PCDH17 Antibody is a polyclonal antibody targeting the PCDH17 protein, which belongs to the cadherin superfamily. It is primarily used in Western blotting (WB) and immunofluorescence (IF) to study PCDH17’s role in cancer, neurodevelopment, and cell adhesion .
Target: PCDH17 protein (molecular weight: ~126 kDa, observed as 160–170 kDa or 150 kDa in WB due to post-translational modifications) .
Host: Rabbit or goat polyclonal antibodies, depending on the manufacturer .
PCDH17 Antibody is validated for diverse experimental approaches, with species-specific reactivity and optimized dilution ranges.
Application | Details | Citation |
---|---|---|
Western Blot | Detects PCDH17 in lysates | |
ELISA | Used in immunoassays | |
Immunofluorescence | Visualizes protein localization |
Species | Antibody Source | Observed Band | Dilution |
---|---|---|---|
Mouse | Proteintech (Rabbit) | 160–170 kDa | 1:500–1:1000 |
Mouse | Abcam (Goat) | 150 kDa | 0.2 µg/mL |
Rat | Proteintech (Rabbit) | 160–170 kDa | 1:500–1:1000 |
Human | Proteintech (Rabbit) | 160–170 kDa | 1:500–1:1000 |
Parameter | Details |
---|---|
Immunogen | PCDH17 fusion protein (Ag12322) |
Purification | Antigen affinity chromatography |
Storage | PBS with 0.02% sodium azide, -20°C |
Conjugate | Unconjugated |
Parameter | Details |
---|---|
Immunogen | Synthetic peptide (aa 1050–1150) |
Purification | Not specified |
Storage | Aliquots stored at -20°C |
Conjugate | Unconjugated |
PCDH17 Antibody has been instrumental in studying PCDH17’s role in cancer biology and therapeutic resistance.
Breast Cancer: Overexpression of PCDH17 via antibody-validated studies suppressed Wnt/β-catenin signaling, reduced metastasis, and induced apoptosis in xenograft models .
Colorectal Cancer: PCDH17 expression correlated with 5-fluorouracil (5-FU) sensitivity, as PCDH17 upregulation enhanced autophagy and apoptosis in resistant cells .
Protocadherin 17 (PCDH17) is a 120-160 kDa glycoprotein belonging to the δ2-group of the protocadherin subfamily within the cadherin family of molecules. It functions primarily as a cell adhesion molecule expressed on endothelium and various epithelia, including stratified squamous epithelium and gastric columnar epithelium . PCDH17 has gained significant research interest due to its roles in multiple biological processes, including neuronal development, cell cycle regulation, cell migration inhibition, and its implications in various pathological conditions . Research has demonstrated PCDH17's involvement in cancer progression, particularly colorectal cancer sensitivity to chemotherapy, as well as its roles in neuropsychiatric conditions including major mood disorders .
PCDH17 is a type I transmembrane protein with a molecular weight ranging from 120-160 kDa, though the calculated molecular weight is approximately 126 kDa (1159 amino acids) . The mature human PCDH17 protein contains:
A 690 amino acid extracellular domain (ECD) spanning residues 18-707
Six cadherin domains within the ECD (residues 18-695)
A 431 amino acid cytoplasmic region
At least two splice variants involving substitutions in the cytoplasmic region
PCDH17 is primarily localized to the cell membrane, consistent with its function in cell-cell adhesion . The protein undergoes post-translational modifications, particularly glycosylation, which accounts for the discrepancy between its calculated molecular weight (126 kDa) and observed molecular weight (160-170 kDa) in Western blot analyses .
Based on current research protocols, PCDH17 antibodies have been validated for the following applications:
It is strongly recommended that each laboratory determine optimal dilutions for their specific experimental systems, as antibody performance can vary based on sample type, detection method, and experimental conditions . When using PCDH17 antibodies in Western blot applications, researchers should expect to detect bands at approximately 120-170 kDa, depending on the sample type and specific antibody used .
For optimal Western blot detection of PCDH17, researchers should follow these methodological guidelines:
Sample preparation: Brain tissue (particularly cerebellum or cortex) provides strong PCDH17 signal in mouse and rat models . Lysates should be prepared under reducing conditions.
Recommended protocol:
Expected results: PCDH17 should be detected at approximately 160-170 kDa in human samples and 120-160 kDa in mouse and rat brain tissues . The specific band size may vary slightly depending on tissue type and post-translational modifications.
Controls: Include positive controls such as mouse brain cortex or rat brain cerebellum tissues, which have been validated to express PCDH17 . Negative controls should include tissues known not to express PCDH17 or siRNA knockdown samples.
To preserve antibody performance and ensure experimental reproducibility, PCDH17 antibodies should be handled according to these storage guidelines:
Long-term storage: Store antibodies at -20°C to -70°C. Most PCDH17 antibodies remain stable for up to 12 months from the date of receipt when stored properly .
