CPNE7 Antibody is a specialized immunological reagent designed to detect and bind to Copine VII (CPNE7), a calcium-dependent phospholipid-binding protein involved in calcium-mediated intracellular processes . These antibodies are typically raised in rabbits against human CPNE7 protein and are available in various forms including non-conjugated and conjugated versions to suit different experimental applications .
The development of specific antibodies against CPNE7 has enabled researchers to investigate the expression patterns, subcellular localization, and functional implications of this protein in both normal physiological conditions and disease states. As research continues to uncover the significant role of CPNE7 in cancer progression, particularly in colorectal cancer, these antibodies have become invaluable tools for advancing our understanding of disease mechanisms and identifying potential therapeutic targets.
CPNE7 belongs to the copine family of calcium-dependent phospholipid-binding proteins that play crucial roles in membrane trafficking and signal transduction. The full-length human CPNE7 protein consists of 558 amino acids with a calculated molecular weight of approximately 62 kDa, though the observed molecular weight in laboratory analyses typically ranges between 65-70 kDa .
The CPNE7 gene is identified in various biological databases with the following identifiers:
While originally characterized as a membrane protein, immunohistochemical studies have revealed that CPNE7 can also be found in the cytoplasm and nucleus, suggesting diverse cellular functions depending on its subcellular localization .
CPNE7 antibodies have been employed in a wide range of research applications, enabling investigators to study the expression, localization, and function of CPNE7 in various biological contexts.
Western blotting represents one of the most common applications for CPNE7 antibodies, with recommended dilutions ranging from 1:500 to 1:8000 depending on the specific product . This technique allows for the quantification and molecular weight determination of CPNE7 protein in diverse biological samples. The Proteintech CPNE7 antibody (17396-1-AP) has demonstrated successful detection of CPNE7 in multiple sample types, including COLO 320 cells, mouse brain tissue, and rat brain tissue .
Immunohistochemical (IHC) staining with CPNE7 antibodies has proven particularly valuable for examining CPNE7 expression patterns in tissue samples. With typical recommended dilutions between 1:50 and 1:500 , these antibodies have been instrumental in revealing increased CPNE7 expression in colorectal cancer tissues compared to normal tissues . IHC applications have also helped elucidate the subcellular distribution of CPNE7, showing its presence not only in the cell membrane but also in the cytoplasm and nucleus .
For immunofluorescence applications, which enable visualization of CPNE7 within cells and tissues with high resolution, dilutions of 1:50 to 1:200 are typically recommended . FITC-conjugated CPNE7 antibodies are particularly suited for this technique, allowing for direct fluorescent detection without secondary antibodies .
Several CPNE7 antibodies, particularly those conjugated with HRP or Biotin, have been specifically developed for ELISA applications . This technique allows for quantitative detection of CPNE7 in solution, providing a valuable tool for high-throughput screening and quantitative analysis.
A critical application of CPNE7 antibodies has been in validating the efficiency of CPNE7 knockdown or knockout in experimental studies investigating the functional role of this protein . These antibodies have been essential for confirming reduced CPNE7 expression at the protein level following siRNA or shRNA treatment in studies examining CPNE7's involvement in cancer progression .
Recent studies have highlighted the significant role of CPNE7 in cancer biology, particularly in colorectal cancer (CRC). CPNE7 antibodies have been instrumental in these investigations, enabling researchers to analyze CPNE7 expression patterns and correlate them with clinical outcomes.
Multiple studies have demonstrated that CPNE7 is significantly upregulated in CRC patient tissues and CRC cell lines compared to normal tissues . This elevated expression has been confirmed through various techniques including immunohistochemical staining, quantitative real-time PCR (qRT-PCR), and western blotting using CPNE7-specific antibodies . Importantly, CPNE7 expression has been shown to correlate positively with cancer progression, with higher expression levels observed in more advanced TNM stages .
