PDIA5, also known as protein disulfide isomerase-related protein (PDIR), is a member of the PDI gene family that exhibits chaperone-like activity. It was first identified in 1995 and is naturally expressed in the brain, liver, kidney, and lungs . PDIA5 functions as part of multi-protein complexes engaging with other chaperones to assist in folding newly synthesized proteins. It participates in protein folding and maintaining cell function by ensuring proper protein structure through disulfide bond formation . PDIA5 is particularly important in research because its overexpression is associated with poor prognosis in multiple cancer types, making it a potential biomarker and therapeutic target .
PDIA5 antibodies are versatile tools with multiple research applications:
Western Blotting (WB): For protein expression quantification and validation
Immunohistochemistry (IHC): For tissue localization studies
Immunocytochemistry (ICC): For cellular localization analysis
Immunoprecipitation (IP): For protein-protein interaction studies
ELISA: For quantitative protein detection
Immunofluorescence (IF): For co-localization studies with other proteins
Most commercially available antibodies have been validated for WB and IHC applications with human samples, with some also showing reactivity with mouse and rat tissues .
Methodological validation should include:
Positive control testing: Use cell lines known to express PDIA5 like HepG2, HeLa, or U251 glioma cells
Western blot analysis: Confirm a single band at approximately 60 kDa (predicted size)
Knockdown verification: Use siRNA against PDIA5 to confirm specificity of antibody signal
Cross-reactivity assessment: Test the antibody against related PDI family members
Application-specific validation: For IHC, include positive control tissues like liver samples
Based on research protocols, optimal Western blotting conditions for PDIA5 detection include:
Sample preparation: Complete cell lysis in RIPA buffer with protease inhibitors
Transfer conditions: Semi-dry or wet transfer to PVDF membrane
Blocking: 5% non-fat milk or BSA in TBST for 1 hour
Primary antibody dilution: 1:1000-1:2000 for most commercial antibodies
Incubation time: Overnight at 4°C for optimal signal
Detection: Enhanced chemiluminescence (ECL)
A comprehensive experimental approach should include:
Expression analysis:
Functional studies:
Mechanistic investigations:
In vivo validation:
Xenograft models with PDIA5-modulated cell lines
Correlation with immune infiltration in animal models
PDIA5 plays a multifaceted role in tumor immune regulation:
Research indicates PDIA5 may be a predictive biomarker for immunotherapy response:
Clinical correlation data:
Patients with high PDIA5 expression show better response to anti-PD-1 and anti-CTLA-4 therapies
In anti-PD-L1 cohorts, high PDIA5 expression correlates with significant clinical survival benefits
Complete response rates are higher in patients with high PDIA5 expression (19.35% vs. 6.7% in low PDIA5)
Molecular mechanisms:
Predictive algorithms:
The interaction between PDIA5 and UPR in cancer involves:
ATF6α activation mechanism:
Therapeutic resistance:
Cell survival regulation:
Multiple complementary approaches provide comprehensive analysis:
Transcriptomic analysis:
Protein expression analysis:
Bioinformatic approaches:
Clinical correlation:
A systematic approach includes:
Co-culture systems:
Flow cytometry analysis:
Assessment of immune cell phenotypes in PDIA5-modulated systems
Measurement of immune activation markers and cytokine production
Cell sorting for further molecular analysis
Multiplex immunohistochemistry:
CIBERSORT analysis:
Implementation strategies include:
Biomarker development:
Standardized IHC protocols for PDIA5 detection in clinical samples
Establishment of expression thresholds that correlate with therapy response
Integration with existing biomarkers like PD-L1 expression and tumor mutational burden
Predictive algorithms:
Patient stratification strategies:
Key considerations include:
Target specificity:
PDIA5 belongs to a family of related proteins with similar functions
Development of highly specific inhibitors to avoid off-target effects
Potential compensatory mechanisms within the PDI family
Delivery challenges:
For CNS tumors like gliomas, blood-brain barrier penetration
Targeted delivery systems to minimize systemic toxicity
Combination with existing standard-of-care therapies
Biomarker integration:
Identification of patient subgroups most likely to benefit
Development of companion diagnostics for PDIA5-targeted therapies
Monitoring for acquired resistance mechanisms
Immune modulation complexity:
Methodological approaches to improve specificity:
Antibody selection optimization:
Use antibodies targeting unique regions of PDIA5 to minimize cross-reactivity
Consider monoclonal antibodies for higher specificity in complex applications
Validate with multiple antibodies targeting different epitopes
Protocol optimization:
Increase blocking concentration (5-10% BSA or milk)
Optimize primary antibody concentration through titration experiments
Increase washing duration and frequency
Use additives like 0.1% Tween-20 to reduce non-specific binding
Sample preparation improvements:
Fresh tissue preparation and proper fixation for IHC
Complete protein denaturation for Western blotting
Appropriate antigen retrieval methods for fixed tissues
Controls implementation:
Quantification methodologies include:
Western blot quantification:
IHC scoring systems:
H-score methodology (intensity × percentage of positive cells)
Semi-quantitative scoring (0, 1+, 2+, 3+)
Digital image analysis for more objective quantification
Pathologist validation of scoring methods
Multi-parameter analysis:
Reference standards:
Inclusion of known positive controls in each experiment
Consistent imaging and analysis parameters
Inter-observer validation for subjective scoring methods