Cyclin B1 (CCNB1), a regulatory protein encoded by the CCNB1 gene, is critical for controlling the G2/M transition of the cell cycle. It forms a complex with cyclin-dependent kinase 1 (CDK1) to create the maturation-promoting factor (MPF), which triggers mitotic events such as nuclear envelope breakdown and chromatin condensation . Overexpression of CCNB1 is linked to aggressive tumor behavior in cancers like breast, prostate, and non-small cell lung cancer, making it a focal point for diagnostic and therapeutic research .
CCNB1 monoclonal antibodies are laboratory tools designed to detect and study this protein. They are engineered to bind specifically to CCNB1, enabling applications in immunohistochemistry (IHC), western blotting (WB), flow cytometry (FC), and immunoprecipitation (IP). Below is a detailed analysis of their characteristics, clinical relevance, and technical specifications.
Notes:
MA1-155 detects a prominent ~52 kDa band in treated cells, with additional bands at ~35 kDa and ~75 kDa .
MA5-15714 targets a ~60 kDa CCNB1 isoform and shows species-specific reactivity .
CCNB1/1098 is validated for paraffin-embedded tissue staining .
High CCNB1 expression is strongly associated with lymphovascular invasion (LVI), tumor aggressiveness, and poor survival in breast cancer (BC):
Key Findings:
CCNB1 mRNA and protein levels correlate weakly (r = 0.136) .
CCNB1 interacts with LVI-related biomarkers (e.g., N-cadherin, TWIST2) .
CCNB1 monoclonal antibodies enable:
Cell Cycle Analysis: Detection of G2/M phase cells via FC or IF .
Tumor Diagnosis: IHC staining to assess CCNB1 overexpression in paraffin sections .
Drug Response Studies: Monitoring CCNB1 degradation under chemotherapeutic agents (e.g., camptothecin, hydroxyurea) .
Antibody | Cross-Reactivity | Validation Methods |
---|---|---|
MA1-155 | Cyclin B2 (WB) | IP, WB, IHC, IF, FC |
GNS-1 | Hamster, Mouse | WB, IHC, FC, IP, IF |
RM281 | N/A | WB, IHC, IP |
Challenges:
Epitope Variability: MA1-155 binds an epitope near the N-terminus (AA 1-21) , while others target different regions (e.g., 5G6 binds a ~60 kDa isoform) .
Species Limitations: Most antibodies are human-specific, though MA1-155 and GNS-1 show cross-reactivity with rodent models .
CCNB1 (Cyclin B1) is a regulatory protein involved in mitosis that complexes with p34(cdc2) to form the maturation-promoting factor (MPF). The protein exists in two alternative transcripts: a constitutively expressed transcript and a cell cycle-regulated transcript predominantly expressed during G2/M phase . CCNB1 has emerged as an important biomarker in cancer research, particularly breast cancer, where high expression correlates with aggressive tumor behavior, presence of lymphovascular invasion (LVI), larger tumor size, higher histological grade, hormonal receptor negativity, and HER2 positivity . These correlations make CCNB1 a valuable target for understanding cancer progression mechanisms and identifying potential therapeutic targets.
Recent research has established CCNB1 as an independent predictor of shorter breast cancer-specific survival (HR = 1.3; 95% CI 1.2–1.5; p = 0.010), highlighting its prognostic value . Given these associations, CCNB1 monoclonal antibodies serve as essential tools for investigating cell cycle regulation abnormalities in cancer cells and assessing patient prognosis.
When performing immunohistochemistry with CCNB1 monoclonal antibodies, researchers should consider the following optimized protocol based on published research:
Sample preparation: Use formalin-fixed, paraffin-embedded tissue sections (4-5 μm thickness). For tissue microarrays (TMAs), ensure cores contain at least 15% invasive tumor area .
Antibody selection: Choose validated antibodies with demonstrated specificity, such as mouse monoclonal anti-CCNB1 antibody (ab72, Abcam) used in published research .
Antibody validation: Confirm specificity using western blot prior to IHC staining using appropriate breast cancer cell lines (e.g., MCF-7, SK-BR-3, MB-MDA-231) .
Dilution optimization: For immunofluorescence, a dilution of 0.5-1 μg/mL is recommended as a starting point . The optimal working dilution should be determined empirically for each experimental system.
Scoring system: Use the H-score method, which combines staining intensity (negative=0, weak=1, moderate=2, strong=3) and percentage of positive cells (0-100%), resulting in a score range of 0-300 .
Concordance assessment: For reliable results, scoring should be performed by at least two observers (e.g., a researcher and a pathologist) with concordance evaluation (aim for ICC ≥ 0.9) .