Kaplan-Meier survival analysis has revealed a strong correlation between CPNE7 expression levels and patient outcomes in colorectal cancer. At 40 months post-diagnosis, approximately 27% of patients with low CPNE7 expression had died, compared to approximately 58% of patients with high CPNE7 expression . These significant differences in survival rates highlight the potential value of CPNE7 as a prognostic biomarker in CRC.
Experimental studies utilizing CPNE7 knockdown approaches have provided valuable insights into the functional importance of CPNE7 in cancer progression. The suppression of CPNE7 expression through siRNA or shRNA techniques has been shown to significantly inhibit multiple cancer-associated processes in CRC cells:
Cell proliferation was markedly reduced in CPNE7 knockdown cells, which showed approximately 90% growth at 72 hours compared to 300% in control cells
Cell migration capacity was reduced to 65% of control levels following CPNE7 inhibition
Invasion capability decreased to approximately 50% in CPNE7-suppressed cells
Wound healing assays demonstrated impaired cell motility, with CPNE7 knockdown cells showing incomplete wound closure (96%) at 48 hours, while control cells achieved complete closure
These findings collectively suggest that CPNE7 plays a crucial role in promoting the aggressive behavior of colorectal cancer cells, contributing to their proliferative, migratory, and invasive capabilities.
Mechanistic investigations have revealed that CPNE7 promotes colorectal tumorigenesis through specific molecular interactions and signaling pathways:
CPNE7 interacts with NONO (Non-POU domain-containing octamer-binding protein) to initiate ZFP42 transcription, thereby promoting CRC progression
CPNE7 influences epithelial-mesenchymal transition (EMT), a critical process in cancer metastasis. Suppression of CPNE7 leads to increased expression of epithelial genes such as E-cadherin (CDH1) and Collagen A1, with concurrent decreased expression of mesenchymal genes
The identification of CPNE7's role in cancer progression has revealed potential therapeutic strategies:
Delivery of CPNE7 shRNA has been demonstrated to hinder tumor growth in vivo
Small molecules such as gramicidin, which block the interaction between CPNE7 and NONO, also inhibit tumor growth in vivo
These findings suggest that targeting CPNE7 or its molecular interactions represents a promising approach for CRC treatment. The CPNE7-NONO-ZFP42 axis has been proposed as a novel potential therapeutic target for colorectal cancer .
As our understanding of CPNE7 and its roles in normal physiology and disease continues to evolve, several promising avenues for future research emerge:
The continued refinement of CPNE7 antibodies with enhanced specificity, sensitivity, and application versatility will further advance research in this field. Future developments may include:
Antibodies targeting specific CPNE7 domains or post-translational modifications
Monoclonal antibodies with increased specificity for particular epitopes
Novel conjugates for expanded detection capabilities
Antibodies optimized for specific applications such as super-resolution microscopy or chromatin immunoprecipitation
While current research has focused predominantly on colorectal cancer, the potential role of CPNE7 in other cancer types warrants investigation. CPNE7 antibodies will be essential tools in these exploratory studies to determine whether the oncogenic functions observed in CRC extend to other malignancies.
The established oncogenic role of CPNE7 in colorectal cancer suggests potential for therapeutic intervention. Future research may focus on:
Development of specific inhibitors targeting CPNE7 or its interactions
Refinement of RNA interference approaches for CPNE7 suppression
Exploration of combination therapies targeting CPNE7 alongside established treatment modalities
CPNE7 (Copine-7) is a calcium-dependent phospholipid-binding protein that plays critical roles in calcium-mediated intracellular processes. It belongs to the copine family, which includes other members such as CPNE1, CPNE3, and CPNE6 . The significance of CPNE7 in cancer research stems from its differential expression and functional impact across multiple cancer types. In colorectal cancer (CRC), CPNE7 is significantly upregulated in tumor tissues compared to normal tissues, with expression patterns correlating with disease progression and patient survival outcomes . While initially characterized as a potential tumor suppressor gene in some cancers like breast cancer, recent evidence demonstrates its oncogenic role in colorectal cancer and oral squamous cell carcinoma, highlighting its context-dependent functionality in different cancer types . This dual nature makes CPNE7 an intriguing target for cancer biology investigations.
