Applications : Immunofluorescence (IF)
Sample type: human cell
Review: Immunofluorescence using the WFDC8 antibody was performed in both human and mouse epididymis and testis tissues, and in mouse sperm and extracellular vesicles isolated from the epididymis (epididymosomes). We found online that CUSABIO had an antibody against WFDC8 and with species reactivity in humans. Derived from our results, we demonstrated through immunofluorescence that this antibody against WFDC8 works for humans and mice.
WFDC8 belongs to the WAP-type four-disulfide core (WFDC) domain family. The WFDC domain contains eight cysteines that form four disulfide bonds at the protein's core, functioning primarily as a protease inhibitor. The WFDC8 protein contains a Kunitz-inhibitor domain and three WFDC domains, making it structurally unique within its family. Most WFDC genes are localized to chromosome 20q12-q13 in two clusters (centromeric and telomeric), with WFDC8 belonging to the telomeric cluster . Research interest in WFDC8 has increased due to its association with Kallmann Syndrome and its functional roles in protease inhibition, particularly through its serine-type endopeptidase inhibitor activity and peptidase inhibitor activity . SPINT1 is recognized as an important paralog of this gene, suggesting potential functional relationships worthy of investigation .
Based on current research resources, multiple types of WFDC8 antibodies are available for different experimental applications. These primarily include polyclonal antibodies targeting the human WFDC8 protein. The Atlas Antibodies collection offers rabbit polyclonal anti-WFDC8 antibodies at 0.2 mg/ml concentration, designed specifically for human WFDC8 research . Abbexa also provides WFDC8 antibodies applicable for multiple applications and available in various quantities ranging from 20 μg to 1 mg . Most commercially available WFDC8 antibodies are non-conjugated, though this should be verified when selecting an antibody for specific applications . It's important to note that while there are numerous antibodies for other WFDC family members (particularly WFDC1 and WFDC2), specific WFDC8 antibodies are more limited but highly targeted for research applications.
WFDC8 antibodies support multiple research applications crucial for investigating this protein's expression, localization, and function. According to available sources, WFDC8 antibodies are validated for use in:
These applications enable researchers to investigate WFDC8 expression patterns across different tissues, subcellular localization, protein-protein interactions, and functional relationships with other proteins. Selecting the appropriate application depends on your specific research question and experimental design requirements.
Antibody specificity verification is critical for generating reliable research data. For WFDC8 antibodies, consider implementing these validation approaches:
Western blot analysis using positive and negative control samples (tissues/cells known to express or not express WFDC8)
Immunoprecipitation followed by mass spectrometry confirmation
RNA interference (siRNA or shRNA against WFDC8) to create knockdown controls
Use of recombinant WFDC8 protein as a competitive inhibitor in your application
Cross-validation with a second antibody targeting a different epitope of WFDC8
Many manufacturers provide validation data for their antibodies. For example, Atlas Antibodies states that their antibodies undergo rigorous validation in IHC, ICC-IF, and WB applications to ensure specificity and reproducibility . When selecting a WFDC8 antibody, examine the manufacturer's validation data and consider performing additional validation steps specific to your experimental system.
When using WFDC8 antibodies for Western blotting, consider these optimized methodological parameters:
Remember that these conditions should be optimized for your specific experimental setup and antibody source. Always perform preliminary experiments to determine the optimal conditions for your particular research context.
