Overexpression: CCNI2 is significantly upregulated in CRC tissues and cell lines (e.g., HCT 116, RKO), correlating with advanced pathological grading and poor patient prognosis .
Functional Impact:
CCNI2 binds CDK5 with higher affinity than its homolog CCNI, enhancing CDK5 kinase activity by ~40% compared to CCNI .
Subcellular localization: CCNI2 retains CDK5 in the cytoplasm and plasma membrane, distinct from CCNI’s nuclear localization .
Prognostic Marker: High CCNI2 expression in CRC tissues predicts shorter survival (Kaplan-Meier analysis, p < 0.05) .
Therapeutic Target: Silencing CCNI2 disrupts CRC progression via apoptosis induction and proliferation inhibition, suggesting potential for RNAi-based therapies .
CCNI2 (Cyclin I-like or Cyclin I2) is a protein that plays a significant role in cell cycle regulation with an approximate mass of 35 kDa . As a member of the cyclin family, CCNI2 functions as a cell cycle regulator involved in critical cellular processes. Research has demonstrated that CCNI2 contributes to cancer progression through several mechanisms:
Cell proliferation regulation: CCNI2 promotes cancer cell proliferation, with knockdown studies showing reduced proliferation rates in colorectal and gastric cancer cell lines
Cell cycle control: CCNI2 knockdown arrests cell cycle in G2 phase in colorectal cancer cells
Apoptosis modulation: Reduced CCNI2 expression increases susceptibility to apoptosis by affecting key apoptotic factors including BID, BIM, Caspase3, and DR6
For experimental investigations, researchers should consider using specific antibodies that target different epitopes of CCNI2, particularly those corresponding to amino acids 150 to C-terminus of the human protein, which have been validated for western blot analysis .
CCNI2 exhibits distinct expression patterns that vary between cancer and normal tissues:
Colorectal Cancer (CRC):
Immunohistochemical staining shows significantly higher CCNI2 expression in CRC tissues compared to paracarcinoma tissues
qPCR analysis confirms elevated CCNI2 mRNA levels in CRC cell lines (CACO2, RKO, SW480, and HCT 116) compared to normal colorectal mucosal cells (FHC)
Gastric Cancer:
Based on TCGA database analysis of 407 gastric cancer samples, CCNI2 expression is significantly higher in tumor tissues compared to normal samples
High expression of CCNI2 was observed in 43 of 93 tumor tissues (46.2%) and in 0 of 101 normal tissues in clinical samples
When designing experiments to evaluate CCNI2 expression, researchers should implement both protein-level detection (immunohistochemistry, western blot) and transcript-level analysis (qPCR) to obtain comprehensive expression profiles across different sample types.
Based on published research protocols, the following methodologies are recommended for optimal CCNI2 detection:
Immunohistochemical Staining:
Formalin-fixed paraffin-embedded samples should be cut into 5-μm sections, deparaffinized and rehydrated
Primary antibody: Use anti-CCNI2 antibody (1:50 dilution), incubate at 4°C overnight
Secondary antibody: HRP goat anti-rabbit IgG (1:200 dilution), incubate at room temperature for 30 min
Signal detection: Stain with DAB and hematoxylin at room temperature
Western Blot Analysis:
Total cellular proteins (20 μg) should be subjected to 10% SDS-PAGE
Transfer proteins to PVDF membranes via wet transfer
Blocking: Use TBST with 5% skim milk at 4°C for 1 hour
Primary antibody: Anti-CCNI2 antibody (1:1000, Abcam ab97767) at 4°C overnight
Secondary antibody: HRP-conjugated goat anti-rabbit IgG (1:3000) at room temperature for 2 hours
Detection: Use ECL-Plus™ Western blotting system kit for visualization
qPCR Analysis:
Design primers specific to CCNI2 gene (NM_001287253.1)
Normalize expression data to appropriate housekeeping genes
When validating CCNI2 knockdown efficiency, a multi-method approach is essential:
shRNA Design and Selection:
Design multiple shRNA sequences targeting different regions of CCNI2 (e.g., shCCNI2-1: 5'-ATCTGCGACGCCTTCGAGGAA-3'; shCCNI2-2: 5'-TACCTGCATTGCGCCACAATT-3'; shCCNI2-3: 5'-CCTGGAAGGCGACCTGGACGA-3')
Include appropriate negative control (e.g., shCtrl: 5'-TTCTCCGAACGTGTCACGT-3')
Transduction Efficiency Assessment:
Use lentiviral vectors with GFP tag to visually confirm transduction (infection efficiency should exceed 80%)
Knockdown Validation:
qPCR Validation: Quantify relative CCNI2 mRNA expression levels compared to control
Western Blot Validation: Confirm protein level reduction
Functional Validation: Perform cellular assays to confirm phenotypic changes
For comprehensive investigation of CCNI2 function in cancer progression, researchers should implement the following experimental design strategy:
In Vitro Studies:
Cell Proliferation Assessment:
Colony Formation Analysis:
Cell Cycle Analysis:
Apoptosis Assessment:
In Vivo Studies:
Mouse Xenograft Models:
Tumor Analysis:
Mechanism Investigation:
Pathway Analysis:
Correlation with Clinical Data:
Distinguishing the specific effects of CCNI2 from other cyclins requires careful experimental design:
Specificity Controls:
Expression Correlation Analysis:
Selective Knockdown and Rescue Experiments:
Design highly specific shRNAs targeting unique regions of CCNI2
Confirm specificity by measuring expression levels of other cyclin family members
Perform rescue experiments with shRNA-resistant CCNI2 constructs to confirm phenotype specificity
Functional Discrimination:
Cell Cycle Phase Analysis:
Protein Interaction Network Analysis:
Identify CCNI2-specific binding partners via co-immunoprecipitation followed by mass spectrometry
Compare with interaction networks of other cyclins
Focus on unique interactions as potential mediators of CCNI2-specific effects
Data Integration Approach:
| Cyclin | Primary Cell Cycle Phase | Key Binding Partners | Cancer Association | Key Distinguishing Features |
|---|---|---|---|---|
| CCNI2 | G2 phase arrest upon knockdown | Unknown (potential: CDK5) | Colorectal, Gastric | High correlation with pathological grade |
| CCNI | Regulates MAPK signaling | CDK5 | Various cancers | Prevents injury-induced apoptosis |
| CCND2 | G1/S transition | CDK4, CDK6 | Lymphoma, Leukemia | Often associated with MYC activation |
| CCNB2 | G2/M transition | CDK1 | Various solid tumors | Multiple phosphorylation sites (Ser10, Ser11, Ser22) |
This comprehensive comparative approach allows researchers to isolate CCNI2-specific effects from those of other cyclins.
