PPIL4 (Peptidylprolyl Isomerase Cyclophilin-Like 4) is a protein encoded by the PPIL4 gene in humans (Gene ID: 85313) . It functions as a peptidylprolyl isomerase, facilitating protein folding by catalyzing the cis-trans isomerization of proline imidic peptide bonds. The protein has a calculated molecular weight of approximately 57 kDa, though it is typically observed at 58-65 kDa in experimental conditions . PPIL4 is studied for its potential roles in RNA processing, protein folding, and cellular signaling pathways. Research on PPIL4 contributes to our understanding of fundamental cellular processes and may have implications for various pathological conditions where protein folding and RNA processing are disrupted.
PPIL4 antibodies are utilized across multiple experimental applications, with the most common being:
The optimal application depends on your specific research question, with Western blotting being the most widely validated method for PPIL4 detection in experimental systems .
Multiple cell lines have been experimentally validated for PPIL4 detection using available antibodies:
These cell lines consistently show PPIL4 expression and have been used as positive controls in various experimental settings. When establishing a new experimental system, these cell lines can serve as reliable positive controls for antibody validation .
Differentiating specific from non-specific binding requires implementing multiple validation approaches:
Molecular weight verification: PPIL4 has a calculated molecular weight of 57 kDa but is typically observed between 58-65 kDa on Western blots . Any bands significantly outside this range may represent non-specific binding.
Positive and negative controls: Include lysates from cells known to express PPIL4 (e.g., HeLa or HEK-293) as positive controls . For negative controls, consider using:
Cross-validation with multiple antibodies: Using antibodies targeting different epitopes of PPIL4 can confirm specificity. Available options include antibodies targeting:
Immunogen comparison: Compare your experimental results with the specific immunogen sequence used to generate your antibody. For example, the sequence "EKEDEDYMPI KNTNQDIYRE MGFGHYEEEE SCWEKQKSEK RDRTQNRSRS RSRERDGHYS NSHKSKYQTD LY" is the immunogen for one monoclonal antibody .
Detecting PPIL4 across tissue types requires optimization based on expression levels and tissue characteristics:
Sample preparation:
For mouse kidney tissue (validated source ): Complete protease inhibitor cocktail is essential during homogenization
For cell lines: Lysis in RIPA buffer supplemented with phosphatase inhibitors improves detection
For human tissues: Antigen retrieval methods may need adjustment based on fixation method
Antibody selection based on tissue:
Protocol modifications:
Detection systems:
Achieving reproducible immunofluorescence results with PPIL4 antibodies depends on several critical parameters:
Fixation method:
Paraformaldehyde (4%) for 15 minutes at room temperature preserves epitope accessibility
Methanol fixation (-20°C, 10 minutes) may enhance detection of certain epitopes
Avoid over-fixation which can mask epitopes
Antibody dilution optimization:
Permeabilization protocol:
0.1-0.3% Triton X-100 for 5-10 minutes is typically sufficient
Saponin (0.1%) provides gentler permeabilization for some epitopes
Inadequate permeabilization is a common cause of false negatives
Antigen retrieval considerations:
Heat-induced epitope retrieval may be necessary for some fixation methods
Citrate buffer (pH 6.0) or Tris-EDTA (pH 9.0) are common retrieval solutions
Optimization of retrieval time is critical (typically 10-20 minutes)
Controls and validation:
The following protocol has been optimized for reliable PPIL4 detection by Western blotting:
Sample preparation:
Gel electrophoresis and transfer:
10% SDS-PAGE gel provides optimal resolution for 58-65 kDa PPIL4
Transfer to PVDF membrane at 100V for 60-90 minutes or 30V overnight at 4°C
Verify transfer with reversible Ponceau S staining
Blocking and antibody incubation:
Detection and imaging:
Validation controls:
Successful immunoprecipitation of PPIL4 requires careful optimization:
Lysate preparation:
Antibody binding:
Bead selection and binding:
For rabbit host antibodies: Protein A or Protein A/G mix beads
For mouse host antibodies: Protein G beads
Add pre-washed beads and incubate 1-4 hours at 4°C
Washing and elution:
Wash 3-5 times with cold lysis buffer
Additional high-salt wash can reduce non-specific binding
Elute with Laemmli buffer at 95°C for 5 minutes
Controls:
Input control (5-10% of lysate used for IP)
IgG control from same species as primary antibody
IP-Western validation using a different PPIL4 antibody targeting another epitope
When facing inconsistent results with PPIL4 antibodies, systematic troubleshooting approaches can help identify and resolve issues:
