PCDH17 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to PCDH17 and Its Antibodies

PCDH17 (Protocadherin 17), a 126 kDa calcium-dependent cell adhesion protein, belongs to the cadherin family and plays critical roles in neuronal connectivity and epithelial integrity . Its dysregulation is linked to cancers, including colorectal and breast, where it acts as a tumor suppressor . Antibodies targeting PCDH17 are essential for studying its expression, subcellular localization, and functional mechanisms.

HRP-conjugated PCDH17 antibodies combine the specificity of anti-PCDH17 antibodies with horseradish peroxidase (HRP), an enzyme that catalyzes oxidative reactions for colorimetric detection in assays like ELISA and Western blot. These conjugates enhance sensitivity and streamline workflows in immunodetection .

Overview of HRP-Conjugated PCDH17 Antibodies

HRP-conjugated PCDH17 antibodies are engineered for high-performance detection in enzymatic assays. Key features include:

FeatureDetails
Host/IsotypeRabbit polyclonal (e.g., United States Biological, CUSABIO)
ReactivityPrimarily human (e.g., United States Biological, CUSABIO)
ApplicationsELISA, Western blot (WB), and immunohistochemistry (IHC)
Conjugation AdvantageEnables enzymatic detection via HRP-substrate reactions (e.g., TMB, DAB)
Molecular WeightDetects PCDH17 at 160–170 kDa (observed), likely due to glycosylation

These antibodies are validated for specificity and sensitivity, with dilutions typically optimized per assay (e.g., 1:500–1:1000 for WB) .

Key Applications

ApplicationPurposeExample Protocol
ELISAQuantitative detection of PCDH17 in lysates or serumHRP-antibody binds PCDH17; TMB substrate adds color
Western BlotAnalysis of PCDH17 expression in tissue/cell lysatesHRP enables chemiluminescent detection
ImmunohistochemistryLocalization of PCDH17 in paraffin-embedded or frozen tissue sectionsHRP-DAB staining for visualization

Research Highlights

  1. Cancer Biology

    • PCDH17 antibodies identified its tumor-suppressive role in colorectal cancer (CRC), correlating with 5-fluorouracil (5-FU) sensitivity. High PCDH17 expression promoted apoptosis and autophagy, enhancing chemotherapeutic efficacy .

    • In breast cancer, PCDH17 promoter methylation linked to downregulation, suggesting epigenetic silencing drives tumorigenesis .

  2. Mechanistic Insights

    • PCDH17 modulates autophagy via JNK pathway activation, as shown in CRC models. Autophagy inhibitors (e.g., chloroquine) reversed PCDH17-induced cell death .

    • Antibodies confirmed PCDH17’s cytoplasmic localization in cancer tissues, aligning with its role in cell adhesion and signaling .

Table 2: PCDH17 Antibody Validation in Research

StudyMethodFindings
(Colorectal Cancer)IHC, WBPCDH17 overexpression correlated with 5-FU sensitivity via autophagy induction
(Breast Cancer)MSP, IHCPromoter methylation silences PCDH17 in 89% of primary tumors

Challenges and Considerations

  • Limited Cross-Reactivity: Most HRP-conjugated PCDH17 antibodies target human PCDH17, limiting cross-species studies .

  • Optimal Dilution: Requires titration for each assay to avoid non-specific binding .

  • Epitope Specificity: Variability in immunogen regions (e.g., AA 39–89, AA 1050–1150) may affect detection outcomes .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on the purchase method or location. For specific delivery timeframes, please consult your local distributor.
Synonyms
Protocadherin-17 (Protocadherin-68), PCDH17, PCDH68 PCH68
Target Names
PCDH17
Uniprot No.

