Detects CTNND1 in lysates of tumor cells, including NSCLC , HCC , and pancreatic cancer .
Identifies isoform-specific expression (e.g., isoform 1A in melanocytes vs. isoform 3A in keratinocytes) .
Validated for paraffin-embedded tissues (e.g., colon, prostate) .
Demonstrates CTNND1 overexpression in HCC and pancreatic cancer .
Used to localize CTNND1 at adherens junctions and the nucleus .
Confirms its role in EMT/MET transitions in astrocytic tumors .
NSCLC: High CTNND1 expression correlates with immunosuppressive tumor microenvironments (TME) and predicts poor immunotherapy response .
HCC: Promotes cell proliferation, migration, and metastasis via Wnt/β-catenin signaling .
Pancreatic Cancer: Knockdown reduces proliferation (CCK-8 assay) and induces apoptosis (flow cytometry) .
CTNND1 encodes the p120-catenin protein, which has multiple crucial biological functions. It maintains cell-cell junctions, regulates epithelial-mesenchymal transition, and participates in transcriptional signaling pathways . The protein is widely expressed across tissues, with notable presence in vascular endothelium . Different isoforms show tissue-specific expression patterns; for example, melanocytes and melanoma cells primarily express the long isoform 1A, whereas keratinocytes express shorter isoforms, especially 3A . CTNND1 has been implicated in various pathological conditions, including craniofacial disorders such as cleft palate, blepharocheilodontic syndrome, and several cancer types .
CTNND1 can be detected through multiple complementary techniques:
Protein detection: Western blot analysis using specific antibodies (e.g., rabbit polyclonal antibodies) is effective for quantifying CTNND1 protein levels . Immunohistochemistry (IHC) can visualize CTNND1 expression patterns in tissue sections .
mRNA detection: Quantitative real-time PCR (qRT-PCR) measures CTNND1 transcript levels . In situ hybridization with digoxigenin-labeled antisense mRNA probes can visualize tissue-specific expression patterns .
Functional analysis: Cell migration and invasion can be measured by Transwell and Matrigel analyses to assess the functional consequences of CTNND1 manipulation .
The optimal method depends on your specific research question. For comparing expression across different cell lines or tissues, a combination of protein and mRNA detection methods is recommended for robust validation.
When selecting a CTNND1 antibody, researchers should consider:
Isoform specificity: CTNND1 has multiple isoforms with tissue-specific expression. Ensure your selected antibody recognizes the relevant isoform(s) for your study. Some antibodies detect all isoforms, while others may be isoform-specific .
Application compatibility: Verify the antibody is validated for your intended application (Western blot, IHC, etc.). For example, the AF7636 antibody is specifically validated for Western blot applications .
Species reactivity: Confirm the antibody recognizes CTNND1 in your study species. Some antibodies, like AF7636, react with human, mouse, and rat CTNND1, with predicted reactivity to other species including pig, zebrafish, bovine, horse, and rabbit .
Epitope information: Understanding which domain of CTNND1 the antibody recognizes is crucial, especially when studying truncated forms or specific domains .
Validation data: Request validation data showing specificity through knockdown/knockout controls to ensure antibody specificity.
The apparent contradictory roles of CTNND1 in different cancers reflect context-dependent functions that require careful interpretation:
Expression pattern analysis: In hepatocellular carcinoma (HCC), CTNND1 is significantly upregulated and promotes tumor progression . Conversely, in triple-negative breast cancer (TNBC), CTNND1 is downregulated in both primary tumors and bone metastases . These opposite expression patterns suggest cancer-specific roles.
Mechanistic differences: In HCC, CTNND1 promotes metastasis by enhancing Wnt/β-catenin signaling . In TNBC, decreased CTNND1 accelerates bone metastasis through different mechanisms, including increased CXCR4 expression via PI3K/AKT pathway activation .
Microenvironment interactions: CTNND1 knockdown in TNBC leads to increased neutrophil infiltration in the bone microenvironment, affecting cytotoxic T lymphocyte function . This demonstrates how CTNND1 can influence tumor-immune interactions differently across cancer types.
When interpreting seemingly contradictory data, researchers should consider:
Tissue-specific isoform expression
Interaction with different signaling pathways
Effects on the tumor microenvironment
Differences in experimental models
A comprehensive approach comparing signaling pathway activation, cellular phenotypes, and in vivo models across cancer types can help reconcile these differences.
