Binds CDK2 to sequester its monomeric form, targeting it for proteasomal degradation and inhibiting G1/S phase transition .
Interacts with DNA polymerase alpha/primase to block DNA replication initiation during S phase .
Modulates Retinoblastoma (Rb) protein and TGF-β signaling to suppress tumor growth .
Tumor-Suppressive Activity:
Oncogenic Potential:
In HCC, CDK2AP1 expression correlates with immune infiltration (B cells, CD4+/CD8+ T cells, macrophages) and checkpoint gene expression (e.g., PD-1, CTLA-4) .
Associates with C1 immune subtype and iCluster1 molecular subtype in HCC, suggesting a role in immunotherapy response .
Cancer Type | Expression Trend | Correlation with Survival |
---|---|---|
Hepatocellular Carcinoma | Upregulated | Poor prognosis (AUC 0.72 at 1 year) |
Kidney Chromophobe (KICH) | Downregulated | Not reported |
Glioblastoma (GBM) | Upregulated | Shorter progression-free survival |
Immunotherapy: CDK2AP1’s association with immune checkpoints (e.g., PD-L1) positions it as a biomarker for immune checkpoint inhibitor response .
Chemosensitization: Overexpression enhances cisplatin and paclitaxel efficacy in lung cancer .
Context-Dependent Roles:
Stem Cell Biology:
Mechanistic Studies: Clarify CDK2AP1’s role in Rb phosphorylation and TGF-β pathway crosstalk.
Clinical Trials: Validate its utility as a biomarker for immunotherapy in HCC and other CDK2AP1-overexpressing cancers.
Targeted Therapies: Develop small-molecule inhibitors or gene-editing approaches to modulate CDK2AP1 activity in context-specific malignancies.
CDK2AP1 (also known as DOC-1 or deleted in oral cancer-1) is a highly conserved, ubiquitously expressed gene located on chromosome 12q24 in humans. It encodes a 115-amino acid nuclear polypeptide with a molecular weight of 12.4 kDa and an isoelectric point of 9.62. The human CDK2AP1 cDNA is approximately 1.6 kilobase pairs in length . The protein functions as a specific CDK2-associated protein that negatively regulates CDK2 activity, which is critical for cell cycle progression, particularly in the transition from G1 to S phase .
CDK2AP1 exerts its cell cycle regulatory effects through multiple mechanisms:
It sequesters monomeric CDK2, preventing its activation
It targets CDK2 for proteolysis, reducing its cellular levels
It interacts with DNA polymerase alpha/primase and mediates the phosphorylation of the large p180 subunit, suggesting a regulatory role in DNA replication during S phase
It functions in the TGF-β signaling pathway, where SMAD induced by TGF-β1 binds at the proximal promoter of the CDK2AP1 gene
These mechanisms collectively contribute to CDK2AP1's role as a negative regulator of cell proliferation in normal cells.
CDK2AP1 has been shown to interact with several key proteins:
Direct interaction with Cyclin-dependent kinase 2 (CDK2)
Interaction with DNA polymerase alpha/primase
Interaction with p53, suggesting involvement in p53-mediated tumor suppression pathways
Possible interaction with an unnamed protein product (BC006130) which may mediate CDK2AP1's inhibitory effect on cell proliferation
These interactions position CDK2AP1 at a critical junction in cell cycle regulation and DNA replication control.
CDK2AP1 expression shows remarkable variability across different cancers:
Cancer Type | CDK2AP1 Expression Pattern | Clinical Correlation |
---|---|---|
Oral/Head and Neck | Downregulated in ~70% of cases | Correlates with disease progression |
Prostate Cancer | High expression in metastatic disease | Associated with poor survival |
Lung Cancer | Generally downregulated | Acts as a growth suppressor |
Breast Cancer | Decreased expression | 39-fold higher in normal vs. tumor tissue |
Glioma | Increased expression | Knockdown reduces proliferation |
This complex pattern suggests context-dependent roles of CDK2AP1 in different cancer types .
