CDKN2C Human refers to the human gene CDKN2C (Cyclin-Dependent Kinase Inhibitor 2C) and its encoded protein, p18<sup>INK4C</sup>. This gene is a member of the INK4 family of cyclin-dependent kinase inhibitors, which regulate cell cycle progression by binding to CDK4/6 kinases and blocking their interaction with cyclin D, thereby inducing G1-phase arrest . CDKN2C plays critical roles in tumor suppression, metabolic regulation, and viral infection pathways, making it a focal point in oncology and molecular biology research .
Protein: 168-amino acid protein (18–19 kDa) containing five ankyrin repeats for CDK4/6 binding .
Isoforms: Two splice variants (NM_001262.2 and NM_078626.2) encode identical proteins .
Interaction Partner | Functional Outcome |
---|---|
CDK4/CDK6 | Inhibition of kinase activity, G1-phase arrest |
RB1 (Retinoblastoma protein) | Synergistic tumor suppression |
CDKN2C inhibits CDK4/6-cyclin D complexes, preventing phosphorylation of RB1 and halting cell cycle progression from G1 to S phase. This checkpoint control is critical for maintaining genomic stability .
Tumor suppression: Loss-of-function mutations or deletions in CDKN2C are linked to malignancies, including gliomas, bone cancers, and endometrial tumors .
Mechanism: Dysregulation leads to uncontrolled cell proliferation and evasion of senescence .
Reduced CDKN2C expression in adipose tissue correlates with insulin resistance and type 2 diabetes (T2D). Knockdown experiments show impaired adipocyte differentiation and lipid storage .
CDKN2C overexpression enhances hepatitis B virus (HBV) replication by inducing G1 arrest, which upregulates host factors necessary for viral transcription .
CDKN2C Human Recombinant: Produced in E. coli as a 20.7 kDa His-tagged protein (PKA-020). Used in kinase inhibition assays and functional studies .
Anti-p18<sup>INK4C</sup> Antibody (AF7746): Detects human CDKN2C in ELISAs and Western blots. Specificity confirmed via CRISPR-Cas9 knockout validation .
CDK4/6 inhibitors: Drugs like palbociclib mimic CDKN2C’s function and are FDA-approved for breast cancer .
HBV therapy: Targeting CDKN2C-mediated cell cycle arrest could reduce viral replication .
Condition | Trial Phase | CDKN2C Role |
---|---|---|
Malignant glioma | Phase II/III | Biomarker for therapy response |
Hepatocellular carcinoma | Preclinical | HBV replication modulation |
Study: CDKN2C mRNA levels are inversely correlated with hyperglycemia and visceral adiposity in T2D patients. CRISPR-Cas9 knockdown in preadipocytes reduced lipid accumulation and differentiation markers (e.g., CEBPA, ADIPOQ) .
CDKN2C is a member of the INK4 family of cyclin-dependent kinase inhibitors that acts as a cell growth regulator by controlling cell cycle G1 progression. It functions by directly interacting with CDK4 or CDK6, preventing their activation and subsequent phosphorylation of the retinoblastoma (RB1) protein . This inhibition maintains RB1 in its active, hypophosphorylated state, suppressing E2F transcription factor activity and arresting cells in G1 phase. Through this mechanism, CDKN2C serves as an important tumor suppressor, as its inactivation can lead to uncontrolled cell proliferation.
CDKN2C is located on chromosome 1p32.3 and encodes a protein containing ankyrin repeats essential for CDK4/6 interaction . Two alternatively spliced transcript variants have been reported, both encoding identical proteins . The gene spans approximately 6.7 kb (chr1:51,206,196–51,212,897) . In experimental studies, researchers often analyze a broader region (approximately 223,049 bp from 51,086,606 to 51,309,654 on chromosome 1p) to fully capture the regulatory landscape of CDKN2C .
CDKN2C expression exhibits tissue-specific patterns with distinct regulatory mechanisms. In normal liver tissue, CDKN2C is consistently expressed while other family members like CDKN2A (p16) and CDKN2B (p15) are transcriptionally silenced . This suggests differential regulation of INK4 family members in various tissues. CDKN2C also plays important roles in spermatogenesis regulation , indicating specialized functions in reproductive tissues. When designing experiments, researchers must account for this tissue-specific variation by including appropriate controls and reference tissues.