Cut-off determination: Establish a cut-off for CCNB1 positivity based on the cohort's median H-score (e.g., 100 H-score in breast cancer studies) .
This protocol has been successfully applied in large breast cancer cohorts (n=2480) with long-term outcome data, demonstrating its reliability for clinical research applications .
Validating CCNB1 monoclonal antibody specificity requires a multi-faceted approach:
Western blot validation:
Use multiple relevant cell lines (e.g., MCF-7, SK-BR-3, MB-MDA-231 for breast cancer research)
Apply primary antibody at 1:1000 dilution and appropriate secondary antibody (e.g., IRDye 700CW Donkey anti-mouse at 1:15,000)
Confirm a specific band at the expected molecular weight (40-62 kDa for CCNB1)
Include endogenous control (e.g., GAPDH with anti-GAPDH primary antibody at 1:5000)
Positive and negative controls:
Include known positive tissues/cells with high CCNB1 expression (e.g., highly proliferative tumors)
Use appropriate negative controls (antibody diluent only) on serial sections
Consider siRNA knockdown of CCNB1 in cell lines as an additional specificity control
Cross-reactivity assessment:
Functional validation:
Proper validation ensures experimental results accurately reflect CCNB1 biology and avoids misinterpretation of data due to non-specific antibody binding.
Studying CCNB1's role in lymphovascular invasion (LVI) requires sophisticated experimental approaches:
Transcriptomic-proteomic correlation:
Combine mRNA expression analysis from large cohorts (e.g., METABRIC n=1980, TCGA n=854) with protein expression using tissue microarrays (n=2480)
Assess correlation between mRNA and protein levels (note: previous studies found weak correlation r=0.136)
Compare expression between LVI-positive and LVI-negative tumors using appropriate statistical tests
Co-expression analysis with LVI-related biomarkers:
Investigate associations between CCNB1 and established LVI markers including N-cadherin, P-cadherin, and TWIST2
Perform multiplex immunofluorescence to visualize co-localization in tumor sections
Quantify co-expression using digital pathology platforms
Functional studies in model systems:
Modulate CCNB1 expression in breast cancer cell lines using overexpression or knockdown approaches
Assess effects on invasion capabilities using transwell invasion assays
Evaluate impact on LVI-related biomarkers at protein and mRNA levels
Patient-derived xenograft models:
Create PDX models from tumors with varying CCNB1 expression levels
Monitor LVI development using specialized staining methods
Test targeted interventions against CCNB1 or downstream pathways
Multivariate analysis for clinical significance:
Perform Cox regression analysis adjusting for confounding factors
Calculate hazard ratios for association between CCNB1 expression and LVI
Develop and validate predictive models incorporating CCNB1 and other biomarkers
This integrated approach has revealed that high CCNB1 expression is significantly associated with LVI presence (p<0.0001) and serves as an independent predictor of shorter breast cancer-specific survival (HR=1.3; 95% CI 1.2–1.5; p=0.010) .
CCNB1 nuclear translocation is a critical regulatory event in cell cycle progression. Research strategies to study this process include:
Cellular fractionation with western blotting:
Live-cell imaging with fluorescently tagged CCNB1:
Create fluorescent protein fusions with CCNB1
Perform time-lapse confocal microscopy during cell cycle progression
Quantify nuclear/cytoplasmic signal intensity ratios
Correlate with cell cycle phase markers
Fixed-cell immunofluorescence:
Modulation of transport mechanisms:
Proximity ligation assays:
Detect interactions between CCNB1 and nuclear transport factors
Visualize spatial relationships at specific cell cycle stages
Quantify interaction frequency under different conditions
This methodological framework has been successfully applied to demonstrate that circ-CCNB1 suppresses CCNB1 nuclear expression in trophoblast cells, providing insights into regulatory mechanisms .
