Cyclin-dependent kinase subunit 2 (CKS2) is a regulatory protein encoded by the CKS2 gene located on chromosome 9q22 . It belongs to the CKS protein family, which includes CKS1, with which it shares 81% amino acid homology . CKS2 plays a critical role in cell cycle regulation, particularly in the G2/M phase transition, and is implicated in cancer progression, prognosis, and therapeutic strategies .
CKS2 is overexpressed in multiple cancers and contributes to tumorigenesis through mechanisms such as:
Cell proliferation: Enhancing CDK1/CDK2 activity to drive S-phase entry and mitosis .
Metastasis: Promoting epithelial-to-mesenchymal transition (EMT) in gliomas and breast cancers .
Drug resistance: Reducing apoptosis induced by chemotherapy .
CKS2 serves as a biomarker for prognosis and treatment response:
Prognostic value: High CKS2 expression correlates with advanced tumor stages, lymph node metastasis, and poor survival in breast, lung, and esophageal cancers .
Therapeutic response: Silencing CKS2 enhances chemotherapy efficacy by inducing apoptosis and cell cycle arrest .
Kaplan-Meier plots from the breast cancer (BC)-GenExMiner database show:
CKS2 inhibition offers a promising strategy for cancer treatment:
Preclinical models: CKS2 knockdown in nude mice inhibits tumor growth and metastasis .
CKS2 (CDC28 protein kinase regulatory subunit 2) is a member of the cell cycle dependent protein kinase subunits family, located at chromosome 9q22 . It functions primarily as a cell cycle regulator involved in the control of cell division. In normal physiological conditions, CKS2 plays critical roles in:
Early embryonic development
Somatic cell division regulation
Cell cycle progression, particularly through the G2/M checkpoint
Meiotic cell division
CKS2 achieves these functions by binding to cyclin-dependent kinases and modulating their activity to ensure proper progression through cell cycle checkpoints. Research methodologies to study its normal function typically include gene knockout models, protein interaction studies, and cell cycle synchronization experiments combined with CKS2 expression analysis.
CKS2 expression shows significant differences between normal and cancerous tissues, with multiple regulatory mechanisms:
Transcriptional regulation: In cancer tissues, CKS2 shows significantly upregulated mRNA expression compared to normal tissues across multiple cancer types .
Epigenetic regulation: DNA methylation status of the CKS2 gene significantly impacts its expression. Cancer tissues often show hypomethylation of CKS2, leading to increased expression .
Copy number variation (CNV): While CNV amplification of CKS2 is not the predominant mechanism, increased CKS2 CNV is observed in 7.2% of glioma patients and correlates with higher CKS2 mRNA expression .
To investigate these differences, researchers should employ matched normal-tumor tissue comparisons using qRT-PCR, microarray analysis, or RNA-seq approaches. For methylation studies, bisulfite sequencing or methylation-specific PCR are recommended methodologies.
When investigating CKS2 function in cancer research, several experimental approaches have proven effective:
Gene knockdown studies: siRNA or shRNA targeting CKS2 are effective for examining its role in proliferation, invasion, and cell cycle progression . For example, in glioma cell lines (U251 and U87), CKS2 knockdown significantly inhibited cell proliferation and invasion capabilities.
Cell viability assays: MTS colorimetric assays can quantify the effects of CKS2 manipulation on cancer cell proliferation over time (24h, 48h, 72h, 96h intervals) .
Invasion assays: Transwell assays effectively demonstrate the impact of CKS2 expression on cancer cell invasiveness .
Colony formation assays: These evaluate the long-term effects of CKS2 expression on cancer cell self-renewal and proliferation .
Cell cycle analysis: Flow cytometry following CKS2 manipulation helps determine its specific effects on cell cycle checkpoints.
For meaningful results, researchers should include appropriate controls and validate findings across multiple cell lines representing the cancer type of interest.