Target Background

Function
PCDH17 is a potential calcium-dependent cell-adhesion protein.
Gene References Into Functions
  1. Methylation of PCDH17 has been linked to the development and progression of high-grade serous ovarian carcinoma (HGSOC), suggesting its potential as a target for novel clinical biomarkers. PMID: 29991130
  2. Serum PCDH17 methylation has emerged as a potential prognostic biomarker for patients undergoing surgery for renal cell carcinoma. PMID: 28688232
  3. PCDH17 acts as a tumor suppressor in breast cancer, inhibiting Wnt/beta-catenin signaling and metastasis. However, its frequent methylation in primary tumors indicates potential as a biomarker for the disease. PMID: 27351130
  4. Aberrant methylation of protocadherin 17 has been associated with acute lymphoblastic leukemia. PMID: 27643535
  5. PCDH-17 has been shown to inhibit metastasis through the EGFR/MEK/ERK signaling pathway. PMID: 26386721
  6. Combined methylation analysis of POU4F2/PCDH17 has demonstrated high sensitivity and specificity (90.00% and 93.96%, respectively) for detecting bladder cancer across various pathological sample groups. PMID: 26700620
  7. PCDH17 methylation in serum is a frequent occurrence in early-stage prostate cancer and serves as an independent predictor of biochemical recurrence after radical prostatectomy. PMID: 26683656
  8. PCDH17 methylation is more prevalent in clear cell renal cell carcinoma patients and is associated with malignant clinicopathological characteristics and a poorer prognosis. PMID: 26404644
  9. PCDH17 promoter methylation is strongly associated with bladder cancer malignancy and can serve as an independent predictor of clinical outcomes in patients with this disease. PMID: 24567353
  10. PCDH17 promoter methylation is significantly linked to malignant behavior and poor prognosis in patients with bladder cancer. PMID: 24366498
  11. Research suggests that PCDH17 plays a crucial role in the synaptic development of specific corticobasal ganglia circuits and may be involved in these circuits in depressive behaviors. PMID: 23684785
  12. PCDH17 acts as a tumor suppressor, suppressing proliferation through apoptosis and autophagy induction. It is frequently silenced in gastric and colorectal cancers. PMID: 22926751
  13. Studies have shown that PCDH17 is transcriptionally downregulated in gastric cancer due to aberrant promoter CpG island methylation. PMID: 22207556
  14. A statistically significant downregulation of PCDH17/PCH68 and PTPRD has been observed. PMID: 21213369
  15. Findings suggest that silencing of PCDH17 expression through promoter hypermethylation or other mechanisms leads to the loss of its tumor-suppressive activity, potentially contributing to the carcinogenesis of a subgroup of ESCCs. PMID: 20200074
  16. Downregulation of CDH18 and PCDH17 has been observed in azoospermic testis. PMID: 20180417
  17. The occurrence of an unusual TG 3' splice site in intron 2 has been discussed. PMID: 17672918

Show More

Hide All

Database Links

HGNC: 14267

OMIM: 611760

KEGG: hsa:27253

UniGene: Hs.106511

Subcellular Location
Cell membrane; Single-pass type I membrane protein.

Q&A

What is PCDH17 and why is it significant in research?

PCDH17 (Protocadherin 17) is a 1,159 amino acid single-pass type I membrane protein containing six cadherin domains. It functions as a calcium-dependent cell adhesion protein that plays a crucial role in establishing cell-cell connections within brain tissue. The gene encoding PCDH17 maps to human chromosome 13, which houses over 400 genes, including BRCA2 and RB1, and comprises nearly 4% of the human genome . Recent research has revealed that PCDH17 functions as a tumor suppressor in colorectal cancer (CRC) and is frequently methylated in these cancers . The significance of PCDH17 in research has grown substantially as studies demonstrate its potential as a prognostic marker, particularly for predicting 5-fluorouracil (5-FU) sensitivity in CRC patients .

What are the recommended applications and dilutions for PCDH17 Antibody, HRP conjugated?

The PCDH17 Antibody, HRP conjugated is versatile for multiple research applications with specific recommended dilutions for optimal results. For Western Blot applications, a dilution range of 1:100-1000 is recommended to achieve optimal signal-to-noise ratio. For immunohistochemistry on paraffin-embedded tissues (IHC-P), a dilution range of 1:100-500 is suggested . These recommendations serve as starting points, and researchers should perform optimization procedures for their specific experimental conditions, including antibody titration, blocking optimization, and incubation time adjustments. The antibody demonstrates reactivity across human, mouse, and rat samples, making it suitable for comparative studies across these species .

How should PCDH17 Antibody, HRP conjugated be stored and handled to maintain optimal activity?

To maintain optimal activity of PCDH17 Antibody, HRP conjugated, proper storage and handling procedures are critical. The antibody should be stored at -20°C for long-term preservation and at 4°C for short-term use (1-2 weeks). Avoid repeated freeze-thaw cycles, as these can significantly degrade the antibody and reduce its binding efficiency. When working with the antibody, aliquot into single-use volumes before freezing to prevent the need for multiple thaws. Always centrifuge the antibody vial briefly before opening to collect liquid at the bottom of the tube. For dilutions, use sterile buffers with pH compatibility for HRP activity (typically pH 6.8-7.5). Minimize exposure to light, as HRP conjugates can be photosensitive. During experimental procedures, maintain samples at appropriate temperatures (typically 4°C for storage of diluted antibody and room temperature for incubations) to ensure consistent performance across experiments.