Antibody validation and storage issues:
Sample-specific considerations:
Protein degradation: Ensure complete protease inhibitor cocktail use
Post-translational modifications: Consider phosphatase inhibitors if phosphorylation affects epitope recognition
Expression levels: Different cell lines show variable PPIL4 expression; HeLa and HEK-293 are reliable positive controls
Technical modifications:
For weak signals: Extended primary antibody incubation (overnight at 4°C)
For high background: Increase blocking time and washing steps
For multiple bands: Try reducing agent concentration adjustment or different antibody targeting another epitope
Cross-validation approaches:
Use multiple antibodies targeting different PPIL4 epitopes
Combine detection methods (e.g., validate WB results with IF)
Consider RNA-level validation (qPCR) to confirm protein-level observations
Application-specific adjustments:
PPIL4 has potential roles in RNA processing, making its antibodies valuable tools in this research area:
Co-immunoprecipitation studies:
RNA immunoprecipitation (RIP):
Cross-link RNA-protein complexes with formaldehyde or UV
Immunoprecipitate with PPIL4 antibodies (1-4 μg per reaction)
Extract and analyze bound RNAs by sequencing or qPCR
Include appropriate controls (IgG, input RNA)
Immunofluorescence co-localization:
Cellular fractionation validation:
RNA processing under stress conditions:
Expose cells to transcriptional inhibitors or stress conditions
Monitor PPIL4 redistribution using immunofluorescence
Compare results across cell types with different PPIL4 expression levels
When working with PPIL4 antibodies across species, several important considerations must be addressed:
Validated species reactivity:
Epitope conservation analysis:
Application-specific validation:
Western blot: Start with 25 μg protein per lane for initial cross-species testing
Immunohistochemistry: May require species-specific optimization of antigen retrieval methods
Immunofluorescence: Fixation protocols may need adjustment based on species and tissue type
Control selection:
Protocol modifications:
Primary antibody concentration may need adjustment (typically 1.5-2× higher for non-validated species)
Extended incubation times may improve detection in non-validated species
Secondary antibody selection should match the host species of primary antibody
For reliable quantitative analysis of PPIL4 using antibody-based methods, follow these best practices:
Western blot quantification:
Use gradient loading to establish linear detection range
Include recombinant standards when absolute quantification is needed
Normalize to multiple housekeeping proteins (e.g., GAPDH, β-actin)
Use fluorescent secondary antibodies rather than chemiluminescence for wider linear range
Perform at least three biological replicates with consistent loading amounts (25 μg recommended)
Immunofluorescence quantification:
Standardize image acquisition parameters (exposure, gain, offset)
Include fluorescence standards for calibration
Analyze multiple fields (>5) and cells (>50) per condition
Use appropriate software for unbiased quantification
Control for cell size and morphology variations
ELISA development:
Use purified recombinant PPIL4 to generate standard curves
Optimize antibody concentrations (capture and detection)
Validate specificity using knockout or knockdown samples
Assess matrix effects from different sample types
Determine limits of detection and quantification
Normalization strategies:
For cell-based assays: Normalize to cell number or total protein
For tissue analysis: Consider section thickness and area measurement
For Western blots: Total protein staining (e.g., Ponceau S) may provide better normalization than single reference proteins
Statistical considerations:
Power analysis to determine appropriate sample sizes
Account for technical and biological variation
Apply appropriate statistical tests based on data distribution
Consider batch effects in multi-experiment analyses
PPIL4 antibodies can be integrated into several advanced imaging approaches:
Super-resolution microscopy:
Live-cell imaging approaches:
For indirect visualization: Express PPIL4 with small epitope tags (FLAG, HA)
Use fluorescently labeled nanobodies against tags for live imaging
Validate with fixed-cell immunofluorescence using PPIL4 antibodies
Consider photobleaching concerns for long-term imaging
Proximity ligation assay (PLA):
Combine PPIL4 antibodies with antibodies against potential interaction partners
Select antibodies from different host species (e.g., rabbit anti-PPIL4 with mouse anti-partner)
Optimize antibody dilutions specifically for PLA (typically higher concentrations than standard IF)
Include appropriate controls (single antibody, non-interacting protein pairs)
Correlative light and electron microscopy (CLEM):
Immunogold labeling with PPIL4 antibodies for electron microscopy