Working solution preparation:
Short-term storage:
Buffer conditions: Most PCDH17 antibodies are provided in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3, which helps maintain stability . Some smaller-volume preparations may contain 0.1% BSA as a stabilizer .
Validation of antibody specificity is critical for obtaining reliable results. For PCDH17 antibodies, consider these validation approaches:
Positive and negative controls:
Molecular approaches:
siRNA or shRNA knockdown of PCDH17 in cell lines expressing the protein should reduce or eliminate the signal
Overexpression systems can confirm the molecular weight of the detected protein
CRISPR/Cas9 knockout cells provide definitive negative controls
Cross-reactivity assessment:
Application-specific validation:
For Western blot, verify the band appears at the expected molecular weight (160-170 kDa)
For immunohistochemistry, compare staining patterns with published literature on PCDH17 expression
Research has identified PCDH17 as a potential tumor suppressor and modulator of chemotherapy sensitivity, particularly in colorectal cancer (CRC). Methodological approaches using PCDH17 antibodies include:
Expression correlation studies:
Mechanistic studies of chemosensitivity:
Investigate the relationship between PCDH17 expression and apoptotic markers following 5-FU treatment
Examine JNK pathway activation (a key mechanism in PCDH17-induced autophagy) using phospho-specific antibodies alongside PCDH17 detection
Use antibodies to monitor autophagic flux in cells with manipulated PCDH17 expression
Prognostic marker development:
Research has demonstrated that PCDH17 increases CRC sensitivity to 5-FU by inducing both apoptosis and JNK-dependent autophagic cell death, making it a potential prognostic marker for predicting 5-FU sensitivity in CRC patients .
PCDH17 has been implicated in neuronal development, synaptogenesis, and psychiatric conditions, particularly major mood disorders. Advanced research approaches include:
Structural and functional neuroimaging correlation:
Use PCDH17 antibodies in postmortem tissue studies to correlate protein expression with neuroimaging findings
Examine PCDH17 expression in specific neuronal populations, particularly in the amygdala where PCDH17 is expressed in a subset of neurons
Correlate PCDH17 levels with amygdala volume and function as identified in imaging studies
Synaptic development analysis:
Use PCDH17 antibodies to study dendritic spine morphology and density in primary neuronal cultures
Combine with transfection of PCDH17 variants to study effects of overexpression on synaptic development
Research has shown that elevated expression of PCDH17 in primary neuronal cultures results in decreased spine density and aberrant dendritic morphology, which may underlie synaptic dysfunction in mood disorders
Genetic-molecular correlations:
Examine how PCDH17 risk alleles (identified in genome-wide association studies) affect protein expression in postmortem brain samples
Risk alleles have been shown to predict higher PCDH17 mRNA levels, consistent with increased expression in patients with bipolar disorder compared to healthy controls
Animal model studies:
PCDH17 exists in multiple isoforms due to alternative splicing, particularly in the cytoplasmic region. Advanced detection approaches include:
Isoform-specific antibody selection:
Choose antibodies raised against specific regions that differ between isoforms
For human PCDH17, at least two splice variants have been identified that involve substitutions in the cytoplasmic region (a 14 amino acid substitution in one variant and a 25 amino acid substitution in another, both affecting residues 876-1159)
Molecular weight discrimination:
Use high-resolution SDS-PAGE (6-8% gels) to separate closely migrating isoforms
Western blot analysis can reveal subtle differences in molecular weight between isoforms
Combine with isoform-specific antibodies for definitive identification
RT-PCR validation:
Design primers specific to different splice junctions
Use RT-PCR to confirm the presence of specific mRNA isoforms
Correlate protein detection with mRNA expression patterns
Mass spectrometry confirmation:
For definitive isoform identification, immunoprecipitate PCDH17 using the antibody
Analyze the precipitated proteins by mass spectrometry to identify specific peptide sequences unique to each isoform
When working with PCDH17 antibodies in Western blot applications, researchers may encounter several technical challenges:
High molecular weight detection issues:
Problem: Difficulty detecting the high molecular weight PCDH17 protein (160-170 kDa)
Solution: Use lower percentage gels (6-8%), extend transfer time, and employ pulsed or high-molecular-weight transfer protocols
Multiple bands or smearing:
Problem: Detection of multiple bands or smearing around the expected molecular weight
Cause: Post-translational modifications (particularly glycosylation), protein degradation, or non-specific binding
Solution: Include protease inhibitors during sample preparation, optimize primary antibody dilution (1:500-1:1000), and consider using freshly prepared tissue samples
Weak signal:
Problem: Low or undetectable signal despite proper sample preparation
Solution: Increase antibody concentration, extend primary antibody incubation time (overnight at 4°C), use more sensitive detection methods, or increase protein loading (50-100 μg of total protein is often needed for endogenous detection)
High background:
Problem: Non-specific staining obscuring specific PCDH17 signal