When facing weak or absent signals in immunohistochemistry with WFDC8 antibodies, consider these methodological troubleshooting steps:
Antigen retrieval optimization:
Try different antigen retrieval methods (heat-induced epitope retrieval using citrate buffer pH 6.0 vs. EDTA buffer pH 9.0)
Extend retrieval time from standard 20 minutes to 30-40 minutes
Ensure complete cooling before proceeding to blocking step
Antibody concentration adjustment:
Perform a titration experiment using different antibody dilutions (1:50, 1:100, 1:200, 1:500)
Extend primary antibody incubation time to overnight at 4°C instead of standard 1-2 hours
Signal amplification strategies:
Implement tyramide signal amplification (TSA) for significantly enhanced sensitivity
Consider using polymer-based detection systems rather than standard ABC methods
Tissue preparation considerations:
Ensure tissue fixation was appropriate (overfixation can mask epitopes)
Use freshly cut tissue sections (aged sections may have decreased antigenicity)
Validate the presence of the target using an alternative method (e.g., RT-PCR)
Controls and validation:
Always include positive control tissue known to express WFDC8
Implement dual staining with a different WFDC8 antibody to confirm expression patterns
If signal remains problematic despite these optimizations, consider alternative detection methods such as RNA in situ hybridization to confirm expression patterns at the mRNA level before revisiting protein detection strategies.
Investigating WFDC8 protein interactions requires careful experimental design. Consider these methodological approaches:
Co-immunoprecipitation (Co-IP) strategies:
Use gentle lysis buffers (e.g., 0.5% NP-40) to preserve protein-protein interactions
Perform reciprocal Co-IPs (IP with anti-WFDC8 followed by Western blot for interaction partner, and vice versa)
Include appropriate negative controls (IgG control, lysates from cells with WFDC8 knockdown)
Consider crosslinking approaches for transient interactions
Proximity ligation assays (PLA):
Effective for visualizing protein interactions in situ with single-molecule sensitivity
Requires two primary antibodies raised in different species (one for WFDC8, one for interaction partner)
Provides spatial information about where interactions occur within cells or tissues
Bimolecular fluorescence complementation (BiFC):
Allows visualization of protein interactions in living cells
Requires genetic fusion of potential interaction partners with complementary fragments of a fluorescent protein
Particularly useful for dynamic studies of WFDC8 interactions
Pull-down assays with recombinant proteins:
Use purified recombinant WFDC8 as bait protein
Can identify direct physical interactions without cellular context
Useful for confirming interactions identified through other methods
Protease inhibition activity assays:
Since WFDC8 functions as a protease inhibitor, design functional assays measuring inhibition of candidate proteases
Use fluorogenic or chromogenic substrates to measure protease activity in presence/absence of WFDC8
Determine kinetic parameters (Ki values) to quantify interaction strength
When designing these experiments, consider that WFDC8 contains multiple domains, including three WFDC domains and a Kunitz-inhibitor domain , each potentially mediating different protein interactions. Domain-specific mutants can help identify which regions are responsible for specific interactions.
Designing robust experiments to investigate WFDC8 expression patterns requires a multi-method approach:
Tissue selection strategy:
Include tissues from multiple organ systems with emphasis on reproductive tissues, immune tissues, and epithelial surfaces
Consider developmental stages if investigating temporal expression patterns
Include tissues known to express other WFDC family members for comparative analysis
Methodological approach combination:
Validation through orthogonal methods:
RNA-seq for comprehensive transcriptional profiling
In situ hybridization to confirm cellular localization of mRNA
Mass spectrometry for unbiased protein detection and quantification
Controls and standards implementation:
Include tissues from WFDC8 knockout models (if available) as negative controls
Use recombinant WFDC8 protein as positive control for antibody-based methods
Apply consistent quantification standards across different tissue types
Data normalization approach:
Normalize protein expression to total protein content rather than single housekeeping proteins
Use multiple reference genes for qRT-PCR normalization
Apply appropriate statistical analysis for comparing expression across tissues
By implementing this comprehensive approach, you can generate reliable data on WFDC8 tissue distribution patterns that accounts for potential methodological limitations of any single technique.