What controls should be implemented when using PCDH17 Antibody, HRP conjugated in Western blot experiments?

When designing Western blot experiments with PCDH17 Antibody, HRP conjugated, a comprehensive control strategy should be implemented to ensure reliable and interpretable results. Include the following controls:

  • Positive Control: Cell lines or tissues with known PCDH17 expression (e.g., specific neural tissues or colorectal cancer cell lines with confirmed PCDH17 expression).

  • Negative Control: Samples lacking PCDH17 expression or samples from PCDH17 knockout models.

  • Loading Control: Use housekeeping proteins (β-actin, GAPDH, or α-tubulin) to normalize protein loading between samples.

  • Antibody Controls:

    • Primary antibody omission control to assess non-specific binding of the secondary detection system

    • Isotype control (rabbit IgG-HRP at the same concentration) to identify potential non-specific binding

  • Molecular Weight Marker: To confirm the detected band corresponds to the expected molecular weight of PCDH17 (approximately 135-140 kDa).

  • Blocking Peptide Control: Pre-incubation of the antibody with the immunizing peptide should eliminate specific binding and the corresponding signal.

To validate PCDH17 knockdown or overexpression experiments, include samples with confirmed altered expression levels, as demonstrated in studies where PCDH17 expression was modulated using shRNA or overexpression vectors .

What is the optimal protocol for detecting PCDH17 expression in paraffin-embedded tissues using HRP-conjugated antibodies?

For optimal detection of PCDH17 in paraffin-embedded tissues using HRP-conjugated antibodies, the following methodological approach is recommended:

  • Tissue Preparation:

    • Fix tissues in 10% neutral buffered formalin for 24-48 hours

    • Process and embed in paraffin according to standard histological procedures

    • Section tissues at 4-5 μm thickness onto adhesive slides

  • Deparaffinization and Rehydration:

    • Xylene: 3 changes, 5 minutes each

    • 100% ethanol: 2 changes, 3 minutes each

    • 95%, 80%, 70% ethanol: 3 minutes each

    • Distilled water: 5 minutes

  • Antigen Retrieval (critical for PCDH17 detection):

    • Heat-induced epitope retrieval in citrate buffer (pH 6.0) at 95-98°C for 20 minutes

    • Allow slides to cool in buffer for 20 minutes at room temperature

  • Endogenous Peroxidase Blocking:

    • Incubate sections in 3% hydrogen peroxide in methanol for 15 minutes

    • Wash in PBS (3 × 5 minutes)

  • Non-specific Binding Blocking:

    • Incubate with 5% normal goat serum in PBS for 1 hour at room temperature

  • Primary Antibody Incubation:

    • Apply PCDH17 Antibody, HRP conjugated at 1:100-500 dilution

    • Incubate overnight at 4°C in a humidified chamber

    • Wash thoroughly in PBS (3 × 5 minutes)

  • Chromogenic Detection:

    • Apply DAB substrate solution for 5-10 minutes (monitor under microscope for optimal signal)

    • Wash in distilled water

  • Counterstaining and Mounting:

    • Counterstain with hematoxylin for 1-2 minutes

    • Dehydrate through graded alcohols

    • Clear in xylene and mount with permanent mounting medium

This protocol has been validated in studies examining PCDH17 expression in colorectal cancer tissues, where PCDH17 showed predominantly cytoplasmic localization in cancer cells .

How can PCDH17 Antibody, HRP conjugated be used to evaluate autophagy in cancer research?

