allows researchers to establish both the timing and mechanistic role of CDKN3 in cell cycle regulation, particularly its peak expression during mitosis and effects on G0/G1 transition .

What bioinformatic approaches are valuable for CDKN3 research in patient cohorts?

Several bioinformatic approaches have proven valuable for CDKN3 research in patient cohorts:

  • Survival analysis:

    • Kaplan-Meier method to assess correlation between CDKN3 expression and patient outcomes

    • Stratification of patients into high and low CDKN3 expression groups

    • The R2 Scan mode can be used to determine optimal cut-off points for grouping patients

    • Log-rank tests to determine statistical significance, with both raw and Bonferroni-adjusted p-values

  • Gene expression correlation:

    • Pearson correlation analysis to identify genes co-expressed with CDKN3

    • STRING database to identify experimentally determined CDKN3-binding proteins

    • GEPIA2 "Similar Gene Detection" module to find genes functionally related to CDKN3

  • Pathway enrichment analysis:

    • Gene Ontology (GO) analysis for biological processes, cellular components, and molecular functions

    • Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis

    • Select pathways with corrected p-value filter (qvalueFilter = 0.05) for statistical significance

  • Immune infiltration analysis:

    • TIMER 2.0 "Immune-Gene" module to explore associations between CDKN3 expression and immune cell infiltration

    • Algorithms like EPIC, MCP-COUNTER, CIBERSORT, CIBERSORT-ABS, and QUANTISEQ for estimating immune cell composition

    • Purity-adjusted Spearman's rank correlation test for statistical validation

These approaches provide a comprehensive framework for investigating CDKN3's clinical relevance, molecular interactions, and potential mechanistic pathways in cancer progression.

How does CDKN3 function in neuroblastoma pathogenesis?

CDKN3 plays a significant role in neuroblastoma pathogenesis through several mechanisms:

  • Cell differentiation inhibition: High-content screening revealed that CDKN3 knockdown promotes neuroblastoma cell differentiation, as evidenced by increased neurite outgrowth. This suggests CDKN3 normally functions to maintain the undifferentiated state characteristic of aggressive neuroblastomas .

  • MYCN-mediated regulation: In neuroblastoma, CDKN3 is directly regulated by MYCN, a critical oncogenic driver. MYCN binds to the E-box element (CAACTG) in the CDKN3 promoter. Luciferase reporter assays confirmed this direct regulation, with mutated E-box sites (ACATCT) showing reduced activity. This establishes a mechanistic link between MYCN amplification and CDKN3 expression .

  • Prognostic significance: Analysis of multiple neuroblastoma patient datasets revealed that high CDKN3 expression strongly correlates with poor prognosis. This correlation was observed across different patient subgroups, including:

    • Age groups (<18 months and >18 months)

    • Gender (male and female)

    • MYCN non-amplified tumors

    • Various tumor stages (particularly stages 2-4)

  • Differentiation therapy potential: CDKN3 appears to be downregulated by multiple differentiation-inducing molecules in neuroblastoma, suggesting it may be a convergent target for differentiation therapy approaches .

These findings collectively suggest CDKN3 functions as an oncogenic driver in neuroblastoma, with particular relevance to cell differentiation pathways and MYCN-driven tumorigenesis.

What is known about CDKN3's role in esophageal squamous cell carcinoma (ESCC)?

CDKN3 functions as an oncogenic driver in esophageal squamous cell carcinoma (ESCC) through several mechanisms:

  • Overexpression pattern: CDKN3 is significantly upregulated in ESCC cell lines (EC-1, EC-7, Eca-109, and TE-1) compared to normal esophageal epidermal cells (Het1A), as confirmed by both qPCR and western blot analyses .

  • Proliferation promotion: Knockdown of CDKN3 using siRNA significantly reduces ESCC cell proliferation, as demonstrated by colony formation assays. siCDKN3 treatment decreases both the number and size of colonies formed by ESCC cells .

  • Cell cycle regulation: CDKN3 silencing induces G0/G1 phase arrest in ESCC cells, suggesting it normally promotes cell cycle progression through the G1/S checkpoint .