CPNE7 expression demonstrates significant variation across colorectal cancer cell lines compared to normal colon cells. Quantitative RT-PCR analysis reveals that CPNE7 expression is consistently higher in colorectal cancer cell lines (SW480, SW620, HCT116, HT29) compared to the normal colon fibroblast cell line CCD-18-co . Among the cancer cell lines, SW480 and HCT116 exhibit particularly elevated expression levels. Western blotting confirms these findings at the protein level, with all tested CRC cell lines showing high CPNE7 protein expression . This differential expression pattern provides a molecular basis for studying CPNE7's role in colorectal carcinogenesis and validates the relevance of these cell line models for CPNE7-focused research. These expression patterns also suggest that colorectal cancer cells may depend on elevated CPNE7 levels for maintaining their malignant phenotype.
CPNE7 exhibits complex subcellular localization patterns that vary by cell type and experimental conditions. While traditionally classified as a membrane protein due to its phospholipid-binding properties, immunohistochemical staining of colorectal cancer patient tissues reveals CPNE7 presence in the cytoplasm and nucleus as well as the cell membrane . Western blot analysis using cellular fractionation confirms this diverse localization pattern, with CPNE7 detected in both nuclear and cytoplasmic fractions of HeLa cells . This multifaceted distribution pattern suggests CPNE7 may perform distinct functions in different cellular compartments, potentially interacting with varied binding partners depending on its localization. When designing immunofluorescence experiments with CPNE7 antibodies, researchers should anticipate detecting signals across multiple cellular compartments and consider using co-localization studies with compartment-specific markers to fully characterize CPNE7's distribution patterns in their specific experimental system.
CPNE7 functions as a positive regulator of epithelial-mesenchymal transition (EMT) in colorectal cancer, as evidenced by gene expression changes following CPNE7 knockdown. When CPNE7 is suppressed in CRC cell lines, significant alterations occur in key EMT markers: epithelial markers E-cadherin and Collagen A1 increase dramatically (by approximately 600% and 1800%, respectively), while mesenchymal markers generally decrease . Specifically, EMT-associated transcription factors ZEB1 and ZEB2 decrease by approximately 1100% and 250% respectively, while MMP2, MMP3, and SNAIL show approximately 40% reduction in expression . These changes indicate that CPNE7 normally promotes the mesenchymal phenotype in CRC cells. The mechanism likely involves CPNE7's participation in signaling pathways that regulate these EMT genes, though the precise molecular interactions require further elucidation. Researchers investigating EMT mechanisms should consider CPNE7 as a potential upstream regulator and may benefit from examining its interaction with known EMT pathway components such as Wnt/β-catenin or TGF-β signaling pathways.
Multiple experimental approaches have demonstrated efficacy for CPNE7 knockdown studies, with both transient and stable suppression systems yielding consistent functional effects. Research shows successful CPNE7 suppression using:
siRNA-mediated transient knockdown: Achieved greater than 80% reduction in CPNE7 expression across multiple colorectal cancer cell lines, with SW480 showing the most significant mRNA reduction and SW620 exhibiting the most pronounced protein level reduction .
shRNA-mediated stable knockdown: Generated through viral infection and selection, providing sustained CPNE7 suppression for long-term studies including in vivo experiments .
CPNE7 expression demonstrates significant negative correlation with patient survival in colorectal cancer, as evidenced by comprehensive Kaplan-Meier survival analysis. Data from approximately 250 CRC patients shows that those with high CPNE7 expression exhibit markedly worse survival outcomes compared to those with low expression . At the 40-month follow-up point, approximately 58% of patients with high CPNE7 expression had died, compared to only 27% in the low-expression group . The survival disparity becomes even more pronounced at longer follow-up periods, with 120-month data showing virtually complete mortality in the high-expression group (127 deaths out of 128 patients) versus significantly better survival in the low-expression cohort (3 deaths out of 123 patients) . Statistical analysis confirms this correlation is significant (p=0.048 for univariate and p=0.032 for multivariate analysis) . Importantly, CPNE7 expression also correlates with lymphatic invasion (p=0.003), suggesting a potential biological mechanism for its impact on survival through promotion of metastatic spread . This survival correlation indicates CPNE7 may serve as a valuable prognostic biomarker in CRC and justifies further investigation into therapeutic strategies targeting CPNE7 or its downstream pathways.
