A PPIA antibody pair typically consists of:
Capture antibody: Binds to PPIA in solid-phase assays (e.g., ELISA)
Detection antibody: Recognizes a distinct PPIA epitope, often conjugated with labels like HRP or biotin
Key applications supported by experimental validation:
Immunohistochemistry (IHC) and flow cytometry with lyophilized formulations reconstituted to 500 µg/mL
HIV-1 replication studies due to PPIA’s interaction with viral Gag proteins
Proteintech’s 35001-1-AP shows no cross-reactivity with PPIF due to absorption-based purification .
Boster Bio’s A01308 demonstrates >95% specificity in WB and IHC using immortalized MSC lines .
PPIA antibody pairs have identified apoptosis-related biomarkers in gastric cancer proteomics .
In HIV-1 studies, PPIA antibodies confirmed its role in viral capsid assembly via Gag interaction .
Reference gene stability: PPIA ranked highest in BestKeeper analysis (Pearson r = 0.98) when paired with YWHAZ in MSC studies .
Sample Preparation: Use RIPA buffer for tissue lysates.
Antibody Dilution:
Detection: Chemiluminescence for WB; fluorophore conjugates for imaging .
PPIA (Peptidylprolyl Isomerase A/Cyclophilin A) is an 18 kDa protein that belongs to the peptidyl-prolyl cis-trans isomerase (PPIase) family. It catalyzes the cis-trans isomerization of proline imidic peptide bonds in oligopeptides and accelerates protein folding .
PPIA antibody pairs are significant because they enable:
Detection of PPIA in multiple experimental contexts (Western blotting, immunohistochemistry, flow cytometry)
Quantification of PPIA expression levels through sandwich ELISA techniques
Investigation of PPIA's roles in HIV-1 replication, cancer progression, and protein translation
Study of PPIA interactions with other proteins like NRF2 in cancer pathways
The canonical protein structure has 165 amino acid residues and can be found in the nucleus, cytoplasm, and extracellular space .
When selecting antibody pairs for PPIA detection, researchers should consider:
Epitope targeting: Ensure antibodies recognize distinct, non-overlapping epitopes. From the literature, antibodies targeting the C-terminal region (like ABIN2855908) and those targeting N-terminal regions can form effective pairs .
Host species diversity: Select antibodies from different host species (e.g., rabbit and mouse) to avoid cross-reactivity in sandwich assays.
Validation status: Prioritize antibodies validated in multiple applications with knockout validation where possible .
Species cross-reactivity: Consider whether the antibodies need to work across species (human, mouse, rat) based on your experimental model .
Performance in specific applications: Some antibodies perform better in certain applications than others. For instance, the Picoband antibody A01308 shows strong performance in Western blot, IHC, ICC, and flow cytometry .
Application | Recommended Validation Method |
---|---|
ELISA | Recombinant protein standard curves |
Western Blot | Positive control lysates, knockouts |
IHC/ICC/IF | Known positive tissues, blocking peptides |
Flow Cytometry | Positive control cell lines, isotype controls |
Epitope mapping is crucial for ensuring your antibody pair targets distinct epitopes. Based on the comparative study of epitope mapping technologies (search result 4), we recommend:
Hydrogen-Deuterium Exchange (HDX): This technique identified epitopes in 5/5 antibody-antigen pairs tested and provides high-resolution mapping of conformational epitopes .
Chemical Cross-Linking (XL): While it missed some antibody CDR3 residues, XL provides clear interaction interfaces between antibody and antigen, with identified residues typically 15-30Å apart .
Alanine Scanning Mutagenesis: This approach can identify critical residues for antibody binding.
Peptide Array Analysis: While this approach worked for only 2/5 antibody-antigen pairs in the study, it may be useful for linear epitopes in PPIA .
For PPIA specifically, antibodies targeting the C-terminal region (residues 116-165) have been documented for successful detection .
When conducting co-immunoprecipitation studies with PPIA antibody pairs:
Negative controls:
IgG isotype control to assess non-specific binding
Lysates from PPIA-knockout cells or tissues (when available)
Competitive blocking with recombinant PPIA protein
Positive controls:
Known PPIA interaction partners (e.g., NRF2 as demonstrated in search result 3)
Recombinant PPIA-tagged protein
Validation approaches:
As shown in the study of PPIA-NRF2 interactions, co-immunoprecipitation revealed that PPIA's enzymatic activity is required for the interaction, since no binding was observed with catalytically dead PPIA variants .
To study PPIA's role in HIV-1 infection:
Selecting appropriate antibodies: Choose antibodies that don't interfere with the Gag-binding region of PPIA, as this interaction is critical for HIV-1 replication .
Critical experimental design elements:
Include proper controls with cyclosporin A (CsA), which disrupts PPIA-Gag interactions
Consider genetic approaches using cells from individuals with different PPIA SNPs
Examine both virion-incorporated and cellular PPIA separately
Genetic considerations: The SNP3 and SNP4 promoter polymorphisms in PPIA are associated with more rapid CD4+ T-cell loss and disease progression in HIV-infected individuals . Design experiments to compare PPIA levels in cells with different genotypes using your antibody pairs.
According to research, "Cyclophilin A binds to the Gag protein of human immunodeficiency virus type 1 (HIV-1)" and "may have an essential function in HIV-1 replication" .
Recent research has revealed PPIA's role in supporting translation of intrinsically disordered proteins . When investigating this function:
Experimental design considerations:
Use paired antibodies to detect both PPIA and its substrate proteins (~20% of which engage in protein phase separation)
Employ PPIA inhibitors (e.g., TMN355) as controls to confirm translation effects are PPIA-dependent
Consider combined approaches (e.g., IP-western blot) to validate proteomic findings
Translation assay approaches:
Control methodologies:
Use Ppia knockout and heterozygous models
Monitor expression of IDR-rich PPIA substrates in both epithelial and hematopoietic cell lines
Research demonstrates "a significantly decreased rate of de novo translation in stem cells that had been treated with the potent PPIA inhibitor TMN355," confirming PPIA's direct role in protein translation .