Solution: Increase blocking time/concentration, optimize antibody dilution, include additional wash steps, and consider using TBS instead of PBS for antibodies sensitive to phosphate buffers
Different sample types require specific optimization strategies for PCDH17 detection:
Brain tissue samples (highest endogenous expression):
Cell line samples:
Transient transfection with PCDH17 expression constructs may be necessary for cell lines with low endogenous expression
For endogenous detection, neuronal cell lines typically show higher expression levels
Confluency can affect expression levels; standardize cell culture conditions
Consider using concentrated protein samples (e.g., through immunoprecipitation) for low-expressing lines
Human patient samples:
Sample-specific recommendations:
Researchers often encounter discrepancies between PCDH17 mRNA levels and protein detection, which can be addressed through these methodological approaches:
Protein stability assessment:
PCDH17 protein may have different half-lives across tissue types
Use protein synthesis inhibitors (e.g., cycloheximide) to determine protein turnover rates
Compare with mRNA stability using actinomycin D treatment
Post-transcriptional regulation:
Investigate the role of microRNAs in regulating PCDH17 translation
Examine RNA-binding proteins that might affect PCDH17 mRNA stability or translation
Consider alternative splicing events that might affect antibody binding sites
Technical considerations:
Ensure the antibody epitope matches the specific PCDH17 isoform being studied
Use multiple antibodies targeting different regions of PCDH17
Combine protein detection methods (Western blot, immunoprecipitation, immunohistochemistry) to validate results
Biological variability:
PCDH17's role in neuronal development and synaptogenesis makes it relevant for neurodevelopmental research:
Developmental expression profiling:
Synaptic function studies:
Investigate PCDH17's role in synapse formation and maintenance
Combine antibody labeling with synaptic markers to assess colocalization
Research has shown that elevated PCDH17 expression leads to decreased spine density and aberrant dendritic morphology, suggesting its importance in synaptic development
Circuit formation analysis:
Use PCDH17 antibodies to identify specific neuronal populations during circuit development
Combine with tract-tracing studies to understand connectivity patterns
Assess how PCDH17 expression correlates with functional circuit establishment
Neurodevelopmental disorder research:
Examine PCDH17 expression in models of neurodevelopmental disorders
Investigate potential disruptions in expression patterns or localization
Consider PCDH17 as a potential biomarker for developmental abnormalities
While PCDH17's role in colorectal cancer has been well-studied, emerging research suggests broader implications in oncology:
Tumor suppressor function:
Use PCDH17 antibodies to assess protein expression across various cancer types
Correlate expression levels with clinical outcomes and tumor characteristics
Investigate mechanisms of PCDH17 downregulation, particularly through promoter methylation
Cell migration and invasion studies:
Therapeutic response prediction:
Mechanistic pathway studies:
As research on PCDH17 continues to evolve, several cutting-edge technologies show promise for advancing our understanding:
CRISPR/Cas9 genome editing:
Generate precise PCDH17 knockout or knock-in models
Create tagged versions of endogenous PCDH17 for localization studies
Introduce specific mutations corresponding to human variants
Super-resolution microscopy:
Examine PCDH17 localization at synapses with nanometer precision
Study dynamic changes in PCDH17 distribution during neuronal development
Investigate co-localization with other synaptic proteins
Single-cell approaches:
Combine single-cell RNA sequencing with protein detection
Identify cell populations with unique PCDH17 expression patterns
Correlate with functional characteristics of specific neuronal subtypes
Proximity labeling techniques:
Use BioID or APEX2 fusion proteins to identify PCDH17 interacting partners
Map the PCDH17 interactome in different cellular contexts
Discover novel functional relationships
The multifaceted roles of PCDH17 in development and disease suggest several therapeutic avenues:
Cancer treatment strategies:
Neuropsychiatric disorder therapeutics:
Regenerative medicine applications:
Investigate PCDH17's potential role in guiding neuronal growth and connectivity
Explore its use in promoting appropriate synaptic development in regenerative approaches
Study its function in neuronal differentiation for stem cell-based therapies
Biomarker development:
Despite significant advances, several important questions about PCDH17 remain unanswered:
Structural and functional relationships:
How do specific domains of PCDH17 contribute to its diverse functions?
What are the binding partners of PCDH17 in different cellular contexts?
How does the cytoplasmic domain mediate intracellular signaling?
Regulatory mechanisms:
What controls PCDH17 expression during development and disease?
How is PCDH17 trafficking and localization regulated?
What post-translational modifications affect PCDH17 function?
Cell type-specific functions:
Translational research needs:
Validation of PCDH17 as a therapeutic target or biomarker across larger patient populations
Development of standardized assays for clinical implementation
Establishment of animal models that accurately recapitulate human PCDH17-related pathologies