PCDH17 Antibody, HRP conjugated can be effectively employed to investigate the relationship between PCDH17 expression and autophagy in cancer research through the following methodological approaches:

  • Co-detection of PCDH17 and Autophagy Markers:

    • Use PCDH17 Antibody, HRP conjugated in Western blots alongside autophagy markers like LC3B-II, BECN1 (Beclin-1), and p62/SQSTM1

    • In dual immunofluorescence or sequential IHC, coordinate PCDH17 detection with autophagy markers to assess co-localization or expression correlation

  • Autophagy Flux Analysis:

    • Treat cells with autophagy modulators (e.g., rapamycin, chloroquine) in the presence or absence of PCDH17 expression

    • Monitor changes in LC3B-II turnover and BECN1 expression in relation to PCDH17 levels

    • Use Western blot to quantify these changes with PCDH17 Antibody, HRP conjugated at 1:100-1000 dilution

  • Functional Studies:

    • Implement PCDH17 knockdown or overexpression systems (as performed in HCT116 and SW480 cells)

    • Measure autophagy responses using PCDH17 Antibody to confirm expression changes

    • Correlate PCDH17 expression with autophagy markers and cell death indicators

Research has demonstrated that PCDH17 reexpression in colorectal cancer cells augments 5-FU sensitivity by promoting apoptosis and autophagic cell death. Studies showed that PCDH17 knockdown significantly inhibited LC3B-II turnover and the expression of BECN1, while BECN1 overexpression did not affect PCDH17 expression, indicating that PCDH17 modulates BECN1 and apoptosis .

How does PCDH17 expression correlate with chemosensitivity in colorectal cancer?

PCDH17 expression demonstrates a significant positive correlation with chemosensitivity in colorectal cancer, particularly to 5-fluorouracil (5-FU) treatment. A comprehensive analysis of PCDH17 expression in chemosensitive and chemoresistant CRC tissues revealed several important patterns:

PCDH17 was significantly more highly expressed in 5-FU-sensitive CRC tissues (52.4%, 11/21 cases) compared to 5-FU-resistant tissues (7.7%, 2/39 cases) . This expression pattern was mirrored by BECN1 (autophagy-related protein), which showed high expression in 81% (17/21 cases) of chemosensitive tissues versus 30.8% (12/39 cases) in chemoresistant tissues .

The correlation between PCDH17 expression and clinical parameters revealed that high PCDH17 expression was significantly associated with reduced lymph node metastasis (pN0/1 categories) compared to advanced nodal involvement (pN2) (p=0.0109) . This relationship is detailed in the following table:

Clinical ParameterNHigh PCDH17 (%)Low PCDH17 (%)p value
pN categories
pN0/13111 (35.5)20 (64.5)0.0109
pN2292 (6.9)27 (93.1)
Chemosensitivity
Sensitive2111 (52.4)10 (47.6)<0.0001
Resistant392 (7.7)37 (92.3)

Functionally, restoring PCDH17 expression in CRC cell lines significantly increased their sensitivity to 5-FU treatment, while PCDH17 knockdown attenuated 5-FU sensitivity in a dose-dependent manner . These findings collectively suggest that PCDH17 expression status could serve as a valuable predictive biomarker for 5-FU sensitivity in CRC patients.

What molecular mechanisms underlie the relationship between PCDH17 and autophagy in cancer cells?

The relationship between PCDH17 and autophagy in cancer cells is mediated through a complex molecular signaling cascade primarily involving the c-Jun NH2-terminal kinase (JNK) pathway. Several key mechanisms have been elucidated:

  • JNK Pathway Activation:

    • PCDH17 upregulation activates the JNK signaling pathway in colorectal cancer cells

    • This activation is characterized by increased phosphorylation of JNK and its downstream target c-Jun

    • JNK activation serves as the central mediator between PCDH17 expression and autophagy induction

  • Autophagy Regulation:

    • PCDH17 expression positively regulates autophagy-related proteins, particularly increasing BECN1 (Beclin-1) expression and LC3B-II turnover

    • PCDH17 knockdown experiments demonstrated significant inhibition of LC3B-II turnover and BECN1 expression

    • Importantly, BECN1 overexpression did not affect PCDH17 expression, suggesting a unidirectional regulatory relationship

  • Experimental Validation:

    • Pharmacological inhibition of JNK using SP600125 (10 μM) in PCDH17-transfected CRC cells significantly decreased phosphorylated c-Jun levels and reduced the LC3-II/I ratio, without affecting PCDH17 expression

    • This confirms JNK's position downstream of PCDH17 but upstream of autophagy induction

    • PCDH17-induced autophagic cell death was attenuated when JNK was inhibited

  • 5-FU Response Mechanism:

    • 5-FU treatment induces PCDH17 upregulation in a dose-dependent manner

    • This upregulation correlates with concurrent JNK activation

    • The enhanced 5-FU sensitivity in PCDH17-expressing cells is significantly mediated through this JNK-dependent autophagic cell death pathway

This mechanistic understanding provides valuable insights for targeted approaches in cancer therapy, particularly for enhancing chemosensitivity in colorectal cancer through modulation of the PCDH17-JNK-autophagy axis.