Optimizing co-immunoprecipitation (co-IP) protocols for CCNB1 interactions requires careful consideration of experimental conditions:
Antibody selection and amount:
Lysis buffer optimization:
For stable interactions (e.g., CCNB1-CDK1): use RIPA buffer with protease/phosphatase inhibitors
For transient interactions: use milder NP-40 or digitonin-based buffers
Avoid harsh detergents that may disrupt protein complexes
Cross-linking considerations:
For weak interactions: incorporate reversible cross-linkers (e.g., DSP)
Optimize cross-linking time and concentration
Include appropriate controls to confirm specificity
Cell synchronization:
Synchronize cells at G2/M phase to maximize CCNB1-complex formation
Verify synchronization efficiency by flow cytometry
Compare interaction profiles across cell cycle phases
Validation approaches:
Perform reverse co-IP (precipitate with partner antibody, detect CCNB1)
Include IgG control and CCNB1-depleted lysate controls
Confirm functional relevance of interactions with kinase assays
Harvest synchronized cells in mid-M phase
Lyse in buffer containing 0.5% NP-40, 150mM NaCl, 50mM Tris pH 7.5, protease/phosphatase inhibitors
Clear lysate by centrifugation (14,000g, 10 min, 4°C)
Pre-clear with protein A/G beads (1 hour, 4°C)
Incubate cleared lysate with 1-2 μg anti-CCNB1 antibody overnight at 4°C
Add protein A/G beads for 2 hours
Wash 4-5 times with lysis buffer
Elute in Laemmli buffer and analyze by western blot
This approach has been successfully used to precipitate active CDK1/cyclin B1 complexes in research applications .
Flow cytometry using CCNB1 antibodies requires careful experimental design to generate reliable data:
Sample preparation optimization:
For cell lines: gentle fixation with 4% paraformaldehyde followed by permeabilization with 0.1-0.5% Triton X-100
For primary tissues: optimize dissociation protocols to maintain epitope integrity
Use appropriate blocking (5-10% serum from secondary antibody host species)
Antibody titration and controls:
Multi-parameter analysis:
Combine with DNA content measurement (PI, DAPI) to correlate with cell cycle phase
Include markers of proliferation (Ki-67) or mitosis (phospho-histone H3)
Consider co-staining for CDK1 to evaluate complex formation
Signal amplification considerations:
For low abundance detection: consider secondary antibody signal amplification
Evaluate fluorochrome brightness relative to expected expression level
Optimize signal-to-noise ratio through careful titration
Data analysis approaches:
Gate on single cells using FSC-H vs. FSC-A
Analyze CCNB1 expression relative to cell cycle phase
Consider bivariate analysis of CCNB1 vs. DNA content
Gate | Purpose | Parameters |
---|---|---|
1 | Cell selection | FSC vs. SSC |
2 | Single cell selection | FSC-A vs. FSC-H |
3 | Viable cell selection | Viability dye negative |
4 | Cell cycle phase | DNA content (PI/DAPI) |
5 | CCNB1 expression | CCNB1 fluorescence intensity |
This methodical approach allows researchers to accurately quantify cell cycle-dependent expression patterns of CCNB1 and correlate them with other cellular parameters.
Discordant results between CCNB1 mRNA and protein expression levels are commonly observed in research. A systematic approach to understanding these differences includes:
Statistical evaluation of correlation:
Biological explanations for discordance:
Post-transcriptional regulation: miRNAs targeting CCNB1 mRNA
Post-translational modifications affecting protein stability
Protein compartmentalization (nuclear vs. cytoplasmic localization)
Alternative splicing generating different protein isoforms
Technical considerations:
Different detection thresholds between RNA sequencing and IHC
Tissue heterogeneity in bulk samples vs. cellular resolution in IHC
Antibody specificity to certain isoforms or modified forms
RNA quality and degradation effects
Validation strategies:
Single-cell approaches to correlate mRNA and protein in individual cells
Pulse-chase experiments to assess protein turnover rates
Investigation of regulatory factors using CCNB1 reporter constructs
Targeted inhibition of degradation pathways
Functional significance assessment:
Determine whether mRNA or protein better predicts biological outcomes
Evaluate which measure correlates more strongly with clinical parameters
Develop integrated biomarker signatures combining both parameters
Step | Approach | Expected Outcome |
---|---|---|
1 | Verify technical reliability | Confirm measurement accuracy for both mRNA and protein |
2 | Assess spatiotemporal factors | Determine if sampling timing/location explains differences |
3 | Investigate regulatory mechanisms | Identify post-transcriptional/post-translational regulators |
4 | Evaluate functional relevance | Determine which measure better correlates with phenotype |
5 | Develop integrated models | Create predictive models incorporating both parameters |
This systematic approach helps researchers interpret seemingly contradictory results and gain deeper insights into CCNB1 biology and its regulatory mechanisms.