CKS2 demonstrates significant prognostic value across multiple malignancies, with distinct patterns emerging:
For researchers investigating prognostic implications, methodological approaches should include:
Kaplan-Meier survival analysis stratified by CKS2 expression levels
Multivariate Cox regression analysis to determine independence from other prognostic factors
ROC curve analysis to evaluate predictive accuracy for survival outcomes
Integration of CKS2 expression with established clinical parameters to develop comprehensive prognostic models
Gene set enrichment analysis (GSEA) and functional studies have identified several key pathways and processes affected by CKS2 overexpression:
Cell cycle regulation: CKS2 significantly impacts E2F targets and G2M checkpoint signaling .
Epithelial-mesenchymal transition (EMT): High CKS2 expression correlates with EMT signature genes, suggesting a role in cancer invasion and metastasis .
Hypoxia response: CKS2 expression affects hypoxia-related pathways, potentially influencing tumor adaptation to low-oxygen environments .
Immune response modulation: CKS2 correlates with allograft rejection and complement pathway activation, suggesting immunomodulatory functions .
Tumor microenvironment interaction: Single-sample Gene Set Enrichment Analysis (ssGSEA) indicates that high CKS2 expression increases infiltration of specific immune cell populations, particularly Th2 cells .
Research methodologies should include transcriptome analysis following CKS2 manipulation, protein-protein interaction studies, and pathway analysis using established bioinformatic pipelines.
CKS2's potential as a diagnostic biomarker is supported by consistent findings of its differential expression between normal and malignant tissues:
In gliomas, CKS2 demonstrates high diagnostic value with area under the curve (AUC) reaching 0.941 in ROC analysis .
CKS2 expression correlates with pathological staging, IDH mutation status, and 1p/19q co-deletion in gliomas .
Multiple cancer types show significant CKS2 upregulation that correlates with malignancy grade .
For clinical implementation, researchers should consider:
Tissue-based detection: Immunohistochemistry (IHC) on formalin-fixed paraffin-embedded tissues with validated antibodies and standardized scoring systems.
mRNA-based detection: qRT-PCR using primers such as 5′-CACTACGAGTACCGGCATGTT-3′ (forward) and 5′-CATGTAATGAACCCAGCCTAGA-3′ (reverse) for CKS2 .
Combined biomarker panels: Integrating CKS2 with other established biomarkers to improve diagnostic accuracy.
Cut-off optimization: Determining optimal expression thresholds for diagnostic classification using training and validation cohorts.
Validation across multiple independent patient cohorts is essential before clinical implementation.
Research demonstrates significant interactions between CKS2 expression and tumor immune microenvironment:
Methodological approaches for investigating these relationships include:
Immune cell deconvolution methods applied to transcriptomic data
Multiplex immunofluorescence to visualize immune cell populations in relation to CKS2 expression
Correlation analysis between CKS2 expression and immune checkpoint molecules
In vitro co-culture systems with immune and cancer cells under CKS2 manipulation
These findings suggest potential for targeting CKS2 in combination with immunotherapies, though more research is needed to establish direct mechanistic relationships.
The relationship between CKS2 and therapy response is emerging as an important research area:
Studies indicate that CKS2 depletion can enhance chemotherapy-induced apoptosis in cancer cells, particularly in hepatocellular carcinoma models .
Computational prediction models using pharmacogenomic databases suggest correlations between CKS2 expression and IC50 values for various chemotherapeutic agents .
For researchers investigating this relationship, recommended approaches include:
Cell viability assays following CKS2 manipulation in combination with chemotherapeutic agents
Analysis of therapy response in patient cohorts stratified by CKS2 expression
Development of prediction models incorporating CKS2 expression to estimate chemotherapy sensitivity
Mechanistic studies examining how CKS2 affects drug resistance pathways
These investigations could potentially identify CKS2 as both a predictive biomarker and a therapeutic target to overcome chemoresistance.
When designing experiments investigating CKS2, researchers should implement several control measures:
Tissue controls: Always include matched normal tissue controls when analyzing tumor samples to establish baseline expression.
Cell line selection: Use multiple cell lines representing the cancer type of interest, ideally with varying endogenous CKS2 expression levels .