  • Invasion and migration enhancement: CDKN3 knockdown significantly impairs the invasive and migratory capabilities of ESCC cells, indicating CDKN3 normally promotes these metastasis-associated phenotypes .

  • AKT pathway activation: Mechanistic studies revealed that CDKN3 promotes ESCC progression by activating the AKT signaling pathway. Western blot analysis showed that siCDKN3 treatment downregulates AKT pathway-related proteins, positioning CDKN3 as an upstream regulator of this oncogenic pathway .

This evidence establishes CDKN3 as a potential therapeutic target in ESCC, with implications for controlling both primary tumor growth and metastatic spread through modulation of the AKT pathway.

How might CDKN3 be targeted therapeutically in cancer treatment?

Therapeutic targeting of CDKN3 in cancer presents several potential strategies based on current understanding:

  • Direct enzyme inhibition: As a dual-specificity phosphatase, CDKN3 could be targeted with small molecule inhibitors designed to block its catalytic activity. This approach would require detailed structural understanding of CDKN3's active site and development of highly specific inhibitors to minimize off-target effects on other phosphatases.

  • RNA interference-based approaches: siRNA or shRNA strategies have demonstrated efficacy in experimental models. For clinical application, these could be delivered via nanoparticle formulations or other delivery vehicles to achieve tumor-specific targeting. The significant anti-tumor effects observed in CDKN3 knockdown experiments suggest this approach could be clinically valuable .

  • Disruption of protein-protein interactions: Targeting the interaction between CDKN3 and its substrates (such as CDK1/CDK2) or other binding partners could provide another therapeutic avenue. This would require detailed mapping of the interaction interfaces.

  • Combination with differentiation therapy: In neuroblastoma, CDKN3 inhibition promotes cell differentiation. This suggests potential synergy with existing differentiation agents like retinoic acid. Such combinations could enhance therapeutic efficacy while potentially reducing required dosages of individual agents .

  • Biomarker-guided therapy: Given the prognostic significance of CDKN3 expression, stratifying patients based on CDKN3 levels could identify those most likely to benefit from CDKN3-targeted therapies or alternative approaches for high-risk individuals .

While these approaches show promise, further research is needed to validate CDKN3 as a therapeutic target, particularly regarding potential toxicity given its role in normal cell cycle regulation.

What contradictory findings exist in CDKN3 research and how might they be reconciled?

Several contradictory findings exist in CDKN3 research that require careful reconciliation:

  • Tumor suppressor vs. oncogene contradiction:

    • Early classification: CDKN3 was initially classified as a putative tumor suppressor based on its ability to inhibit CDKs

    • Current evidence: Consistent overexpression in multiple cancers and correlation with poor outcomes suggest oncogenic properties

    • Reconciliation: The current consensus leans toward CDKN3 functioning primarily as an oncogene in most cancer contexts, with its overexpression in tumors likely reflecting increased mitotic cell populations rather than loss of function

  • Expression patterns in MYCN-amplified neuroblastoma:

    • General pattern: High CDKN3 correlates with poor outcomes in most neuroblastoma subgroups

    • Exception: In the MYCN-amplified group in the NRC dataset, this correlation was inconsistent

    • Reconciliation: MYCN amplification may override or alter CDKN3's prognostic significance through additional oncogenic mechanisms, or sample size limitations in subgroup analyses may affect statistical power

  • Stage-specific correlations:

    • Advanced stages: Strong correlation between high CDKN3 and poor outcomes in stages 2-4

    • Early stages: Weaker or inconsistent correlations in stage 1 and 4s neuroblastoma

    • Reconciliation: CDKN3's impact may depend on the broader molecular context of different disease stages, with additional factors potentially influencing outcomes in very early or special stage disease

  • Splice variant interpretation:

    • Earlier hypothesis: Aberrant CDKN3 transcripts were proposed to encode dominant-negative inhibitors, explaining overexpression

    • Current evidence: Aberrant transcripts occur infrequently and at lower levels than canonical forms

    • Reconciliation: The mitotic peak expression of CDKN3 provides a more plausible explanation for its overexpression in cancer than dominant-negative inhibition by rare variants

These contradictions highlight the context-dependent nature of CDKN3 function and underscore the importance of comprehensive experimental approaches that consider cell type, cancer stage, and molecular context when interpreting CDKN3's role in cancer biology.