Comprehensive validation of CPNE7 antibodies requires a multi-platform approach to ensure specificity and reliability across different experimental applications. Based on current research protocols, the following validation workflow is recommended:
Western blot validation:
Test the antibody against both recombinant CPNE7 protein and endogenous CPNE7 from cell lysates
Include positive controls (cells known to express CPNE7, such as SW480 or HCT116)
Include negative controls (CPNE7 knockdown cells or non-expressing cells like CCD-18-co)
Verify the detected band matches the predicted molecular weight (approximately 70 kDa)
Immunohistochemistry validation:
Specificity validation:
Application-specific optimization:
These rigorous validation steps ensure reliable antibody performance across experimental platforms and strengthen the validity of subsequent research findings.
Designing robust CPNE7 functional studies requires careful consideration of several technical factors to ensure reliable and reproducible results. Key considerations include:
Cell model selection:
Choose cell lines with varying baseline CPNE7 expression levels (e.g., SW480 and HCT116 for high expression; CCD-18-co for low expression)
Consider patient-derived primary cells to complement established cell lines
For in vivo studies, both knockout mouse models and xenograft models have proven effective
Knockdown/overexpression strategies:
For transient effects, siRNA approaches achieve >80% knockdown efficiency
For stable manipulation, shRNA or CRISPR-Cas9 systems provide sustained alteration
Rescue experiments with wild-type CPNE7 are essential to confirm specificity of observed phenotypes
Inducible expression systems help study dose-dependent effects
Functional assay selection:
Proliferation: Multiple timepoints (24h, 48h, 72h) are necessary to capture growth kinetics
Migration/invasion: Both Transwell and wound-healing assays provide complementary data
Colony formation: Semi-solid agar assays assess anchorage-independent growth
EMT assessment: Combine morphological analysis with molecular marker quantification (E-cadherin, N-cadherin, etc.)
Controls and normalization:
Include both positive controls (known EMT inducers) and negative controls (non-targeting siRNA)
Normalize cell functional data to baseline proliferation rates
Control for transfection/transduction efficiency in all experiments
Account for cell-cycle effects when interpreting proliferation data
Downstream molecular analysis:
Examine both mRNA (qRT-PCR) and protein (Western blot) levels of CPNE7 and target genes
Consider pathway analysis to identify signaling networks affected by CPNE7 manipulation
Evaluate calcium-dependent effects given CPNE7's calcium-binding properties
Addressing these technical considerations ensures robust experimental design and facilitates meaningful interpretation of CPNE7's functional impact.
Differentiating between CPNE7's direct molecular interactions and its secondary effects on signaling pathways requires sophisticated experimental approaches that establish causality and temporal relationships. Researchers should implement the following strategies:
Temporal analysis of signaling events:
Conduct time-course experiments following CPNE7 manipulation to establish the sequence of molecular changes
Early events (0-6 hours) after CPNE7 knockdown or overexpression are more likely to represent direct effects
Late events (24+ hours) may reflect secondary or compensatory mechanisms
Protein-protein interaction studies:
Conduct co-immunoprecipitation (Co-IP) assays to identify direct binding partners of CPNE7
Perform proximity ligation assays (PLA) to visualize in situ protein interactions
Use mass spectrometry-based interactome analysis to comprehensively identify the CPNE7 protein complex
Domain-specific functional analysis:
Generate CPNE7 mutants lacking specific functional domains (calcium-binding C2 domains or von Willebrand A-like domain)
Test these mutants in rescue experiments to determine which domains are essential for specific functions
Identify critical residues through site-directed mutagenesis
Pathway inhibitor studies:
Use specific inhibitors of EMT-related pathways (e.g., TGF-β, Wnt/β-catenin, NF-κB) in combination with CPNE7 manipulation
Determine if pathway inhibition prevents CPNE7-induced phenotypic changes
This approach helps position CPNE7 within established signaling hierarchies
Direct target validation:
Perform chromatin immunoprecipitation (ChIP) if nuclear localization suggests transcriptional regulatory roles
Use reporter assays with promoter regions of putative target genes
Employ EMSA (Electrophoretic Mobility Shift Assay) to detect direct DNA-protein interactions
By integrating these approaches, researchers can construct a mechanistic model that distinguishes CPNE7's direct molecular actions from downstream consequences, providing clearer insights into its role in complex cellular processes like EMT and cancer progression.