Based on published research, effective experimental designs for studying CTNND1 function include:
Expression modulation approaches:
In vitro functional assays:
In vivo models:
Signaling pathway analysis:
Clinical correlation:
To effectively analyze CTNND1's interaction with Wnt/β-catenin signaling:
Protein interaction analysis:
Co-immunoprecipitation to detect physical interactions between CTNND1 and β-catenin
Proximity ligation assay to visualize protein interactions in situ
Subcellular fractionation to track β-catenin nuclear translocation in response to CTNND1 manipulation
Transcriptional activity measurement:
Pathway manipulation:
Use of Wnt pathway activators (e.g., GSK3β inhibitors) or inhibitors in combination with CTNND1 modulation
Rescue experiments with constitutively active β-catenin in CTNND1-depleted cells
Correlation analysis in clinical samples:
Multiplex immunohistochemistry for co-localization of CTNND1 with β-catenin
Expression correlation between CTNND1 and Wnt targets in patient samples
Research has demonstrated that CTNND1 expression positively correlates with β-catenin, WNT11, Cyclin D1, and BMP7/MMP7 expression in human HCC specimens . This correlation supports the functional relationship between CTNND1 and Wnt signaling in cancer progression.
When performing Western blot analysis with CTNND1 antibodies, include these essential controls:
Positive controls:
Negative controls:
Specificity controls:
Peptide competition assay: Pre-incubating the antibody with immunizing peptide should abolish specific signals
Multiple antibodies targeting different epitopes of CTNND1 to confirm band identity
Different isoform-expressing cells to verify isoform-specific detection
Loading controls:
Housekeeping proteins (β-actin, GAPDH) to normalize protein loading
Total protein staining (Ponceau S, SYPRO Ruby) as an alternative normalization method
Molecular weight verification:
For optimal immunohistochemical detection of CTNND1:
Sample preparation:
Proper fixation is critical; overfixation can mask epitopes
For formalin-fixed paraffin-embedded (FFPE) samples, optimize antigen retrieval methods (heat-induced or enzymatic)
Consider using both FFPE and frozen sections for validation
Antibody optimization:
Perform titration experiments to determine optimal antibody concentration
Test multiple antibodies targeting different CTNND1 epitopes
Validate antibody specificity using CTNND1 knockdown/knockout tissues
Detection system selection:
For low expression, use amplification systems like tyramide signal amplification
For co-localization studies, consider multiplex IHC or immunofluorescence
Controls:
Scoring system development:
Variability in CTNND1 detection may stem from several sources. Address these systematically:
Biological variability:
Cell density effects: CTNND1 expression can vary with cell confluence due to its role in cell-cell adhesion
Cell passage number: Expression may change with extended culturing
Solution: Standardize cell culture conditions and document passage numbers
Technical variability:
Antibody lot-to-lot variation
Inconsistent sample preparation
Solution: Use the same antibody lot when possible; implement standardized protocols
Isoform complexity:
Post-translational modifications:
Phosphorylation status can affect antibody binding
Solution: Consider phospho-specific antibodies if relevant
Statistical approach:
Perform sufficient biological replicates (minimum n=3)
Use appropriate statistical tests to determine significance
Consider normalization methods carefully
When correlating CTNND1 expression with patient outcomes:
Stratification approach:
Define clear criteria for "high" versus "low" expression based on quantitative measurements
Consider using median split, quartiles, or optimized cutoff values
Evaluate the robustness of findings across different stratification methods
Statistical analysis:
Clinical correlation:
Analyze associations between CTNND1 expression and clinicopathological parameters
Research has shown significant correlations between CTNND1 expression and tumor diameter, microvascular invasion, and tumor differentiation in HCC
Consider cancer-specific parameters (e.g., HBsAg status in HCC, hormone receptor status in breast cancer)
Tissue heterogeneity:
Validation approaches:
Several emerging technologies hold promise for advancing CTNND1 research:
Single-cell technologies:
Single-cell RNA sequencing to identify cell-specific CTNND1 expression patterns within heterogeneous tumors
Single-cell proteomics to analyze CTNND1 protein levels and modifications at the individual cell level
Spatial transcriptomics to map CTNND1 expression within the tumor microenvironment
Advanced imaging techniques:
Super-resolution microscopy to visualize CTNND1 localization at subcellular structures
Intravital imaging to monitor CTNND1 dynamics during metastasis in live animal models
Multiplexed imaging (e.g., CODEX, MIBI) to simultaneously detect multiple proteins in the CTNND1 pathway
Gene editing advancements:
CRISPR activation/inhibition systems for temporal control of CTNND1 expression
Base editing or prime editing for introducing specific CTNND1 mutations
CRISPR screens to identify synthetic lethal interactions with CTNND1
Liquid biopsy applications:
In silico approaches:
AI/machine learning to predict CTNND1 interaction networks
Structural biology and molecular dynamics simulations to understand CTNND1 protein interactions
Systems biology approaches to model CTNND1's role in cellular signaling networks
CTNND1 research shows potential for clinical translation in several areas: