PDIA3 contains four thioredoxin-like domains (a, b, b′, a′) with distinct functional regions:
Domain | Key Features | Catalytic Motifs | Binding Partners |
---|---|---|---|
a/a′ | Catalytically active | Cys-Gly-His-Cys (C57-G58-H59-C60; C406-G407-H408-C409) | Substrate proteins |
b/b′ | Chaperone binding | K214, K274, R282 residues | Calnexin/calreticulin |
The enzyme forms transient disulfide bonds with substrates through its redox-active sites, while the bb′ domains mediate interactions with lectin chaperones to position glycoproteins for efficient folding .
Core activities include:
Disulfide bond formation/isomerization in nascent glycoproteins
Quality control in MHC class I antigen presentation complex
ER stress response through mTORC1 signaling regulation
Key cellular roles:
Cancer biology: Overexpressed in 70% of malignancies, correlating with:
Neuroprotection:
PDIA3 expression predicts outcomes across malignancies:
PDIA3 inhibition strategies:
The human recombinant protein (UniProt P30101) is engineered for research applications:
Parameter | Specification |
---|---|
Expression System | E. coli |
Molecular Weight | 58.5 kDa |
Purity | >95% (SDS-PAGE) |
Activity | >20 A650/cm/min/mg |
Stabilization | 10% glycerol, 1mM DTT |
This recombinant form enables study of:
PDIA3, also known as ERp57, is a disulfide oxidoreductase and isomerase that primarily resides in the endoplasmic reticulum (ER). It plays critical roles in protein folding by catalyzing the formation, breakage, and rearrangement of disulfide bonds in substrate proteins. PDIA3 is particularly involved in the post-translational folding and refolding of disulfide-bonded domains, which characterize many extracellular matrix (ECM) proteins .
Methodologically, PDIA3's enzymatic activity can be assessed through oxidoreductase assays using fluorogenic substrates. Researchers can monitor PDIA3's isomerase activity by tracking the refolding of denatured proteins with scrambled disulfide bonds. Subcellular localization studies typically employ immunofluorescence with specific antibodies such as the mouse monoclonal IgG1 antibody (Abcam, Ab13506) .
PDIA3 expression and activity are regulated at multiple levels including transcriptional control, post-translational modifications, and subcellular localization. Under normal conditions, PDIA3 primarily functions within the ER, but during cellular stress, it can translocate to the cell surface where it becomes available for antibody binding .
To investigate PDIA3 regulation, researchers should consider experimental approaches that monitor both expression levels (via qPCR and Western blotting) and cellular localization (through subcellular fractionation and immunofluorescence). Stress conditions such as ER stress inducers (tunicamycin, thapsigargin) can be used to study dynamic changes in PDIA3 expression and localization.
Several validated methods exist for PDIA3 detection in human samples:
Western blotting: Using specific antibodies such as mouse monoclonal anti-PDIA3 (1:2,000 dilution)
Immunohistochemistry: For tissue localization studies
ELISA: For quantitative measurement in serum or tissue lysates
Mass spectrometry: For absolute quantitation and post-translational modification analysis
For quantitative proteomics approaches, researchers should consider parallel reaction monitoring (PRM) methods similar to those described in the literature, which have successfully identified PDIA3 peptides using heavy-labeled standards for accurate quantification .
PDIA3 has been implicated in hepatic inflammation through immune autoreactivity mechanisms. In high-fat and high-fructose (HFHF) diet models, PDIA3 epitopes drive TH1- and TH17-polarized immune responses in the liver and promote the generation of pathogenic anti-PDIA3 antibodies .
Methodologically, researchers investigating this pathway should consider:
Diet-induced models (HFHF) to trigger metabolic stress
Analysis of MHC-II presentation of PDIA3-derived peptides via immunopeptidome profiling
Assessment of T cell responses through proliferation assays with PDIA3-specific T cells
Quantification of anti-PDIA3 antibodies via ELISA
Evaluation of hepatocyte damage through serum transaminase measurements
The sequence DGEEAGAYDGPRTADG has been identified as a key PDIA3 peptide presented by MHC-II (I-Ab), while IFRDGEEAGAYDGPRTADGIVSHLK represents a linear peptide uniquely recognized by anti-PDIA3 antibodies in HFHF diet models .
When investigating PDIA3's role in nonalcoholic steatohepatitis (NASH), researchers should employ multiple complementary approaches:
Dietary manipulation: HFHF diet models effectively induce NASH with associated PDIA3 upregulation
Cell surface PDIA3 detection: Flow cytometry or immunofluorescence to quantify PDIA3 translocation to hepatocyte surfaces
Antibody purification: Peptide affinity column methods to isolate anti-PDIA3 antibodies from serum
Passive transfer studies: Administration of PDIA3-specific T cells or antibodies to evaluate pathogenicity
Liver damage assessment: Measurement of ALT/AST and histological evaluation
Research has shown that lipotoxicity and glucotoxicity associated with an HFHF diet promote increased PDIA3 levels at the hepatocyte cell surface, making it available for antibody binding. This mechanism has been linked to immunogenic cell death of metabolically stressed hepatocytes .