Researchers frequently encounter several challenges when performing CCNB1 immunohistochemistry that can affect result interpretation:
Variable staining intensity:
Problem: Inconsistent staining between samples or within the same section
Solution: Standardize fixation time (24-48 hours), use automated staining platforms, and ensure consistent antigen retrieval conditions (buffer pH and temperature)
Validation: Include control tissues on each slide to monitor staining consistency
Background staining:
Problem: Non-specific staining obscuring true CCNB1 signal
Solution: Optimize blocking (5-10% serum, 1 hour), extend washing steps, and titrate primary antibody concentration
Validation: Include isotype control and secondary-only control
Epitope masking:
Problem: Inability to detect CCNB1 despite known expression
Solution: Test multiple antigen retrieval methods (citrate pH 6.0 vs. EDTA pH 9.0), optimize retrieval time (15-30 minutes), and consider alternative fixatives
Validation: Use multiple antibodies targeting different epitopes
Scoring reproducibility:
Cut-off determination:
Problem: Arbitrary thresholds affecting clinical correlations
Solution: Use data-driven approaches based on median or ROC curve analysis
Validation: Apply multiple cut-offs and compare with clinical outcomes
When troubleshooting challenging samples, implement a systematic approach testing one variable at a time and documenting all protocol modifications to ensure reproducibility.
Distinguishing between active and inactive CCNB1 forms is crucial for understanding its functional state in cell cycle regulation:
Phosphorylation-specific antibodies:
Approach: Use antibodies specifically recognizing phosphorylated residues on CCNB1 that indicate activation
Key sites: Ser126, Ser128, Ser133, Ser147 (activating phosphorylations)
Validation: Treatment with phosphatase to confirm specificity
CDK1-CCNB1 complex detection:
Approach: Proximity ligation assay (PLA) to visualize CCNB1-CDK1 interaction
Advantage: Provides spatial information about complex formation
Controls: Serum-starved cells (negative) vs. mitotic cells (positive)
Subcellular localization analysis:
Activity-based probes:
Approach: Use fluorescent substrates that are modified by active CDK1-CCNB1 complex
Readout: Flow cytometry or live-cell imaging to measure substrate modification
Advantages: Direct measurement of functional activity
Immunoprecipitation of active complexes:
By combining these approaches, researchers can distinguish between total CCNB1 levels and the functionally relevant active pool, providing more meaningful insights into cell cycle regulation and dysregulation in disease states.
CCNB1 expression varies significantly across breast cancer molecular subtypes, providing important insights into tumor biology and potential therapeutic strategies:
Expression patterns across PAM50 subtypes:
Correlation with receptor status:
Relationship with tumor grade and proliferation:
Strong positive correlation with high histological grade
Association with proliferation markers
Significantly higher in aggressive tumor phenotypes
Multivariate analysis findings:
Molecular Subtype | Relative CCNB1 Expression | Associated Features |
---|---|---|
Basal-like | Highest | ER/PR negative, high grade, poor prognosis |
HER2-enriched | High | HER2 positive, high grade |
Luminal B | Intermediate | ER positive, higher proliferation |
Luminal A | Low | ER/PR positive, lower grade, better prognosis |
Normal-like | Lowest | Similar to normal breast tissue |
This pattern of expression suggests that CCNB1 may play a particularly important role in more aggressive breast cancer subtypes and could serve as a potential therapeutic target in these difficult-to-treat tumors.
The relationship between CCNB1 and circular RNAs (circRNAs) represents an emerging area of research with implications for disease understanding:
Circ-CCNB1 regulatory functions:
Molecular mechanisms:
Functional consequences:
Affects trophoblast proliferation and invasion
May contribute to pathological processes in pregnancy complications
Creates a regulatory feedback loop affecting CCNB1 activity
Experimental approaches for studying circ-CCNB1/CCNB1 interactions:
Broader implications:
Suggests a novel layer of post-transcriptional regulation
Indicates circRNAs may regulate protein function through localization control
Opens potential therapeutic avenues targeting circular RNA-protein interactions
This research area demonstrates how circRNAs like circ-CCNB1 can regulate CCNB1 function not by changing expression levels but by affecting subcellular localization, representing an important regulatory mechanism with potential implications for various pathological conditions including reproductive disorders and cancer.