The relationship between CDK2AP1 and TGF-β signaling is particularly important in cancer progression. Studies have revealed that:
TGF-β1 treatment increases CDK2AP1 mRNA and protein levels in normal human keratinocytes
SMAD proteins induced by TGF-β1 bind at the proximal promoter of the CDK2AP1 gene
A significant correlative expression exists between TGF-β receptor II (TGFβRII) and CDK2AP1 in human oral squamous cell carcinoma (OSCC) tissues
There is a corresponding decrease in TGFβRII, CDK2AP1, SMAD2, and pSMAD2/3 in human OSCC cell lines
OSCC lines resistant to TGF-β1 fail to induce pSMAD2/3 and expression of CDK2AP1
These findings suggest that disruption of the TGF-β/CDK2AP1 axis may contribute to cancer progression, particularly in oral cancers.
Research has revealed seemingly contradictory roles for CDK2AP1 in different cancer contexts:
In oral, breast, and lung cancers, CDK2AP1 appears to function as a tumor suppressor with decreased expression associated with cancer development
Conversely, in glioma, CDK2AP1 appears to promote tumorigenesis, with CDK2AP1 knockdown reducing cell proliferation and tumor growth
In prostate cancer, the relationship is complex: while initial studies suggested a tumor-suppressive role, more recent research indicates that high CDK2AP1 expression is associated with higher Gleason grade and poor survival
To reconcile these findings, researchers should:
Consider tissue-specific molecular contexts and pathways
Examine tumor microenvironment influences
Investigate post-translational modifications of CDK2AP1
Analyze different isoforms or splice variants
Design experiments that assess both gain and loss of function in the same cancer model
To study CDK2AP1 protein-protein interactions effectively:
Co-immunoprecipitation (Co-IP): This has successfully identified interactions between CDK2AP1 and key proteins like CDK2 and p53. Use appropriate antibodies with proper controls to avoid false positives .
Molecular docking: Computational approaches can predict and visualize potential interaction sites between CDK2AP1 and other proteins, guiding experimental validation .
Proximity ligation assay (PLA): This technique provides visualization of protein interactions with subcellular resolution, ideal for verifying interactions in specific cellular compartments.
FRET or BRET assays: These approaches can detect interactions in living cells, providing temporal information about when and where CDK2AP1 interacts with partners during the cell cycle.
Yeast two-hybrid screening: While less physiological, this can identify novel interaction partners that may have been overlooked.
Mass spectrometry-based interaction proteomics: This unbiased approach can identify the complete interactome of CDK2AP1 in different cellular contexts.
A comprehensive experimental design to study CDK2AP1's epigenetic functions should include:
ChIP-seq analysis: To identify genomic regions where CDK2AP1 binds, potentially in complex with other epigenetic modifiers.
RNA-seq after CDK2AP1 modulation: Compare transcriptomes after CDK2AP1 knockdown, overexpression, and in control cells to identify regulated genes.
DNA methylation analysis: Use bisulfite sequencing or methylation arrays to examine how CDK2AP1 manipulation affects DNA methylation patterns, particularly at the promoters of cell cycle regulators.
Histone modification profiling: Employ ChIP-seq for various histone marks (H3K4me3, H3K27me3, H3K27ac, etc.) to determine how CDK2AP1 influences chromatin states.
Interaction studies with epigenetic modifiers: Use co-IP or proximity labeling to identify CDK2AP1 interactions with DNA methyltransferases, histone modifiers, or chromatin remodelers.
Rescue experiments: Test if CDK2AP1-mediated epigenetic effects can be reversed by inhibiting specific epigenetic pathways .
To accurately assess CDK2AP1's effects on the cell cycle:
Flow cytometry with PI staining: Quantify cell distribution across cell cycle phases after CDK2AP1 manipulation.
EdU incorporation assays: Measure S-phase entry and DNA synthesis rates.
Live-cell imaging with FUCCI system: Track individual cells through the cell cycle in real-time after CDK2AP1 modulation.
CDK2 activity assays: Directly measure CDK2 kinase activity with immunoprecipitated CDK2 from cells with altered CDK2AP1 levels.