For isolation and characterization of anti-PDIA3 antibodies, researchers should follow these methodological steps:
Serum collection from appropriate models (e.g., HFHF diet-fed mice) or human patients
Initial quantification via ELISA using recombinant PDIA3 protein
Antibody titration to determine concentration
Purification using peptide affinity columns specific for PDIA3 epitopes
Specificity confirmation through:
Immunoblotting against recombinant His-tagged PDIA3
ELISA with purified recombinant PDIA3
Comparison with commercial anti-PDIA3 antibodies as positive controls
This approach has successfully demonstrated statistically significant increases in anti-PDIA3 antibodies elicited by HFHF diets in experimental models .
PDIA3 has emerged as a significant factor in breast cancer progression through multiple mechanisms:
Elevated expression in tumors versus normal breast tissue, particularly in invasive ductal breast cancers compared to lobular cancers
Promotion of extracellular matrix (ECM) modifications that support cancer cell migration and invasion
Enhancement of anchorage-independent growth in mammospheres
Facilitation of bone metastasis in metastatic breast cancer cell lines
Experimental approaches to study these mechanisms should include:
Comparative proteomics between normal and cancer tissues
Functional assays of cell spreading, focal adhesion formation, and migration following PDIA3 inhibition
Analysis of PDIA3's effects on the secretome, particularly ECM and heparin-binding proteins
In vivo metastasis models to evaluate the impact of PDIA3 knockdown or inhibition
Notably, PDIA3 inhibition by compounds such as 16F16 (at 5 μM concentration) has been shown to decrease cell spreading, reduce focal adhesions, and inhibit cell migration in MDA-MB-231 breast cancer cells .
For comprehensive investigation of PDIA3 in cancer, researchers should consider these experimental models and approaches:
Cell lines:
MDA-MB-231 human breast cancer cells (particularly for invasive/metastatic phenotypes)
Matched pairs of cell lines with PDIA3 knockdown or knockout
In vitro functional assays:
Cell spreading and adhesion assays
Focal adhesion formation (visualized by vinculin staining)
Migration and invasion assays
Conditioned medium transfer experiments to assess secretome effects
Secretome analysis:
Collection of conditioned medium after 48h culture with or without PDIA3 inhibition
Enrichment of heparin-binding proteins
Quantitative proteomics using mass spectrometry
Bioinformatic analysis of protein-protein interaction networks
In vivo models:
Orthotopic implantation for primary tumor growth
Experimental metastasis models for specific organ tropism
Culture conditions should include serum-free Fibroblast Growth Medium supplemented with L-ascorbic acid (50 μg/mL) to promote collagen synthesis when studying ECM effects .
PDIA3 inhibition profoundly impacts the cancer cell secretome, particularly affecting extracellular matrix components and organization:
Quantitative proteomics analysis of MDA-MB-231 cells treated with the PDIA3 inhibitor 16F16 (5 μM) identified 80 proteins reproducibly decreased at least twofold in the conditioned medium .
Gene Ontology analysis revealed that many affected proteins have roles in:
ECM structure and function
Cell adhesion
Epithelial-mesenchymal transition (EMT)
Ribosomal functions associated with extracellular vesicles
The predominant types of disulfide-bonded domains in the affected extracellular proteins contained β-hairpin folds, with the knottin fold being most common .
Functional effects of these secretome changes include reduced:
Promigratory cell spreading
F-actin organization
Focal adhesion formation
Researchers studying these effects should employ a combination of proteomics, structural biology approaches, and functional assays to fully characterize the impact of PDIA3 inhibition on the cancer microenvironment.
Distinguishing between intracellular and cell surface PDIA3 functions requires sophisticated methodological approaches:
Selective cell surface labeling:
Non-permeabilized cell surface biotinylation followed by streptavidin pull-down
Cell-impermeant cross-linking reagents to identify surface interaction partners
Flow cytometry of non-permeabilized cells using antibodies against extracellular PDIA3 epitopes
Subcellular fractionation:
Differential centrifugation to separate membrane fractions
Density gradient separation of cellular compartments
Western blotting with compartment-specific markers to confirm fraction purity
Functional discrimination:
Cell-impermeable PDIA3 inhibitors to selectively target surface activity
Function-blocking antibodies that cannot enter cells
Expression of engineered PDIA3 with altered trafficking signals
Research has demonstrated that PDIA3 translocation to the cell surface is associated with cellular stress conditions, including metabolic stress in hepatocytes from HFHF diet-fed mice and in aggressive breast cancer cells .
When investigating PDIA3's protein-protein interaction networks, researchers should consider these methodological approaches:
Affinity-based methods:
Co-immunoprecipitation with anti-PDIA3 antibodies
Pull-down assays using recombinant PDIA3 as bait
Proximity labeling techniques (BioID, APEX) for identifying transient interactions
Structural biology approaches:
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction surfaces
Cross-linking mass spectrometry (XL-MS) to identify interaction sites
Crystallography or cryo-EM for detailed structural characterization
Bioinformatic analysis:
Protein-protein interaction network analysis of PDIA3-affected secretome proteins
Domain analysis focusing on disulfide-bonded domains (particularly β-hairpin folds)
Integration with disease-specific datasets (e.g., breast cancer subtypes)
Functional validation:
Site-directed mutagenesis of interaction domains
Domain-swapping experiments
Competitive inhibition studies
Protein-protein interaction analysis of PDIA3-regulated secretome proteins has revealed networks particularly relevant to ECM organization and cell adhesion in breast cancer models .