How can PCDH17 Antibody be used in dual-labeling experiments to investigate its interaction with the JNK pathway?

Dual-labeling experiments utilizing PCDH17 Antibody, HRP conjugated alongside JNK pathway markers provide powerful insights into their functional interactions. The following methodological approach enables comprehensive investigation of these relationships:

  • Sequential Immunohistochemistry (IHC) Protocol:

    • First staining round: Apply PCDH17 Antibody, HRP conjugated (1:100-500) and develop with DAB (brown)

    • Heat inactivation: Microwave slides in citrate buffer (pH 6.0) for 10 minutes

    • Second staining round: Apply antibodies against phospho-JNK or phospho-c-Jun and develop with Vector Blue

    • This approach enables visualization of spatial relationships between PCDH17 and activated JNK pathway components

  • Multiplex Immunofluorescence Strategy:

    • Convert HRP-conjugated PCDH17 Antibody to fluorescent signal using tyramide signal amplification

    • Counter-label with antibodies against JNK pathway components (p-JNK, p-c-Jun)

    • Include autophagy markers (LC3B, BECN1) to visualize the complete signaling axis

    • Employ confocal microscopy to assess co-localization patterns

  • Proximity Ligation Assay (PLA):

    • Use PCDH17 Antibody (after HRP inactivation) alongside JNK pathway antibodies

    • This technique reveals potential protein-protein interactions occurring within 40nm distance

    • Quantification of PLA signals provides insights into the molecular proximity of PCDH17 and JNK pathway components

  • Co-Immunoprecipitation Framework:

    • Use PCDH17 Antibody for immunoprecipitation followed by Western blotting for JNK pathway components

    • Alternatively, immunoprecipitate JNK pathway proteins and probe for PCDH17

    • Include appropriate controls (IgG control, input lysate)

  • Time-course Analysis Post-Treatment:

    • Treat cells with 5-FU at various time points (6h, 12h, 24h, 48h)

    • Perform dual-labeling to track the temporal relationship between PCDH17 expression and JNK activation

    • This reveals whether JNK activation precedes, follows, or occurs simultaneously with PCDH17 upregulation

Research has established that inhibition of JNK in PCDH17-transfected colorectal cancer cells significantly decreases phosphorylated c-Jun levels and reduces the LC3-II/I ratio without affecting PCDH17 expression, confirming JNK's position downstream of PCDH17 but upstream of autophagy induction . These dual-labeling approaches provide critical insights into the spatiotemporal dynamics of the PCDH17-JNK-autophagy signaling axis.

How should researchers address inconsistent PCDH17 staining patterns in immunohistochemistry?

When encountering inconsistent PCDH17 staining patterns in immunohistochemistry experiments using HRP-conjugated antibodies, a systematic troubleshooting approach is essential:

  • Fixation and Processing Evaluation:

    • Inconsistent fixation times can significantly impact PCDH17 epitope availability

    • Monitor and standardize fixation duration (optimal: 24-48 hours in 10% neutral buffered formalin)

    • Assess tissue processing protocols, as excessive dehydration or clearing can denature PCDH17 protein

  • Antigen Retrieval Optimization:

    • PCDH17 detection often requires precise antigen retrieval conditions

    • Compare citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0) for optimal epitope unmasking

    • Adjust retrieval time (15-30 minutes) and temperature (95-98°C) systematically

    • Document conditions that yield reproducible results for your specific tissue types

  • Antibody Dilution Matrix:

    • Create a dilution series (1:50, 1:100, 1:200, 1:500) to identify optimal working concentration

    • Different tissue types may require different antibody concentrations

    • Extend primary antibody incubation time (overnight at 4°C vs. 2 hours at room temperature)

  • Positive Control Validation:

    • Include tissues with known PCDH17 expression patterns in each staining batch

    • For colorectal cancer research, incorporate both 5-FU-sensitive and 5-FU-resistant cases as internal references

    • Use multiple positive controls to account for expression variability

  • Technical Considerations:

    • Ensure even distribution of reagents across tissue sections

    • Minimize section drying during the staining procedure

    • Standardize washing steps (duration, buffer composition, agitation)

    • Consider automated staining platforms for improved consistency

  • Signal Enhancement Strategy:

    • For weak signals, implement tyramide signal amplification (TSA)

    • Increase DAB development time while monitoring under microscope

    • Adjust counterstaining intensity to improve signal-to-background ratio

Research has shown that PCDH17 localizes predominantly in the cytoplasm of colorectal cancer cells . Deviations from this pattern may indicate technical issues or biologically significant variations that warrant further investigation.