CPNE7 demonstrates significant potential as a prognostic biomarker in colorectal cancer, supported by both univariate and multivariate analyses of patient outcomes. Statistical evidence from a cohort of approximately 250 CRC patients reveals that CPNE7 expression is an independent predictor of patient survival with hazard ratios of 1.54 (p=0.048) in univariate analysis and 1.58 (p=0.032) in multivariate analysis . The table below summarizes key prognostic associations of CPNE7 expression:
Developing therapeutic strategies targeting CPNE7 presents several significant challenges that span from basic molecular understanding to practical drug development considerations:
Target specificity challenges:
Functional complexity:
CPNE7 demonstrates context-dependent functions (oncogenic in colorectal cancer but potentially tumor-suppressive in other cancers)
Therapeutic inhibition might have unpredictable effects in different tissues
CPNE7 knockout mice show developmental defects in dental tissues, suggesting potential for systemic toxicity
Molecular targeting approaches:
As a protein without enzymatic activity, CPNE7 lacks obvious catalytic sites for small molecule inhibition
Protein-protein interaction disruption requires identification of critical binding interfaces
Calcium-binding disruption may affect numerous physiological processes beyond CPNE7
Delivery challenges:
Patient selection considerations:
Need for predictive biomarkers to identify patients most likely to benefit from CPNE7 targeting
Development of companion diagnostics to quantify CPNE7 expression levels
Understanding resistance mechanisms to CPNE7-targeted therapies
Validation requirements:
Demonstration of target engagement in vivo
Correlation between CPNE7 inhibition and therapeutic efficacy
Development of pharmacodynamic markers for clinical trials
Addressing these challenges requires collaborative efforts between structural biologists, medicinal chemists, and cancer biologists to develop targeted approaches that can effectively modulate CPNE7 function while minimizing off-target effects and toxicity.
Several cutting-edge technologies hold promise for elucidating CPNE7's complex functions in cancer biology, enabling researchers to address current knowledge gaps with unprecedented precision:
Single-cell multi-omics approaches:
Single-cell RNA sequencing can reveal heterogeneity in CPNE7 expression within tumors
Integration with spatial transcriptomics would map CPNE7 expression patterns within the tumor microenvironment
Single-cell proteomics could identify cell populations where CPNE7 exerts its strongest effects
These approaches would clarify whether CPNE7's oncogenic effects are universal or cell-type specific
Advanced protein structural analysis:
Cryo-electron microscopy could resolve CPNE7's structure in different calcium-binding states
Hydrogen-deuterium exchange mass spectrometry would identify dynamic regions involved in protein interactions
AlphaFold2 and other AI-based structure prediction tools could model CPNE7 complexes with potential partners
These structural insights would facilitate rational design of CPNE7 inhibitors
Genome and epigenome editing technologies:
CRISPR-Cas9 screening approaches could identify synthetic lethal interactions with CPNE7
Base editing technologies allow introduction of specific CPNE7 mutations to assess their functional impact
Epigenome editing could investigate regulatory mechanisms controlling CPNE7 expression
These genetic tools would establish causal relationships between CPNE7 and downstream effects
Advanced in vivo models:
Patient-derived organoids expressing variable CPNE7 levels would provide physiologically relevant models
Humanized mouse models could better recapitulate CPNE7's effects in human cancer
CRISPR-engineered animal models with tissue-specific CPNE7 manipulation would clarify its context-dependent roles
These models would bridge the gap between in vitro findings and clinical observations
Calcium signaling visualization techniques:
Genetically encoded calcium indicators combined with CPNE7 fluorescent tagging would enable real-time visualization of CPNE7 dynamics during calcium fluctuations
Optogenetic tools could trigger calcium release to study immediate CPNE7 responses
These approaches would illuminate CPNE7's calcium-dependent functions in living cells
Integrating these advanced technologies would provide unprecedented insights into CPNE7's multifaceted roles in cancer biology and potentially reveal new therapeutic vulnerabilities in CPNE7-expressing tumors.