CDKN2C's role in disease pathogenesis varies significantly depending on the context:
In cancer: CDKN2C functions as a haploinsufficient tumor suppressor, where loss of a single gene copy can contribute to tumorigenesis, particularly when combined with other oncogenic alterations . In medullary thyroid carcinoma (MTC), CDKN2C loss significantly correlates with more aggressive disease and poorer survival, especially when combined with RET mutations .
In viral infections: CDKN2C has been identified as an important host factor for HBV replication . It is overexpressed in highly permissive cells and HBV-infected patients, inducing cell cycle G1 arrest through CDK4/6 inhibition, which upregulates HBV transcription enhancers . This overexpression correlates with disease progression in HBV-infected patients.
This dual role—tumor suppressor in some contexts and disease promoter in others—highlights the complex biology of CDKN2C in human pathophysiology.
Several experimental models have proven valuable for studying CDKN2C:
Knockout mouse models have revealed CDKN2C's roles in regulating spermatogenesis and tumor suppression . Notably, loss of a single gene copy is sufficient to cause MTC in mice, with accelerated disease when combined with RET mutations .
Cell line models with manipulated CDKN2C expression are useful for mechanistic studies. HBV research demonstrated that a genome-wide gain-of-function screen in poorly permissive hepatoma cell lines could uncover CDKN2C's role in viral replication .
Patient-derived samples with different CDKN2C statuses provide clinically relevant insights. Studies in MTC patients showed that CDKN2C loss is associated with distant metastasis and decreased survival .
When selecting experimental models, researchers should consider the specific biological context and ensure appropriate controls to account for tissue-specific variation in CDKN2C expression.
CDKN2C functions within a complex regulatory network. Its primary interaction is with CDK4/6, preventing their association with D-type cyclins and subsequent RB1 phosphorylation . Research has revealed interesting relationships between CDKN2C and cyclin D expression patterns. In hepatoblastoma samples, a shift from cyclin D1 to cyclin D3 expression occurs during malignant transformation, while CDKN2C expression remains stable .
The table below summarizes key interactions within the CDKN2C pathway:
Interacting Protein | Functional Relationship | Biological Consequence |
---|---|---|
CDK4/CDK6 | Direct binding and inhibition | Prevents RB1 phosphorylation |
Cyclin D (D1, D2, D3) | Competitive inhibition | Blocks Cyclin D-CDK4/6 complex formation |
RB1 | Indirect regulation | Maintains RB1 in active, hypophosphorylated state |
E2F | Downstream effector | Suppresses E2F-mediated transcription |
CDKN2C's growth-suppressive function correlates with wild-type RB1 status , highlighting the importance of studying these proteins as an integrated system rather than in isolation.
Multiple complementary approaches can be used to comprehensively assess CDKN2C alterations:
Copy Number Analysis:
Mutational Analysis:
Expression Analysis:
For optimal results, researchers should:
Include paired normal tissue whenever possible
Use multiple assays to confirm findings
Integrate genomic, transcriptomic, and proteomic data
CDKN2C alterations have demonstrated significant prognostic relevance:
In Medullary Thyroid Carcinoma:
In HBV Infection:
CDKN2C overexpression correlates with disease progression in HBV-infected patients
CDKN2C enhances viral replication through cell cycle modulation
These findings highlight CDKN2C's potential as both a prognostic biomarker and therapeutic target in multiple disease contexts.
Researchers can modulate CDKN2C function through several approaches:
Overexpression Systems:
Plasmid-based transient transfection with CDKN2C expression vectors
Stable cell lines using lentiviral or retroviral vectors
Inducible expression systems (e.g., Tet-On/Off) for temporal control
Gene Silencing Approaches:
siRNA or shRNA targeting CDKN2C
CRISPR-Cas9 genome editing for complete knockout
CRISPRi for transcriptional repression
Protein Function Modulation:
Small molecule inhibitors targeting CDKN2C-CDK4/6 interactions
Peptide mimetics of functional domains
Protein degradation approaches
Validation of successful modulation should include both expression assessments and functional readouts, such as:
Changes in cell cycle distribution (G1 arrest)
CDK4/6 activity and RB1 phosphorylation status
Downstream target gene expression
In HBV research, gain-of-function screens successfully identified CDKN2C as an important host factor for viral replication , demonstrating the utility of functional genomic approaches.