Phospho-Rb analysis: Measure phosphorylation status of retinoblastoma protein, a key CDK2 substrate.
Western blot analysis of cyclins and CKIs: Monitor expression of cyclins A, E and CDK inhibitors (p21, p27) throughout the cell cycle.
Single-cell transcriptomics: Profile cell cycle gene expression at the single-cell level to capture heterogeneous responses to CDK2AP1 modulation .
CDK2AP1 serves as a critical competency factor in embryonic stem cell (ESC) differentiation:
Deletion of CDK2AP1 leads to early embryonic lethality, likely through altered differentiation capability of ESCs
CDK2AP1 regulates stem cell maintenance and differentiation through epigenetic mechanisms
It may help establish the balance between self-renewal and differentiation commitments
This is relevant to cancer research because:
Cancer cells and stem cells share common characteristics, particularly high proliferative potential
Understanding CDK2AP1's role in stem cell differentiation may provide insights into the mechanisms of cancer stem cells
The epigenetic regulatory functions of CDK2AP1 in stem cells may parallel similar functions in cancer cells
Therapeutic strategies targeting CDK2AP1 might affect both cancer cells and cancer stem cells differently
The molecular pathways involving CDK2AP1 show both similarities and differences between stem cells and cancer cells:
In stem cells:
CDK2AP1 is involved in maintaining the pluripotency network
It participates in epigenetic regulation critical for differentiation
It may help coordinate cell cycle progression with differentiation signals
In cancer cells:
CDK2AP1 interacts with the TGF-β signaling pathway
It can influence androgen receptor signaling in prostate cancer cells
It appears to interact with p53, potentially affecting apoptotic responses
Common pathways include:
Cell cycle regulation through CDK2
Epigenetic modification of key target genes
Potential intersection with major developmental signaling pathways
Future research should focus on comparative pathway analysis to better understand context-specific functions.
The dual role of CDK2AP1 as both tumor suppressor and oncogene requires sophisticated research approaches:
Tissue-specific cofactor analysis: Identify tissue-specific interaction partners that may convert CDK2AP1 from suppressor to promoter.
Post-translational modification profiling: Determine if CDK2AP1 undergoes different modifications in different cancer contexts.
Isoform-specific functions: Investigate whether different isoforms or splice variants predominate in different tissues.
Pathway context: Analyze the status of TGF-β, p53, and other key signaling pathways that interact with CDK2AP1 in each cancer type.
Dose-dependent effects: Examine whether CDK2AP1 exhibits threshold effects where different expression levels trigger different cellular outcomes.
Temporal dynamics: Study how CDK2AP1's function may change during cancer progression, potentially explaining its different roles in primary versus metastatic disease .
Studying CDK2AP1's epigenetic functions presents several methodological challenges:
Challenge: Distinguishing direct versus indirect epigenetic effects
Solution: Use rapid inducible systems (e.g., degron-tagged CDK2AP1) to identify immediate epigenetic changes
Challenge: Identifying relevant target genes among global epigenetic changes
Solution: Integrate multi-omics approaches (ChIP-seq, RNA-seq, ATAC-seq) to prioritize targets
Challenge: Determining how CDK2AP1 recruits or modulates epigenetic machinery
Solution: Use proximity labeling approaches (BioID, APEX) to identify transient interactions
Challenge: Separating cell cycle effects from epigenetic effects
Solution: Use synchronized cells and cell cycle inhibitors to isolate phase-specific functions
Challenge: Connecting epigenetic changes to functional outcomes
Solution: Perform targeted epigenetic editing at CDK2AP1-regulated loci to confirm causality
The interaction between CDK2AP1 and p53 represents a critical area for advanced cancer research:
Research implications include:
Potential impact on p53-dependent therapeutic responses
Possibility that CDK2AP1 status could predict efficacy of p53-targeted therapies
Need to evaluate CDK2AP1 status when considering therapies that rely on functional p53 pathways
Future studies should systematically investigate how CDK2AP1-p53 interaction affects response to various cancer therapeutics, particularly in cancers where p53 mutations are common.