What are the potential pitfalls in PCDH17 expression analysis as a predictive biomarker for chemosensitivity?

When evaluating PCDH17 expression as a predictive biomarker for chemosensitivity, researchers should be aware of several potential pitfalls that could affect data interpretation and clinical translation:

  • Intratumoral Heterogeneity Considerations:

    • PCDH17 expression can vary significantly across different regions within the same tumor

    • Single biopsy samples may not accurately represent the entire tumor's PCDH17 status

    • Recommendation: Analyze multiple tumor regions (minimum 3-5 areas) and report both the average and heterogeneity index

  • Scoring System Standardization:

    • Inconsistent scoring methods can lead to discrepant results between studies

    • Establish clear criteria for what constitutes "high" versus "low" PCDH17 expression

    • Consider both staining intensity and percentage of positive cells (e.g., H-score or Allred scoring)

    • The threshold used in published research (52.4% positivity in chemosensitive vs. 7.7% in chemoresistant tissues) should be validated in independent cohorts

  • Confounding Molecular Factors:

    • PCDH17 expression correlates with BECN1 levels, but other autophagy regulators may influence outcomes

    • Additional genetic alterations (e.g., JNK pathway mutations) could modify the PCDH17-chemosensitivity relationship

    • Consider multivariate analysis incorporating known colorectal cancer molecular subtypes

  • Technical Variables:

    • Pre-analytical variables (fixation time, tissue processing) can affect PCDH17 immunoreactivity

    • Antibody selection: polyclonal antibodies may show batch-to-batch variability

    • Detection systems (DAB vs. fluorescence) might yield different sensitivity levels

  • Clinical Context Limitations:

    • The predictive value of PCDH17 has been primarily established for 5-FU monotherapy

    • Modern treatment regimens often combine multiple agents (FOLFOX, FOLFIRI)

    • Validation studies should assess PCDH17's predictive value in current combination therapy settings

  • Alternative Mechanism Consideration:

    • While PCDH17 is linked to autophagy and JNK-dependent mechanisms, alternative pathways may contribute to chemosensitivity

    • PCDH17 methylation status may provide additional or complementary predictive information

    • Consider integrating multiple biomarkers for improved predictive accuracy

Research has shown a significant association between PCDH17 expression and 5-FU sensitivity (p<0.0001) , but these potential pitfalls should be addressed to establish PCDH17 as a robust clinical biomarker.

How can researchers distinguish between PCDH17-induced autophagy and general stress-induced autophagy in experimental systems?

Distinguishing PCDH17-induced autophagy from general stress-induced autophagy requires sophisticated experimental designs and careful controls. The following methodological approaches can help researchers make this critical distinction:

  • Genetic Modulation Systems:

    • PCDH17 Knockdown in Stress Conditions: Apply 5-FU or other stressors to cells with and without PCDH17 knockdown via shRNA

    • Inducible PCDH17 Expression: Use Tet-On/Off systems to control PCDH17 expression independent of stress factors

    • Rescue Experiments: Reintroduce PCDH17 in knockout cells and assess if autophagy is restored specifically through PCDH17-dependent mechanisms

    • Research has shown that PCDH17 knockdown significantly inhibits LC3B-II turnover and BECN1 expression, confirming direct regulation

  • Pathway-Specific Inhibition:

    • JNK Pathway Blockade: Use SP600125 (10 μM) to specifically inhibit JNK and determine if PCDH17-induced autophagy is selectively affected

    • Control Stress Pathways: Simultaneously inhibit multiple stress response pathways (MAPK, p38, NF-κB) to isolate PCDH17-specific effects

    • Autophagy Stage Inhibition: Apply early (3-MA) vs. late (bafilomycin A1) autophagy inhibitors to characterize the stage specificity of PCDH17's effect

    • Data shows that inhibiting JNK in PCDH17-transfected cells specifically decreases the LC3-II/I ratio without affecting PCDH17 expression

  • Temporal Analysis:

    • Time-Course Experiments: Monitor autophagy markers at multiple time points post-stressor application

    • PCDH17 Expression Kinetics: Track the relationship between PCDH17 upregulation and autophagy induction