How does CDKN3 expression correlate with patient stratification in clinical settings?

CDKN3 expression provides valuable insights for patient stratification in clinical settings, with several significant correlations observed across cancer types:

  • Neuroblastoma patient stratification:

    Analysis of three independent neuroblastoma datasets (Kocak, NRC, and SEQC) revealed:

    ParameterSubgroupSurvival ProbabilityStatistical Significance
    All patientsHigh CDKN30.35p=1.58E-34
    Low CDKN30.91
    Age <18 mosHigh CDKN30.71p=5.98E-14
    Low CDKN30.97
    Age >18 mosHigh CDKN30.23p=3.77E-11
    Low CDKN30.93
    MYCN non-amplifiedHigh CDKN30.40p=4.79E-26
    Low CDKN30.94
    Stage 3High CDKN30.00p=1.86E-15
    Low CDKN30.85
    Stage 4High CDKN30.22p=7.16E-07
    Low CDKN30.73

    This stratification demonstrates the robust prognostic value of CDKN3 across various clinical subgroups .

  • Integration with established clinical parameters:

    • CDKN3 expression provides additional prognostic information beyond traditional factors like age, MYCN status, and tumor stage

    • Particularly valuable in MYCN non-amplified cases, where other molecular markers may be lacking

    • High CDKN3 expression in patients with otherwise favorable characteristics (e.g., <18 months age) identifies a subset with unexpectedly poor outcomes

  • Immune microenvironment correlation:

    • CDKN3 expression correlates with specific immune cell infiltration patterns

    • Analysis using algorithms like EPIC, MCP-COUNTER, CIBERSORT, and QUANTISEQ reveals associations with cancer-associated fibroblasts (CAFs), regulatory T cells (Tregs), and T cell follicular helper (TFH) populations

    • These correlations provide additional context for understanding tumor biology and potential immunotherapy responses

  • Multi-cancer application:

    • Similar stratification approaches have demonstrated value in hepatocellular carcinoma and other cancer types

    • The consistent negative correlation between CDKN3 expression and patient outcomes suggests broad applicability as a prognostic biomarker

These findings establish CDKN3 expression as a clinically relevant parameter for patient stratification, with potential applications in treatment planning, monitoring, and clinical trial design.

What are the key unresolved questions regarding CDKN3 function in human cancers?

Despite significant advances in understanding CDKN3, several key questions remain unresolved:

  • Mechanistic basis of context-specific effects:

    • Why does CDKN3 appear to have different effects in different cancer types and stages?

    • What molecular cofactors determine whether CDKN3 promotes or inhibits cancer progression in specific contexts?

    • How do tissue-specific factors influence CDKN3's function and impact on clinical outcomes?

  • Phosphatase substrate specificity:

    • Beyond CDK1/CDK2, what is the complete repertoire of CDKN3 substrates?

    • How is substrate selection regulated in different cellular contexts?

    • What determines the specificity and efficiency of CDKN3-mediated dephosphorylation?

  • Regulation of CDKN3 itself:

    • What mechanisms control CDKN3's cell cycle-dependent expression pattern?

    • How is CDKN3 regulated post-translationally (phosphorylation, ubiquitination, etc.)?

    • What factors control its subcellular localization and activity?

  • Therapeutic targeting considerations:

    • Can CDKN3 be effectively targeted without disrupting normal cell cycle progression?

    • What patient populations would most benefit from CDKN3-targeted therapies?

    • What combination strategies might enhance efficacy while minimizing toxicity?

  • Prognostic vs. predictive biomarker potential:

    • Is CDKN3 expression merely prognostic or can it predict response to specific therapies?

    • How might CDKN3 status inform treatment selection in personalized medicine approaches?

    • Can measurement of CDKN3 activity (rather than just expression) provide additional clinical insights?