Knockdown controls: When performing CKS2 knockdown experiments, include both negative control siRNA/shRNA and validate knockdown efficiency via qRT-PCR and western blot .
Validation cohorts: Findings should be validated across independent patient cohorts and datasets (e.g., combining TCGA, GEO, and CGGA data for glioma studies) .
Expression normalization: Use established housekeeping genes such as GAPDH (5′-CCCATCACCATCTTCCAGGAG-3′ forward and 5′-GTTGTCATGGATGACCTTGGC-3′ reverse) for accurate normalization .
These controls help minimize experimental bias and strengthen the validity of research findings.
Discrepancies between mRNA and protein levels are common in cancer research and require careful methodological consideration:
Multi-level validation: Researchers should validate CKS2 expression at both mRNA level (via qRT-PCR or RNA-seq) and protein level (via IHC or western blot) within the same samples.
Post-transcriptional regulation: Investigate potential microRNA regulation of CKS2 using prediction algorithms and validation experiments.
Protein stability analysis: Assess CKS2 protein half-life and degradation pathways that might differ between cancer types.
Subcellular localization: Evaluate whether CKS2 protein localization (nuclear vs. cytoplasmic) affects functional outcomes and correlates with prognostic significance.
When publishing research, clearly report both mRNA and protein findings rather than assuming direct correlation between transcription and translation.
Based on current evidence, several translational paths show particular promise:
Prognostic biomarker development: CKS2 expression has demonstrated consistent association with survival outcomes across multiple cancer types , making it a strong candidate for inclusion in prognostic panels.
Therapeutic targeting: As a cell cycle regulator with specific overexpression in cancer tissues, CKS2 represents a potential therapeutic target with possibly reduced normal tissue toxicity.
Patient stratification: CKS2 expression might help identify patient subgroups most likely to benefit from specific treatment approaches, particularly cell cycle-targeting therapies.
Combination therapy approaches: The connections between CKS2 and tumor immunity suggest potential synergy between CKS2 inhibition and immunotherapeutic approaches.
For successful translation, researchers should focus on establishing standardized assessment methods, validating findings in prospective clinical trials, and developing specific inhibitors of CKS2 function or expression.
Several methodological advances would significantly enhance CKS2 research:
Single-cell technologies: Application of single-cell RNA sequencing to understand CKS2 expression heterogeneity within tumors and identify specific cell populations most affected by its expression.
CRISPR-based approaches: Precise genome editing to create isogenic cell lines with CKS2 variants to assess functional impacts of specific mutations or alterations.
Structural biology: Detailed protein structure studies to identify potential binding sites for therapeutic targeting.
Patient-derived organoids: Development of 3D culture models maintaining CKS2 expression patterns of original tumors for more physiologically relevant drug testing.
In vivo models: Creation of conditional CKS2 knockout or overexpression animal models to study systemic effects of CKS2 modulation.
CKS2 binds to the catalytic subunit of cyclin-dependent kinases (CDKs) and is essential for their biological function . CDKs are a family of protein kinases that regulate the cell cycle, transcription, mRNA processing, and differentiation of nerve cells. The CKS2 protein is involved in the regulation of the cell cycle, particularly in the transition from the G1 phase to the S phase and the G2 phase to the M phase .
The expression of CKS2 is tightly regulated and varies throughout the cell cycle. In HeLa cells, CKS2 mRNA is found to be expressed in different patterns, reflecting its specialized role in cell cycle regulation . The upregulation of CKS2 has been observed in various cancer types, including lower grade gliomas (LGGs), where it is associated with poor prognosis .
CKS2 has been identified as a prognostic biomarker in lower grade gliomas. Studies have shown that increased expression of CKS2 is associated with poor survival rates in LGG patients . The protein’s involvement in cell cycle regulation makes it a potential target for cancer therapy, as disrupting its function could inhibit the proliferation of cancer cells.
Research on CKS2 has provided insights into its role in cancer and other diseases. The protein’s interaction with CDKs and its regulation of the cell cycle make it a valuable target for drug development. Recombinant CKS2 protein is used in various biochemical assays to study its function and interactions with other proteins.