    • Pulse-Chase Analysis: Use LC3 turnover assays to determine if PCDH17 affects autophagy induction or flux

  • Molecular Interaction Studies:

    • Proximity Ligation Assays: Determine if PCDH17 directly interacts with autophagy machinery components

    • Co-Immunoprecipitation: Assess physical interactions between PCDH17, JNK pathway components, and autophagy proteins

    • Subcellular Fractionation: Analyze whether PCDH17 relocates to autophagosome formation sites during autophagy induction

  • Comparative Systems Biology:

    • Transcriptome Analysis: Compare gene expression patterns between PCDH17-induced and general stress-induced autophagy

    • Phosphoproteomics: Identify unique phosphorylation signatures in PCDH17-mediated autophagic responses

    • Metabolic Profiling: Assess whether PCDH17-induced autophagy has distinctive metabolic consequences

Research has demonstrated that autophagy plays a dominant role in PCDH17-induced cell death, as autophagy inhibitors blocked cell death to a greater extent than pancaspase inhibitors . This distinctive feature helps differentiate PCDH17-mediated autophagy from general stress responses and highlights its potential as a therapeutic target.

What emerging technologies might enhance PCDH17 detection and functional analysis in cancer research?

Several cutting-edge technologies are poised to revolutionize PCDH17 detection and functional analysis in cancer research, offering unprecedented resolution and insights:

  • Advanced Imaging Technologies:

    • Super-Resolution Microscopy: Techniques like STORM and PALM can visualize PCDH17 localization at nanometer resolution, potentially revealing previously undetectable subcellular distribution patterns

    • Lattice Light-Sheet Microscopy: Enables real-time visualization of PCDH17 trafficking and interactions with minimal phototoxicity in living cells

    • Spatial Transcriptomics: Combines PCDH17 protein detection with localized RNA expression analysis for comprehensive spatial context

  • Single-Cell Analysis Platforms:

    • Single-Cell Proteomics: Quantifies PCDH17 protein levels and associated pathway components at individual cell resolution

    • CyTOF/Mass Cytometry: Simultaneously measures PCDH17 alongside dozens of other cancer and immune markers without fluorescence spillover limitations

    • Cellular Indexing of Transcriptomes and Epitopes (CITE-seq): Links PCDH17 protein expression with transcriptomic profiles at single-cell resolution

  • Genome Editing and Screening:

    • CRISPR Base Editing: Introduces specific PCDH17 mutations without DNA double-strand breaks

    • CRISPR Activation/Interference (CRISPRa/CRISPRi): Modulates PCDH17 expression without altering the genetic sequence

    • CRISPR Screens: Systematically identifies genes that modify PCDH17-dependent chemosensitivity

    • Prime Editing: Enables precise introduction of clinically relevant PCDH17 mutations for functional characterization

  • Organoid and Patient-Derived Models:

    • Tumor Organoids: Creates patient-specific 3D models to test PCDH17's role in chemosensitivity

    • Microfluidic Tumor-on-a-Chip: Assesses PCDH17 functions in more physiologically relevant microenvironments

    • Humanized Mouse Models: Evaluates PCDH17-targeted therapies in the context of human immune components

  • Therapeutic Development Platforms:

    • Antibody-Drug Conjugates (ADCs): Utilizes PCDH17-targeting antibodies to deliver cytotoxic payloads specifically to cancer cells

    • mRNA Therapeutics: Temporarily restores PCDH17 expression in tumors with methylated PCDH17

    • Small Molecule Screens: Identifies compounds that modulate PCDH17-JNK-autophagy axis for enhanced chemosensitivity

These technologies could significantly advance our understanding of how PCDH17 expression correlates with chemosensitivity in colorectal cancer and potentially other malignancies, as evidenced by research showing PCDH17's role in promoting JNK-dependent autophagic cell death and enhancing 5-FU sensitivity .

How might PCDH17 status be integrated into personalized medicine approaches for colorectal cancer?