Addressing these questions will require integrated approaches combining structural biology, proteomics, advanced genetic models, and clinical correlations to fully elucidate CDKN3's complex roles in cancer biology and therapeutic potential.

What emerging technologies might advance CDKN3 research?

Several emerging technologies hold promise for advancing CDKN3 research:

  • CRISPR-based approaches:

    • CRISPR activation/interference (CRISPRa/CRISPRi) for precise modulation of CDKN3 expression

    • CRISPR base editing for introducing specific mutations without double-strand breaks

    • CRISPR screens to identify synthetic lethal interactions with CDKN3 in cancer contexts

    • CRISPR-mediated knock-in of reporter tags for live-cell tracking of endogenous CDKN3

  • Single-cell technologies:

    • Single-cell RNA sequencing to resolve CDKN3 expression heterogeneity within tumors

    • Single-cell proteomics to correlate CDKN3 protein levels with phosphorylation states of substrates

    • Spatial transcriptomics to map CDKN3 expression patterns within tumor microenvironments

    • Trajectory analysis to understand CDKN3's role in cancer evolution and metastasis

  • Advanced structural biology techniques:

    • Cryo-electron microscopy to resolve CDKN3-substrate complexes

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic protein interactions

    • AlphaFold and related AI approaches to predict structural interactions and design inhibitors

    • Time-resolved structural studies to capture catalytic intermediates

  • Patient-derived models:

    • Organoids from patient tumors to study CDKN3 function in near-native contexts

    • Patient-derived xenografts for in vivo validation of CDKN3-targeted therapies

    • Ex vivo drug sensitivity testing correlated with CDKN3 expression patterns

    • Humanized mouse models incorporating patient immune components

  • Multi-omics integration:

    • Integrated analysis of genomics, transcriptomics, proteomics, and phosphoproteomics data

    • Network medicine approaches to position CDKN3 within cancer-specific regulatory networks

    • Systems biology models of cell cycle incorporating CDKN3's dynamic expression and activity

    • AI/machine learning to identify patterns in large-scale multi-omics datasets

These technologies, especially when used in combination, could significantly accelerate our understanding of CDKN3's roles in cancer and facilitate development of targeted therapeutic approaches.

Product Science Overview

Introduction

Cyclin-Dependent Kinase Inhibitor 3 (CDKN3), also known as KAP (Kinase-Associated Phosphatase), is a protein encoded by the CDKN3 gene in humans. This protein plays a crucial role in cell cycle regulation by interacting with and dephosphorylating cyclin-dependent kinases (CDKs), particularly CDK2 .

Gene and Protein Structure

The CDKN3 gene is located on chromosome 14 and encodes a dual specificity protein phosphatase. This means that CDKN3 can remove phosphate groups from both tyrosine and serine/threonine residues on its substrates . The protein has several aliases, including CIP2, CDI1, and KAP1 .

Function and Mechanism

CDKN3 is primarily involved in the regulation of the cell cycle. It dephosphorylates CDK2 at the threonine-160 residue, which is essential for CDK2 activation . By doing so, CDKN3 prevents the activation of CDK2, thereby inhibiting cell cycle progression from the G1 to the S phase . This regulatory mechanism is vital for maintaining proper cell division and preventing uncontrolled cell proliferation.

Clinical Significance

CDKN3 has been implicated in various cancers, including hepatocellular carcinoma, breast cancer, and prostate cancer . The gene can be deleted, mutated, or overexpressed in these cancers, leading to dysregulation of the cell cycle and contributing to tumorigenesis . Additionally, CDKN3 has been shown to interact with other CDKs, such as CDK3 and CDC2, further highlighting its role in cell cycle control .

Research and Applications

Recombinant human CDKN3 is used in research to study its function and role in cancer. By understanding how CDKN3 interacts with CDKs and regulates the cell cycle, researchers can develop targeted therapies for cancers where CDKN3 is dysregulated . Various antibodies and assays are available for detecting and studying CDKN3 in laboratory settings .

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 2024 Thebiotek. All Rights Reserved.