Rigorous controls are essential for validating WFDC8 manipulation in functional studies:
Control Type | Purpose | Implementation Method |
---|---|---|
Negative Controls | Establish baseline expression/function | Mock transfection; non-targeting siRNA/shRNA; empty vector controls |
Positive Controls | Confirm experimental system functionality | Known gene with established knockdown/overexpression phenotype |
Rescue Controls | Verify specificity of observed phenotypes | Re-expression of siRNA-resistant WFDC8 construct after knockdown |
Dose-Response Controls | Establish relationship between WFDC8 levels and phenotype | Titration of siRNA/expression vector quantities |
Off-Target Effect Controls | Rule out non-specific effects | Multiple siRNAs targeting different regions of WFDC8 mRNA |
Temporal Controls | Determine kinetics of response | Time-course analysis of phenotype after manipulation |
Additionally, implement these validation strategies:
Expression level verification:
Functional validation:
Assess known WFDC8 functions (protease inhibition) using biochemical assays
Compare results with established phenotypes of other WFDC family members
Specificity confirmation:
Cellular localization monitoring:
Use immunofluorescence with validated antibodies to confirm correct localization of overexpressed protein
Include tagged versions (GFP/FLAG) to monitor expression while verifying tag doesn't disrupt function
These controls ensure that observed phenotypes are specifically attributable to WFDC8 modulation rather than experimental artifacts or off-target effects.
Differentiating functional effects of WFDC8 from other WFDC family members requires careful experimental design:
Domain-specific functional analysis:
Selective targeting strategies:
Design highly specific siRNAs/shRNAs targeting unique regions of WFDC8 mRNA
Utilize CRISPR/Cas9 genome editing for complete WFDC8 knockout
Implement rescue experiments with WFDC8 constructs resistant to siRNA but maintaining function
Comparative expression analysis:
Perform parallel expression analysis of multiple WFDC family members
Create expression correlation matrices across different experimental conditions
Use hierarchical clustering to identify conditions where WFDC8 behaves distinctly from other family members
Protease inhibition profile characterization:
Generate a comprehensive inhibition profile for WFDC8 against a panel of proteases
Compare with published profiles of other WFDC proteins
Identify proteases uniquely or preferentially inhibited by WFDC8
Protein interaction network mapping:
Perform immunoprecipitation followed by mass spectrometry (IP-MS) to identify WFDC8-specific interaction partners
Compare with interaction networks of other WFDC family members
Focus functional studies on pathways uniquely regulated by WFDC8
By implementing these approaches, you can establish functional specificities of WFDC8 distinct from other family members, leading to more precise understanding of its biological roles.
Post-translational modifications (PTMs) can significantly impact both antibody recognition and biological function of WFDC8. Consider these methodological approaches:
PTM identification strategies:
Mass spectrometry analysis of immunoprecipitated endogenous WFDC8
Phospho-specific antibody screening if phosphorylation is suspected
Glycosylation analysis using glycosidase treatments followed by mobility shift assessment
Ubiquitination studies using ubiquitin-specific antibodies or tandem ubiquitin binding entities (TUBEs)
Epitope mapping protocols:
Generate a panel of WFDC8 fragments to determine antibody binding regions
Create site-directed mutants of predicted PTM sites and assess antibody binding
Perform peptide competition assays with modified and unmodified peptides
Functional impact assessment:
Compare activity of native versus deglycosylated/dephosphorylated WFDC8
Generate non-modifiable mutants (e.g., S/T→A for phosphorylation, N→Q for N-glycosylation)
Analyze subcellular localization of PTM-deficient mutants versus wild-type protein
Antibody selection strategy for PTM research:
Choose antibodies raised against recombinant proteins (likely lacking PTMs) versus those against synthetic peptides
Validate antibody recognition using in vitro modified recombinant proteins
Consider using multiple antibodies targeting different epitopes when studying heavily modified proteins
Temporal regulation investigation:
Assess PTM patterns under different cellular conditions (stress, differentiation, etc.)
Correlate changes in PTM status with functional activities
Identify enzymes responsible for adding/removing specific modifications
These approaches will help determine whether PTMs are critical for WFDC8 function and whether they influence experimental outcomes due to differential antibody recognition.