Integration of PCDH17 status into personalized medicine frameworks for colorectal cancer represents a promising frontier that could significantly enhance treatment outcomes through several strategic approaches:

  • Diagnostic and Predictive Biomarker Implementation:

    • Companion Diagnostic Development: Create standardized IHC assays for PCDH17 expression to guide 5-FU-based therapy decisions

    • Multiparameter Biomarker Panels: Incorporate PCDH17 alongside established markers (MSI status, RAS/RAF mutations) for comprehensive profiling

    • Liquid Biopsy Applications: Develop methods to detect PCDH17 methylation patterns in circulating tumor DNA

    • Research has established significant correlations between PCDH17 expression and 5-FU sensitivity (p<0.0001), providing strong rationale for its clinical implementation

  • Therapy Selection Algorithms:

    • Treatment Decision Trees: Develop algorithms incorporating PCDH17 status to guide chemotherapy selection

    • Combination Therapy Stratification: Determine if PCDH17 status predicts response to 5-FU in combination with targeted agents (anti-EGFR, anti-VEGF)

    • Alternative Pathway Selection: For PCDH17-low tumors, recommend therapies targeting alternative pathways

  • Clinical Trial Design and Patient Stratification:

    • Biomarker-Guided Trials: Design trials that prospectively stratify patients based on PCDH17 expression

    • Adaptive Trial Designs: Incorporate PCDH17 testing in basket or umbrella trials to refine patient selection

    • Post-Hoc Analyses: Retrospectively analyze PCDH17 status in completed trials to validate its predictive value

  • Epigenetic Modulation Strategies:

    • Demethylating Agent Combinations: For tumors with methylated PCDH17, explore combining demethylating agents with conventional chemotherapy

    • Histone Deacetylase Inhibitors: Test if epigenetic modifiers can restore PCDH17 expression in resistant tumors

    • Targeted Demethylation: Develop CRISPR-based approaches for locus-specific PCDH17 demethylation

  • Monitoring and Resistance Management:

    • Serial PCDH17 Assessment: Monitor changes in PCDH17 expression during treatment to detect emerging resistance

    • Alternative Pathway Activation: Screen for bypass mechanisms in PCDH17-positive tumors that develop resistance

    • Adaptive Therapy Protocols: Adjust treatment intensity based on dynamic PCDH17 expression

What are the challenges in developing PCDH17-targeted therapies for enhancing chemosensitivity?

Developing PCDH17-targeted therapies to enhance chemosensitivity faces several complex challenges that span basic science, translational research, and clinical implementation domains:

  • Biological Complexity Challenges:

    • Target Specificity: PCDH17 belongs to the protocadherin family with multiple homologous members, creating potential off-target effects

    • Context-Dependent Function: PCDH17's role may vary across cancer types and even within different regions of the same tumor

    • Compensatory Mechanisms: Cancer cells might activate alternative pathways to circumvent PCDH17-mediated effects

    • Pathway Redundancy: Multiple inputs regulate autophagy beyond the PCDH17-JNK axis, potentially limiting therapeutic efficacy

  • Therapeutic Development Hurdles:

    • Protein Restoration Challenge: As a tumor suppressor gene, developing therapeutics to restore PCDH17 function is more difficult than inhibiting oncogenic targets

    • Epigenetic Targeting: Selective demethylation of PCDH17 without affecting other genes remains technically challenging

    • Antibody Accessibility: PCDH17's membrane localization may not be uniformly accessible in solid tumors due to physical barriers

    • Delivery Systems: Targeting deep-seated tumors with PCDH17-modulating agents requires advanced delivery technologies

  • Clinical Translation Obstacles:

    • Patient Selection: Identifying optimal candidates for PCDH17-targeted therapy requires validated biomarker assays

    • Treatment Sequencing: Determining whether PCDH17 modulation should precede, accompany, or follow conventional chemotherapy

    • Resistance Monitoring: Developing methods to track adaptation to PCDH17-targeted interventions

    • Combination Toxicity: Managing potential synergistic toxicities when combining PCDH17 modulators with conventional chemotherapy

  • Technical and Methodological Limitations:

    • Model Systems: Current preclinical models may not fully recapitulate the complexity of PCDH17 regulation in human tumors

    • Functional Readouts: Standardizing methods to assess successful PCDH17 modulation in clinical specimens

    • Pharmacodynamic Markers: Identifying reliable indicators of on-target engagement for PCDH17-directed therapies

    • Long-term Effects: Understanding delayed consequences of manipulating PCDH17-mediated autophagy

  • Regulatory and Commercial Considerations:

    • Novel Endpoint Requirements: Regulatory approval may require innovative endpoints beyond traditional response criteria

    • Companion Diagnostic Development: Synchronized development of therapeutics and diagnostics adds complexity

    • Market Positioning: Determining how PCDH17-targeted approaches would complement or compete with established therapies

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.