PDIA3's role in immunogenic cell death (ICD) represents an emerging area of research with important implications for both cancer and inflammatory conditions:
Current understanding:
PDIA3 is translocated to the cell surface during cellular stress conditions
Surface-exposed PDIA3 can serve as a target for antibody binding
PDIA3 epitopes can drive TH1- and TH17-polarized immune responses
PDIA3-specific antibodies can exacerbate hepatocyte death in metabolically stressed conditions
Experimental investigation approaches:
ICD induction protocols using established ICD inducers (anthracyclines, oxaliplatin)
Monitoring PDIA3 surface exposure during different forms of cell death (flow cytometry)
Assessment of DAMP (damage-associated molecular pattern) release in relation to PDIA3 exposure
Dendritic cell activation assays to evaluate immunogenicity
T cell response profiling following ICD induction with and without PDIA3 inhibition
In vivo vaccination assays to assess immunogenicity of dying cells
Clinical relevance:
Evaluation of anti-PDIA3 antibody levels in patients with chronic inflammatory liver conditions
Analysis of PDIA3 expression in relation to immune infiltration in tumors
Correlation with response to immunotherapy
Research has demonstrated increased humoral responses to PDIA3 in patients with autoimmune hepatitis, primary biliary cholangitis, and type 2 diabetes, suggesting broader clinical implications .
Several approaches show promise for therapeutic targeting of PDIA3:
Small molecule inhibitors:
Blocking antibodies:
Antibodies targeting surface-exposed PDIA3 in disease states
Epitope-specific antibodies to block particular functions
Peptide-based approaches:
Competitive inhibitors based on substrate binding sites
Cell-penetrating peptides targeting intracellular PDIA3
Gene silencing:
siRNA or shRNA approaches for transient or stable knockdown
CRISPR-Cas9 for genetic modification in experimental models
The ideal therapeutic approach depends on the specific disease context, with considerations for:
Target location (intracellular vs. surface PDIA3)
Disease mechanism (enzymatic activity vs. immune recognition)
Delivery challenges (particularly for intracellular targeting)
Potential off-target effects on related PDI family members
Rigorous evaluation of PDIA3 inhibitor specificity and efficacy requires comprehensive testing:
Biochemical assays:
In vitro enzymatic assays using purified PDIA3 and related PDI family members
Determination of IC50 values for PDIA3 vs. other PDIs
Assessment of binding kinetics using surface plasmon resonance or isothermal titration calorimetry
Cellular validation:
Target engagement assays (cellular thermal shift assay, drug affinity responsive target stability)
Phenotypic assays monitoring known PDIA3-dependent functions
Comparison of inhibitor effects with genetic knockdown/knockout phenotypes
Proteomic approaches:
Analysis of disulfide proteome changes following inhibitor treatment
Secretome analysis to monitor effects on extracellular proteins
Comparison with established PDIA3 inhibition signatures
Dose-response considerations:
These approaches collectively provide a comprehensive evaluation of inhibitor quality and suitability for specific research or therapeutic applications.
Accumulating evidence supports PDIA3's potential as a biomarker in several human diseases:
Breast cancer:
Liver diseases:
Methodological considerations for biomarker development:
Standardized ELISA methods for anti-PDIA3 antibody detection
Tissue microarray analysis for PDIA3 expression in tumor samples
Integration with other clinical parameters for enhanced predictive value
Prospective validation in independent patient cohorts
These findings suggest PDIA3 may serve as both a tissue biomarker and a serological biomarker through anti-PDIA3 antibody detection.
For investigating PDIA3-related biomarkers in clinical samples, researchers should consider these methodological approaches:
For developing PDIA3 as a biomarker in breast cancer, researchers should focus on its correlation with the basal subtype and its relationship to distant metastasis-free survival, as supported by analysis against human breast cancer datasets .
PDIA3 consists of four thioredoxin-like domains: a, b, b′, and a′. The a and a′ domains contain Cys-Gly-His-Cys active site motifs and are catalytically active. The bb′ domains contain a binding site for calnexin (CNX), which is composed of positively charged, highly conserved residues that interact with the negatively charged residues of the CNX P domain .
PDIA3 is primarily localized in the endoplasmic reticulum (ER) and interacts with lectin chaperones such as calreticulin and calnexin to modulate the folding of newly synthesized glycoproteins. Complexes of lectins and PDIA3 mediate protein folding by promoting the formation of disulfide bonds in their glycoprotein substrates .
PDIA3 plays a significant role in various biological processes, including:
Recombinant PDIA3 is produced using recombinant DNA technology, which involves inserting the PDIA3 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant form is used in various research and therapeutic applications, including studies on protein folding, cancer progression, and immune response .