CDKN2C has been implicated in longevity regulation through its involvement in the mTOR signaling pathway . A study of Dutch nonagenarians found significant association between genetic variation in mTOR pathway genes (including CDKN2C) and familial longevity . While no individual gene remained significant after multiple hypothesis testing correction, the pathway as a whole showed significant association.
This connection likely stems from CDKN2C's role in regulating cellular senescence and proliferation, which are key processes in aging. The cell cycle regulatory functions of CDKN2C may contribute to maintaining tissue homeostasis and preventing age-related pathologies such as cancer. Further research is needed to elucidate the specific mechanisms by which CDKN2C variants influence longevity.
The INK4 family (CDKN2A, CDKN2B, CDKN2C, and CDKN2D) shares similar functions in CDK4/6 inhibition, creating challenges in isolating CDKN2C-specific effects:
Functional Redundancy: All INK4 proteins inhibit CDK4/6, making it difficult to attribute phenotypes to individual members.
Tissue-Specific Expression Patterns: In normal liver, CDKN2C is expressed while CDKN2A and CDKN2B are silenced , but patterns differ across tissues.
Alternative Transcripts: CDKN2A produces both α and β transcripts with distinct functions , complicating expression analysis.
To address these challenges, researchers should:
Use specific antibodies and probes validated for distinguishing INK4 family members
Employ genetic approaches targeting individual INK4 genes
Perform comprehensive analysis of all family members in parallel
Consider tissue-specific expression patterns when interpreting results
Next-generation approaches can provide deeper insights into CDKN2C regulation:
Single-cell technologies:
Single-cell RNA-seq can reveal cell-type-specific expression patterns
Single-cell ATAC-seq can identify accessible chromatin regions controlling CDKN2C expression
CyTOF or single-cell proteomics can assess protein levels and modifications
Epigenetic profiling:
ChIP-seq for histone modifications and transcription factor binding at the CDKN2C locus
DNA methylation analysis to identify regulatory regions
Chromosome conformation capture methods (Hi-C, 4C) to map enhancer-promoter interactions
CRISPR-based screens:
CRISPRi/a screens targeting non-coding regions to identify CDKN2C regulators
Base editing approaches to introduce specific variants
Perturb-seq to assess functional consequences of CDKN2C modulation at single-cell resolution
Long-read sequencing:
Characterization of complex structural variants affecting CDKN2C
Identification of novel transcripts and isoforms
These approaches can help uncover the complex regulatory networks controlling CDKN2C expression in different physiological and pathological contexts.
Cyclin-Dependent Kinase Inhibitor 2C (CDKN2C), also known as p18, is a member of the INK4 family of cyclin-dependent kinase inhibitors. This protein plays a crucial role in regulating the cell cycle by interacting with cyclin-dependent kinases (CDKs), specifically CDK4 and CDK6 . By inhibiting these kinases, CDKN2C functions as a cell growth regulator, controlling the progression of the cell cycle at the G1 phase .
The CDKN2C protein is encoded by the CDKN2C gene. It binds to CDK4 or CDK6, preventing their activation and thus inhibiting the phosphorylation of the retinoblastoma protein (pRB). This inhibition leads to cell cycle arrest in the G1 phase, thereby controlling cell proliferation . The protein’s structure allows it to effectively bind to CDKs and block their interaction with cyclins, which are necessary for the activation of CDKs .
CDKN2C has been identified as a significant player in the development and progression of various cancers. Its expression levels are often altered in cancerous tissues, leading to uncontrolled cell proliferation. For instance, upregulated CDKN2C expression has been detected in small cell lung carcinoma (SCLC) and is associated with poor prognosis . The protein’s role in cancer is not limited to SCLC; it has also been implicated in other cancers such as breast invasive carcinoma and adrenocortical carcinoma .
The clinical significance of CDKN2C extends beyond its role in cancer development. Its expression levels can serve as a prognostic marker, helping to predict the outcome of cancer treatments. High levels of CDKN2C expression are often associated with poor prognosis, making it a potential target for therapeutic interventions . Additionally, CDKN2C’s involvement in the immune microenvironment suggests its potential usefulness in immunotherapy .