Correlate with fluorescence microscopy data
Requires specialized sample preparation and high-specificity antibodies
Consider pre-embedding vs. post-embedding labeling approaches
Tissue clearing and 3D imaging:
Optimize antibody penetration in cleared tissue samples
Extended incubation times (48-72 hours) with higher antibody concentrations
Validate with conventional thin-section immunofluorescence
Account for increased background in thick samples
Studying PPIL4 in primary cells introduces several considerations distinct from established cell lines:
Expression level variations:
Primary cells typically show lower PPIL4 expression than immortalized lines
Increase protein loading (40-50 μg) for Western blot detection
Use more sensitive detection methods (ECL Prime or fluorescent secondaries)
Consider signal amplification systems for immunofluorescence
Antibody validation strategy:
Always include positive control cell lines (HeLa, HEK-293) alongside primary cells
Validate primary cell-specific findings with multiple antibodies targeting different epitopes
Consider genetic approaches (siRNA) to confirm specificity in primary cells
Account for donor-to-donor variability in primary human cells
Protocol adaptations:
Primary cell fixation may require gentler conditions (2% PFA rather than 4%)
Reduce detergent concentration for permeabilization (0.1% Triton X-100)
Extended blocking times to reduce background (2 hours minimum)
Longer primary antibody incubation (overnight at 4°C) for Western blot and immunofluorescence
Primary cell-specific controls:
Age-matched samples for primary cells from different donors
Passage-matched cells for experiments over time
Tissue-specific positive and negative controls
Consider the impact of isolation methods on protein expression and epitope accessibility
Technical challenges:
Limited material availability may necessitate micro-scale protocols
Increased heterogeneity requires larger sample sizes for statistical power
Sensitivity to culture conditions may affect PPIL4 expression or localization
Shorter experimental windows due to limited passage potential
Current PPIL4 antibodies present several limitations that researchers should be aware of:
Epitope coverage limitations:
Specificity concerns:
Cross-reactivity with related cyclophilin family proteins has been reported
Validation across multiple applications remains incomplete for many antibodies
Future approaches should include systematic validation with PPIL4 knockout controls
Comparative analysis of multiple antibodies is recommended for critical experiments
Application restrictions:
Limited validation for immunohistochemistry on formalin-fixed tissues
Minimal data on chromatin immunoprecipitation applications
Future validation efforts should expand application range
Community-based validation reporting would accelerate progress
Species reactivity limitations:
Technical improvements needed:
Development of monoclonal antibodies for better lot-to-lot consistency
Creation of application-specific antibodies (e.g., ChIP-grade, IHC-specific)
Conjugated versions for multi-parameter flow cytometry
Nanobody or recombinant antibody approaches for enhanced reproducibility
PPIL4 antibodies can significantly contribute to disease mechanism research in several areas:
RNA processing disorders:
PPIL4 antibodies can help characterize aberrant splicing mechanisms
Immunoprecipitation followed by RNA-seq can identify disease-specific RNA interactions
Tissue microarray analysis can assess PPIL4 expression changes across disease states
Recommended approach: Combine 1:500 dilution Western blot with RNA-protein interaction studies
Cancer research applications:
Expression profiling across tumor types (currently validated in several cancer cell lines)
Correlation of subcellular localization with disease progression
Analysis of post-translational modifications in cancer contexts
Patient-derived xenograft models can be analyzed with human-specific PPIL4 antibodies
Neurodegenerative disease research:
Protein aggregation studies may benefit from PPIL4 antibodies
Co-localization with disease-specific protein aggregates
Analysis of stress granule association during cellular stress
Brain region-specific expression analysis requires careful antibody validation
Developmental biology:
Tracking PPIL4 expression during differentiation processes
Analysis of protein-protein interaction networks during development
Combined with lineage markers for tissue-specific expression patterns
Knockout models can be validated using existing antibodies
Therapeutic development:
Target engagement studies for compounds affecting PPIL4 function
Biomarker development for diseases with altered PPIL4 expression
Cellular assays for high-throughput screening of PPIL4 modulators
Proximity-based assays to monitor drug-induced conformational changes