CCNB1 monoclonal antibodies can contribute significantly to the development of prognostic models in cancer through several methodological approaches:
Tissue microarray (TMA) analysis:
Methodology: Immunohistochemical staining of TMAs from large patient cohorts (n>2000)
Scoring system: H-score combining intensity (0-3) and percentage (0-100%)
Validation: Requires blinded scoring by multiple observers with high concordance (ICC≥0.9)
Cut-off determination: Data-driven approaches using median or outcome-based optimization
Integration with molecular data:
Multi-marker prognostic panels:
Quantitative image analysis:
Technology: Digital pathology platforms for automated quantification
Advantages: Reduces subjectivity, increases throughput
Features: Nuclear/cytoplasmic ratio, staining heterogeneity, spatial patterns
Integration: Deep learning algorithms to identify subtle expression patterns
Clinical implementation considerations:
Standardization: Protocol harmonization across laboratories
Quality control: Regular proficiency testing
Reporting: Standardized scoring and interpretation guidelines
Clinical validation: Prospective studies showing impact on patient management
Parameter | Hazard Ratio | 95% CI | p-value |
---|---|---|---|
CCNB1 (high vs. low) | 1.3 | 1.2-1.5 | 0.010 |
Tumor size (>2cm vs. ≤2cm) | 1.4 | 1.2-1.7 | 0.001 |
Nodal status (positive vs. negative) | 1.8 | 1.5-2.2 | <0.001 |
Histological grade (3 vs. 1/2) | 1.5 | 1.3-1.8 | <0.001 |
ER status (negative vs. positive) | 1.7 | 1.4-2.0 | <0.001 |
Based on published research, this integrated approach has demonstrated that CCNB1 provides independent prognostic information beyond standard clinicopathological parameters in breast cancer .
Single-cell analysis of CCNB1 protein expression is advancing rapidly with several innovative methodologies:
Mass cytometry (CyTOF):
Technology: Metal-tagged antibodies for deep phenotyping
Advantage: Minimal spectral overlap allowing 40+ parameters simultaneously
Application: Correlate CCNB1 with multiple cell cycle regulators and signaling pathways
Challenge: Antibody validation for metal conjugation
Single-cell Western blotting:
Approach: Microfluidic platforms for protein separation from individual cells
Benefit: Confirms antibody specificity at single-cell level
Application: Heterogeneity analysis in tumor samples
Advancement: Multiplexed detection of CCNB1 along with binding partners
Imaging mass cytometry:
Methodology: Laser ablation of tissue sections stained with metal-tagged antibodies
Advantage: Spatial information preserved with subcellular resolution
Application: Tumor microenvironment context for CCNB1 expression
Analysis: Machine learning algorithms for pattern recognition
In situ protein sequencing:
Technology: Antibody-based detection with DNA barcodes for spatial mapping
Benefit: Highly multiplexed protein detection in intact tissue
Application: Relationship between CCNB1 and microenvironmental factors
Development stage: Emerging technique with growing applications
Live-cell reporters:
Approach: CRISPR knock-in of fluorescent tags to endogenous CCNB1
Advantage: Real-time monitoring of expression and localization
Application: Cell cycle dynamics in living tumor cells
Extension: Combined with optogenetic tools for functional manipulation
These emerging technologies are transforming our understanding of CCNB1 biology by revealing cell-to-cell variability, spatial context, and temporal dynamics that are masked in bulk analyses, potentially leading to more precise prognostic indicators and therapeutic strategies.
Integrating CCNB1 antibody-derived data with multi-omics approaches creates powerful research frameworks:
Transcriptomic-proteomic integration:
Epigenomic-proteomic correlation:
Technique: Integrate CCNB1 protein data with DNA methylation and histone modification profiles
Application: Identify epigenetic regulators of CCNB1 expression
Analysis: Multivariate regression models linking epigenetic patterns to protein levels
Outcome: Potential epigenetic biomarkers predicting CCNB1 activity
Proteogenomic integration:
Framework: Combine genomic alterations, transcriptomics, and CCNB1 protein data
Advantage: Comprehensive view of regulatory mechanisms
Approach: Pathway analysis incorporating genetic variants affecting CCNB1 network
Clinical relevance: Identify patient subgroups for targeted therapies
Spatial multi-omics:
Technology: Spatial transcriptomics combined with multiplexed protein imaging
Application: Map CCNB1 protein expression to spatially resolved transcriptomes
Benefit: Context-dependent understanding of CCNB1 regulation
Analysis: Spatial statistics and neighborhood analyses
Network-based integration:
Methodology: Construct protein-protein interaction networks centered on CCNB1
Data sources: Antibody-based interactome data, phosphoproteomics, transcriptomics
Analysis: Network perturbation algorithms to identify key regulators
Outcome: System-level understanding of CCNB1 function
Data Layer | Technology | Integration Approach | Outcome |
---|---|---|---|
Genomics | WGS/WES | Variant impact prediction | Genetic modulators |
Transcriptomics | RNA-seq | Correlation analysis | Expression regulation |
Proteomics | IHC/MS | Protein network analysis | Functional complexes |
Epigenomics | ATAC-seq/MethylSeq | Regulatory region identification | Epigenetic control |
Metabolomics | MS | Pathway enrichment | Metabolic impact |