Developing CDK2AP1-targeted therapies presents both opportunities and challenges:
Potential therapeutic approaches:
Small molecule modulators of CDK2AP1-CDK2 interaction
Epigenetic drugs that target CDK2AP1-regulated pathways
Synthetic lethality approaches in tumors with altered CDK2AP1 function
Gene therapy to restore CDK2AP1 expression in cancers where it's downregulated
Patient populations most likely to benefit:
Oral squamous cell carcinoma patients with decreased CDK2AP1 expression
Prostate cancer patients with specific molecular subtypes showing high CDK2AP1 expression
Patients with cancers showing disruption of the TGF-β/CDK2AP1 axis
Given the context-dependent nature of CDK2AP1 function, comprehensive biomarker strategies will be essential for patient selection .
For studying CDK2AP1 in patient-derived samples, researchers should consider:
Multiplex immunohistochemistry: Allows simultaneous detection of CDK2AP1 along with interacting partners and pathway components within the tissue architecture.
Single-cell RNA sequencing: Provides insights into heterogeneity of CDK2AP1 expression within tumors and correlation with cell states.
Spatial transcriptomics: Maps CDK2AP1 expression patterns in relation to tumor microenvironment features.
Patient-derived organoids: Enables functional studies of CDK2AP1 manipulation in systems that better recapitulate patient tumor biology.
Digital spatial profiling: Combines protein and RNA analysis with spatial information to understand CDK2AP1 in its tissue context.
Liquid biopsy approaches: Explores whether CDK2AP1 alterations can be detected in circulating tumor DNA or circulating tumor cells .
To resolve contradictions in CDK2AP1 research across cancer types:
Multi-cancer comparative studies: Directly compare CDK2AP1 function across multiple cancer types using standardized methodologies.
Isogenic cell line panels: Create matched cell line models with CDK2AP1 manipulation across diverse cancer types.
Domain-specific mutants: Generate mutations in different functional domains to dissect context-specific activities.
Conditional expression systems: Use inducible systems to study dose-dependent and temporal effects.
In vivo models with tissue-specific manipulation: Develop genetically engineered mouse models with tissue-specific CDK2AP1 alterations.
Systematic interaction mapping: Compare CDK2AP1 interactomes across cancer types to identify context-specific partners.
Meta-analysis of existing datasets: Integrate public data across multiple cancer types while controlling for confounding variables .
Cyclin-Dependent Kinase 2 Associated Protein 1 (CDK2AP1), also known as p12DOC-1 or doc-1 (deleted in oral cancer-1), is a highly conserved and ubiquitously expressed gene located on chromosome 12q24. This gene encodes a 115-amino acid protein that plays a crucial role in regulating the cell cycle and epigenetic mechanisms.
CDK2AP1 is primarily known for its role as a negative regulator of Cyclin-Dependent Kinase 2 (CDK2). It achieves this by sequestering monomeric CDK2 and targeting it for proteolysis . This regulation is essential for maintaining proper cell cycle progression, particularly during the S-phase, where DNA replication occurs .
Additionally, CDK2AP1 interacts with DNA polymerase alpha/primase and mediates the phosphorylation of the large p180 subunit, suggesting a regulatory role in DNA replication . This protein also forms a core subunit of the nucleosome remodeling and histone deacetylation (NuRD) complex, which epigenetically regulates embryonic stem cell differentiation .
CDK2AP1 has been implicated in various malignancies, including oral cancer and prostate cancer . Its downregulation has been observed in several types of cancer, indicating its potential role as a tumor suppressor . In prostate cancer, high CDK2AP1 expression has been associated with worse overall survival and cause-specific survival .
Human recombinant CDK2AP1 is utilized in research to study its role in cell cycle regulation and its potential as a therapeutic target in cancer treatment. By understanding the mechanisms through which CDK2AP1 regulates CDK2 and other cellular processes, researchers aim to develop novel strategies for cancer therapy.