The translation of CPNE7 research findings from colorectal cancer to other cancer types requires systematic comparative analysis and consideration of tissue-specific factors. Several approaches and considerations are essential for this translation:
Cross-cancer expression analysis:
Comprehensive profiling of CPNE7 expression across cancer types using multi-omics databases (TCGA, ICGC)
Correlation of expression patterns with clinical outcomes in each cancer type
Identification of cancer types with similar CPNE7 expression patterns to CRC
Functional conservation assessment:
Validation of CPNE7's oncogenic functions in cell line models representing diverse cancer types
Comparison of phenotypic changes following CPNE7 manipulation across cancer types
Evaluation of whether CPNE7-regulated EMT processes are consistent across different tissues of origin
Mechanistic pathway comparison:
Analysis of whether CPNE7 interacts with the same downstream effectors across cancer types
Identification of tissue-specific binding partners through comparative interactome studies
Determination if calcium-dependent functions of CPNE7 are universally conserved
Context-dependent effects:
Investigation of potential tumor-suppressive roles in certain cancer types versus oncogenic roles in others
Analysis of how tissue-specific microenvironments might influence CPNE7 function
Consideration of how different genetic landscapes might modify CPNE7's impact
Therapeutic implications:
Assessment of whether CPNE7-targeting strategies developed for CRC could apply to other cancers
Identification of cancer types most likely to benefit from CPNE7 modulation
Development of biomarkers to predict responsiveness to CPNE7-targeted therapies across cancer types
This translational approach would determine whether CPNE7 represents a broad oncogenic driver across multiple cancers or if its functions are context-dependent, guiding the development of targeted therapeutic strategies with appropriate patient selection criteria.
Incorporating CPNE7 analysis in cancer research requires methodological rigor and attention to technical details. Based on current evidence, the following best practices are recommended:
Experimental design considerations:
Include paired tumor and normal tissues when analyzing CPNE7 expression in patient samples
Utilize multiple cancer and normal cell lines to account for biological variability
Design time-course experiments to capture both immediate and delayed effects of CPNE7 manipulation
Implement both gain-of-function and loss-of-function approaches for comprehensive functional assessment
Technical methodology recommendations:
For protein detection: Use validated antibodies with demonstrated specificity against CPNE7 (e.g., mouse monoclonal antibodies at 1 μg/mL concentration for Western blotting)
For gene expression analysis: Design primers spanning exon junctions to prevent genomic DNA amplification
For knockdown studies: Compare siRNA and shRNA approaches, validating knockdown efficiency at both mRNA and protein levels
For functional assays: Implement multiple complementary assays (proliferation, migration, invasion, colony formation) to comprehensively assess phenotypic changes
Data analysis and interpretation:
Stratify patient cohorts by CPNE7 expression levels using clearly defined thresholds
Perform both univariate and multivariate analyses when correlating CPNE7 with clinical outcomes
Consider calcium signaling context when interpreting CPNE7 functional data
Analyze EMT markers comprehensively, including both epithelial (E-cadherin, Collagen A1) and mesenchymal (N-cadherin, ZEB1/2, SNAIL) markers
Reporting standards:
Clearly document antibody validation procedures, including specificity controls
Report full experimental conditions including cell density, passage number, and culture conditions
Include comprehensive methods descriptions enabling reproducibility
Document statistical approaches and significance thresholds