Investigating WFDC8's role in inflammatory and immune processes requires multifaceted experimental design:
Expression regulation in inflammatory contexts:
Assess WFDC8 expression in immune cells (macrophages, neutrophils, epithelial cells) after stimulation with:
Pathogen-associated molecular patterns (PAMPs): LPS, poly(I:C), flagellin
Pro-inflammatory cytokines: TNF-α, IL-1β, IL-6
Anti-inflammatory cytokines: IL-10, TGF-β
Perform time-course and dose-response experiments
Use Western blot with validated antibodies and qRT-PCR for quantification
Functional assessment in inflammatory models:
In vitro models:
WFDC8 overexpression/knockdown in macrophages followed by cytokine profiling
Neutrophil extracellular trap (NET) formation assays with recombinant WFDC8
Bacterial killing assays in presence/absence of WFDC8
Ex vivo models:
Precision-cut tissue slices treated with recombinant WFDC8
Primary cell cultures from WFDC8 knockout models (if available)
In vivo models:
Assess local and systemic inflammation in WFDC8-deficient animal models
LPS challenge studies with WFDC8 administration
Mechanistic investigations:
Identify specific proteases inhibited by WFDC8 in inflammatory contexts
Investigate potential non-protease inhibitory functions (similar to other WFDC family members)
Assess impact of WFDC8 on inflammatory signaling pathways (NF-κB, MAPK, etc.)
Translational relevance assessment:
Analyze WFDC8 levels in patient samples from inflammatory conditions
Correlate WFDC8 expression with disease severity or outcomes
Compare with known inflammatory biomarkers
This comprehensive approach will help elucidate whether WFDC8 has significant immunomodulatory functions similar to other WFDC family members while establishing its unique contributions to immune regulation.
Investigating WFDC8's role in cancer requires systematic experimental approaches:
Expression profile characterization:
Analyze WFDC8 expression across cancer types using:
Correlate expression with clinicopathological parameters and patient outcomes
Functional assessment in cancer models:
In vitro functional assays after WFDC8 modulation:
Proliferation assays (MTT, BrdU incorporation)
Migration and invasion assays (Transwell, wound healing)
Colony formation and soft agar growth
Apoptosis assessment (Annexin V, TUNEL)
In vivo tumor models:
Xenograft studies with WFDC8-overexpressing or WFDC8-knockdown cancer cells
Patient-derived xenografts treated with recombinant WFDC8
Metastasis models to assess impact on cancer dissemination
Mechanistic investigations:
Protease regulation:
Identify cancer-relevant proteases inhibited by WFDC8
Focus on proteases involved in matrix remodeling and metastasis
Signaling pathway analysis:
Assess impact on growth factor signaling (EGFR, FGFR, etc.)
Investigate effects on PI3K/AKT, MAPK, and WNT pathways
Study potential interaction with tumor suppressor pathways
Tumor microenvironment effects:
Evaluate impact on tumor-associated macrophage function
Assess effects on angiogenesis and lymphangiogenesis
Analyze influence on immune cell infiltration and function
Comparative analysis with other WFDC family members:
Compare WFDC8 cancer-related functions with WFDC1 (ps20, a known tumor suppressor)
Contrast with WFDC2 (HE4, a known oncogene and ovarian cancer biomarker)
Develop integrated models of WFDC family function in cancer contexts
This multifaceted approach will establish whether WFDC8 functions primarily as a tumor suppressor (like WFDC1) or promotes cancer progression (like WFDC2), informing its potential as a therapeutic target or biomarker.