Based on current research findings, several molecular mechanisms have been implicated in CCNI2-mediated cancer progression:
Apoptosis Regulation:
CCNI2 knockdown in cancer cells modulates key apoptotic factors:
When investigating this mechanism, researchers should:
Examine mitochondrial membrane potential changes
Monitor caspase activation cascade
Assess cytochrome c translocation from mitochondria to cytosol
Cell Cycle Regulation:
CCNI2 impacts cell cycle progression with knockdown resulting in:
Research approaches should include:
CDK activity assays to identify CCNI2-regulated CDKs
Phosphorylation analysis of cell cycle checkpoint proteins
Time-lapse imaging to visualize cell cycle progression defects
Signaling Pathway Modulation:
Evidence suggests CCNI2 influences multiple signaling pathways:
To elucidate these mechanisms, researchers should:
Perform phosphoproteomic analysis to identify signaling alterations
Use pathway-specific inhibitors to determine functional relevance
Apply CRISPR-Cas9 screening to identify synthetic lethal interactions
Transcriptional Regulation:
CCNI2 may influence gene expression programs:
RNA sequencing following CCNI2 knockdown reveals differentially expressed genes
Potential involvement in transcriptional regulation through interaction with HDGF
Comprehensive analysis should include:
ChIP-seq to identify genomic binding sites of CCNI2-associated factors
RNA-seq time course following CCNI2 modulation
ATAC-seq to assess chromatin accessibility changes
When faced with contradictory data on CCNI2 function across cancer types, researchers should implement the following systematic approach:
1. Context-Dependent Analysis Framework:
Establish a standardized experimental design that accounts for:
Cancer type-specific genetic backgrounds
Tissue microenvironment factors
Patient characteristics (age, gender, tumor stage)
2. Meta-Analysis of Expression Data:
Compare CCNI2 expression patterns across cancer types:
In colorectal cancer, high CCNI2 expression correlates with:
In gastric cancer, CCNI2 expression correlates with:
3. Functional Validation Across Multiple Models:
4. Reconciliation Strategies:
When contradictory results are observed:
Evaluate differences in experimental methodologies
Consider genetic heterogeneity within cancer subtypes
Examine post-translational modifications affecting CCNI2 function
Assess differences in binding partners across tissue types
5. Integrated Mechanism Model:
Develop a comprehensive model that incorporates cancer type-specific variations:
Core conserved functions (proliferation, apoptosis regulation)
Tissue-specific pathways and interactions
Microenvironment-dependent effects
Patient-specific factors (genetic background, comorbidities)
By implementing this systematic approach, researchers can develop a nuanced understanding of CCNI2 function that accounts for context-dependent variations across cancer types.
When designing in vivo experiments to study CCNI2 function, researchers should address several critical considerations:
1. Animal Model Selection:
Xenograft Models:
Genetically Engineered Mouse Models (GEMMs):
Consider tissue-specific CCNI2 knockout or overexpression using Cre-loxP systems
Evaluate tissue-specific promoters for targeted expression (e.g., Villin for intestinal epithelium)
Account for potential developmental effects of CCNI2 modulation
2. CCNI2 Modulation Strategies:
Stable Knockdown Approach:
Inducible Systems:
Consider doxycycline-inducible shRNA systems for temporal control
Implement CRISPR-Cas9 approaches for complete gene knockout
Evaluate pharmacological approaches if available
3. Experimental Design Parameters:
Sample Size Determination:
Timing Considerations:
4. Analysis Methods:
Tumor Growth Assessment:
Molecular and Histological Analysis:
5. Ethical and Regulatory Considerations:
Follow institutional animal care guidelines and obtain proper approvals
Implement humane endpoints based on tumor burden and animal welfare
Consider the 3Rs principle (Replacement, Reduction, Refinement)
Ensure proper reporting of animal experiments following the ARRIVE guidelines
6. Translational Relevance:
Correlate findings with clinical data from patient samples
Consider patient-derived xenograft models for higher clinical relevance
Evaluate potential therapeutic implications through combination studies