PPIA plays important roles in cancer progression, particularly through NRF2 stability in lung cancer . When investigating these mechanisms:
Experimental approach:
Use antibody pairs that can detect both PPIA and its cancer-relevant partners (e.g., NRF2)
Design co-immunoprecipitation experiments to capture protein-protein interactions
Include controls with catalytically inactive PPIA variants (PPIA R55A&F60A)
Validation strategies:
siRNA knockdown of PPIA to confirm antibody specificity
Ubiquitination assays to confirm PPIA's effect on protein stability
Pull-down analysis using recombinant PPIA-conjugated beads
Technical considerations:
CsA treatment blocks PPIA-NRF2 interaction and can serve as an important control
PPIA's enzymatic activity is required for interaction with NRF2, so consider this when selecting antibodies
Research shows that "si PPIA treatment profoundly elevated the NRF2 ubiquitination" and that "PPIA's enzymatic activity is required for the PPIA-NRF2 interaction" .
Common challenges in flow cytometry with PPIA antibodies include:
Intracellular accessibility issues:
Non-specific binding:
Solution: Block with 10% normal goat serum before antibody incubation
Validation: Include rabbit IgG isotype controls at equivalent concentrations
Signal optimization:
Recommended antibody concentration: 1μg/1×10^6 cells
Incubation conditions: 30 min at 20°C
Secondary antibody: DyLight®488 conjugated goat anti-rabbit IgG (5-10μg/1×10^6 cells)
The validated protocol from search result 6 demonstrates successful detection of PPIA in THP-1, U937, and K562 cells with distinct positive population separation from controls .
When facing discrepancies between different antibody-based methodologies:
Common sources of discrepancy:
Epitope accessibility differences between applications
Post-translational modifications affecting antibody recognition
Protein conformation differences in native vs. denatured conditions
Systematic resolution approach:
Validation with multiple antibodies: Use antibodies targeting different PPIA epitopes
Complementary techniques: Combine antibody-based detection with mass spectrometry
Genetic validation: Use PPIA knockout controls or siRNA knockdown
Application-specific considerations:
Western blotting may detect denatured epitopes not accessible in native conditions
IHC/ICC may show different results due to fixation effects on epitope accessibility
ELISA sensitivity often differs from Western blot
The search results show that some antibodies display different molecular weights than expected (e.g., 185 kDa observed vs. 18 kDa calculated) , which may indicate detection of complexes, dimers, or post-translationally modified forms.
For developing a sandwich ELISA for PPIA:
Antibody pair selection:
Optimization parameters:
Validation approach:
Generate standard curves using recombinant PPIA
Test recovery with spiked samples
Perform parallelism testing with diluted biological samples
Parameter | Recommended Range | Optimization Variable |
---|---|---|
Capture Antibody | 1-10 μg/mL | Concentration |
Detection Antibody | 0.5-2 μg/mL | Dilution factor |
Detection Range | 3.12-200 ng/mL | Standard curve range |
Sensitivity | 0.087-0.78 ng/mL | Signal amplification |
Commercial ELISA kits have established detection ranges of "3.12-200ng/ml" with sensitivities as low as "0.087 ng/mL" .
Recent research has revealed PPIA's importance in stem cell biology and aging:
Experimental design considerations:
Use antibody pairs to detect PPIA in hematopoietic stem cells
Combine with functional assays to correlate PPIA levels with stem cell function
Compare young vs. aged stem cell populations
Key methodological approaches:
Immunofluorescence with stem cell markers
Flow cytometry to quantify PPIA in specific stem cell populations
Co-immunoprecipitation to identify age-dependent PPIA interaction partners
Validation strategies:
Genetic approaches with Ppia heterozygous and knockout models
Rescue experiments to demonstrate causal relationships between PPIA and aging phenotypes
Research has shown that "To substantiate the causal relationship between PPIA and the ageing phenotype, we subsequently performed rescue experiments" in hematopoietic stem cells .
For multiplexed detection systems incorporating PPIA antibodies:
Technical requirements:
Ensure antibodies are compatible with multiplexing reagents
Select antibodies from different host species to avoid cross-reactivity
Consider differential labeling strategies (fluorophores, quantum dots)
Validation approach:
Test for signal bleed-through between channels
Verify antibody performance in single-plex before multiplexing
Include single-stained controls in multiplex experiments
Application-specific considerations:
For flow cytometry: carefully select fluorophores with minimal spectral overlap
For multiplexed IHC: sequential staining may be preferable to simultaneous staining
For bead-based multiplexing: test for cross-reactivity with other capture antibodies
Multiplexed approaches allow simultaneous detection of PPIA and its interaction partners or downstream effectors, such as NRF2 in cancer studies .
Genetic variation in PPIA can affect antibody recognition and experimental outcomes:
Key genetic variations to consider:
Experimental design considerations:
When possible, genotype samples for known PPIA polymorphisms
Include samples with different PPIA genotypes as controls
Consider that some epitopes may be affected by genetic variants
Antibody selection strategy:
Choose antibodies targeting conserved regions when working across genetically diverse samples
Use multiple antibodies targeting different epitopes
Validate antibodies against samples with known PPIA variants
Research has shown that "individuals who processed two functional variants in the promoter region of PPIA had higher risk of CD4+ T-cell loss or progression to AIDS-defining diseases" .