Addressing cross-reactivity concerns with WFDC8 antibodies requires systematic validation and experimental design:
Comprehensive specificity testing:
Test antibodies against recombinant proteins of all WFDC family members
Perform Western blots using lysates from cells overexpressing individual WFDC proteins
Conduct peptide blocking experiments with specific peptides from WFDC8 and closely related family members
Implement knockout/knockdown controls for WFDC8 and related proteins
Epitope analysis and selection:
Select antibodies targeting unique regions of WFDC8 with minimal sequence homology to other WFDC proteins
Analyze epitope sequences using multiple sequence alignment tools
Consider custom antibody development against highly specific WFDC8 regions if commercial options show cross-reactivity
Application-specific validation protocols:
For immunohistochemistry: Perform parallel staining with multiple antibodies targeting different WFDC8 epitopes
For Western blotting: Use precise molecular weight differentiation and include positive controls
For immunoprecipitation: Validate pulled-down proteins by mass spectrometry
Combined detection approaches:
Implement dual-labeling approaches (e.g., IF for protein combined with FISH for mRNA)
Correlate antibody staining patterns with known mRNA expression data from RNA-seq or qRT-PCR
Use proximity ligation assay (PLA) with two different WFDC8 antibodies for highly specific detection
Data interpretation guidelines:
Establish clear criteria for positive WFDC8 staining based on validated controls
Document potential cross-reactivity in your experimental reports
Consider multiple methods to confirm key findings
By implementing these rigorous approaches, you can minimize cross-reactivity concerns and generate reliable data on WFDC8 expression and function in complex biological systems.
Proper storage and handling of WFDC8 antibodies is critical for maintaining their performance and extending their useful lifespan:
Storage temperature recommendations:
Aliquoting protocol:
Create small, single-experiment aliquots upon receiving the antibody
Use sterile conditions during aliquoting to prevent contamination
Include carrier protein (0.1% BSA) if diluting before aliquoting
Label tubes with antibody details, concentration, and date
Buffer considerations:
Maintain recommended buffer conditions (typically PBS with preservative)
Consider adding sodium azide (0.02%) to prevent microbial growth
Some applications may require azide-free antibody preparations
Handling precautions:
Minimize exposure to light for fluorophore-conjugated antibodies
Avoid vortexing antibodies; mix by gentle inversion or pipetting
Use clean, nuclease-free tubes for storage
Handle at appropriate temperatures (cold room or on ice)
Performance monitoring strategy:
Include positive controls in each experiment to monitor antibody performance
Document lot numbers and maintain records of performance
Consider implementing a validation protocol for new lots
Track signal intensity over time to detect potential degradation
Shipping and temporary storage considerations:
Use ice packs for short-term transport
For longer shipping, use dry ice
Upon receipt, immediately transfer to appropriate long-term storage
Following these guidelines will help ensure consistent performance of WFDC8 antibodies across experiments and extend their functional lifespan, improving experimental reproducibility and reducing costs associated with antibody replacement.
When facing contradictory results between different methods of WFDC8 detection, implement this systematic troubleshooting approach:
Method-specific limitations assessment:
Antibody-based methods: Evaluate specificity, sensitivity, and potential cross-reactivity issues
mRNA detection: Consider post-transcriptional regulation differences between mRNA and protein levels
Mass spectrometry: Assess sample preparation compatibility with WFDC8 detection
Document technical parameters that might affect detection (fixation methods, extraction protocols)
Sample-specific considerations:
Evaluate tissue/cell heterogeneity that might explain discrepancies
Consider subcellular localization differences that might affect detection by different methods
Assess potential post-translational modifications affecting antibody recognition
Evaluate potential context-dependent expression differences in your experimental system
Validation through orthogonal approaches:
Implement at least three independent detection methods:
Protein level: Western blot, immunohistochemistry, ELISA
mRNA level: qRT-PCR, RNA-seq, in situ hybridization
Functional approaches: Activity assays specific to WFDC8 function
Use genetic manipulation (overexpression/knockdown) to create defined controls
Data integration and reconciliation strategy:
Create a comprehensive table documenting all results and methodological details
Identify patterns in discrepancies (e.g., consistent issues with particular methods)
Weight evidence based on methodological strengths and limitations
Consider biological explanations for apparent contradictions:
Alternative splicing affecting epitope availability
Tissue-specific post-translational modifications
Context-dependent protein stability or turnover
External validation resources:
Compare your findings with published literature and public databases
Consult with experts in WFDC protein biology or specific methodologies
Consider collaborative verification of key findings using complementary expertise
Through this systematic approach, you can resolve contradictions between methods and develop a more accurate understanding of WFDC8 expression in your experimental system. In publications, transparently report discrepancies and provide your interpretation of these differences rather than selectively reporting concordant results.