The DCAF6 Antibody targets the DCAF6 protein, encoded by the DCAF6 gene located on human chromosome 1 (NC_000001.11) . This gene product functions as a ligand-dependent coactivator for nuclear receptors, including NR3C1 (glucocorticoid receptor), facilitating transcriptional activation in response to steroid hormones . DCAF6 interacts with DDB1 and CUL4, components of the E3 ubiquitin ligase complex, suggesting roles in protein stability and chromatin remodeling .
This antibody is validated for:
Immunofluorescence: Subcellular localization studies in human tissues and cancer cells.
Immunohistochemistry: Detection in normal and cancerous tissues via the Human Protein Atlas project .
Protein Array Analysis: Cross-reactivity testing against 364 human recombinant proteins .
While direct preclinical or clinical data on the DCAF6 Antibody is limited, its utility lies in studying DCAF6’s role in nuclear receptor signaling and cancer biology. Emerging evidence suggests:
DCAF6, also known as Nuclear Receptor Interaction Protein (NRIP) or IQ motif and WD repeat-containing protein 1 (IQWD1), is an 860 amino acid protein that localizes to the nucleus and contains one IQ domain and seven WD-repeats . This protein functions as a ligand-dependent coactivator of nuclear receptors and specifically enhances the transcriptional activity of androgen receptor (AR) and glucocorticoid receptor (GR) . DCAF6 is expressed in testis, skeletal muscle, prostate, and heart .
Its importance stems from its role in hormone signaling pathways, particularly in androgen receptor signaling, which has implications for prostate cancer research and other hormone-dependent conditions. Northern Blot analysis has detected high expression of NRIP in skeletal muscle and testis with lower expression in heart, prostate, and adrenal gland .
Currently available DCAF6 antibodies exhibit several common characteristics:
Most DCAF6 antibodies require validation for specific applications and cell/tissue types of interest.
The choice of application method depends on your research question:
Immunohistochemistry (IHC): Optimal for localizing DCAF6 in tissue sections. Verified samples include human colon cancer and human brain tissues . Typical dilutions range from 1:50-1:200 .
Western Blotting (WB): Effective for determining protein expression levels and molecular weight. Several antibodies are specifically validated for this purpose .
Immunofluorescence (IF): Ideal for subcellular localization studies, confirming nuclear localization of DCAF6 .
ELISA: Useful for quantitative detection, with recommended dilutions ranging from 1:2000-1:5000 .
Immunoprecipitation (IP): Valuable for studying protein-protein interactions, particularly for investigating DCAF6's interaction with nuclear receptors .
The nuclear localization of DCAF6 makes nuclear isolation and proper fixation particularly important in all protocols .
For optimal stability and activity:
Avoid repeated freeze-thaw cycles which can cause protein denaturation and loss of activity .
Most DCAF6 antibodies are supplied in glycerol-containing buffers (typically 40-50% glycerol) to prevent freeze damage .
When shipped with ice packs, immediately store antibodies at the recommended temperature upon receipt .
Working dilutions can be prepared and stored at 4°C for short periods (1-2 weeks), but should be aliquoted and frozen for longer storage .
Some products have validated stability of 12 months when properly stored .
Rigorous validation is critical due to potential specificity issues with antibodies:
Knockout/Knockdown Controls: The gold standard for validation. Test the antibody in DCAF6 knockout or knockdown cell lines to confirm signal absence/reduction .
Multiple Antibody Approach: Use at least two different antibodies targeting different epitopes of DCAF6 and compare their staining/banding patterns .
Peptide Competition Assay: Pre-incubate the antibody with the immunizing peptide to confirm signal blockage.
Cross-Species Reactivity Testing: If your antibody is claimed to work across species, validate in each species separately .
Recombinant Protein Controls: Use recombinant DCAF6 proteins as positive controls in your assays .
As noted in recent reproducibility initiatives like YCharOS (Antibody Characterization through Open Science), standardized characterization processes involving knockout cell lines have become critical for evaluating antibody specificity .
When studying DCAF6 interactions with nuclear receptors like AR:
Nuclear Extraction Optimization:
Use specialized nuclear extraction buffers containing DNase/RNase to disrupt chromatin associations
Include phosphatase inhibitors to preserve phosphorylation states that may be critical for interactions
Co-IP Protocol Refinements:
Use mild detergents (0.1-0.3% NP-40 or Triton X-100) to preserve protein complexes
Consider crosslinking with formaldehyde (0.1-0.3%) to stabilize transient interactions
Rotate samples at 4°C overnight for complete antibody binding
Buffer Considerations:
Include 10-20 mM sodium molybdate to stabilize steroid receptor complexes
Add protease inhibitors to prevent degradation during long incubations
Antibody Selection:
Controls:
Include hormone-treated versus untreated samples (DCAF6 functions as a ligand-dependent coactivator)
Use IgG controls matched to your primary antibody species
This approach has successfully identified DCAF6-AR interactions in previous studies .
DCAF6 exists in three isoforms produced by alternative splicing events , creating specific detection challenges:
Isoform-Specific Detection Challenges:
Commercially available antibodies may not distinguish between the three DCAF6 isoforms
Isoforms may have different subcellular distributions or tissue expression patterns
Methodological Solutions:
Epitope Mapping: Select antibodies targeting regions uniquely present or absent in specific isoforms
Molecular Weight Resolution: Use high-resolution SDS-PAGE (8-10% gels) to separate isoforms by size
2D Electrophoresis: Combine isoelectric focusing with SDS-PAGE for improved isoform separation
RT-PCR Validation: Confirm isoform expression at the mRNA level prior to protein detection
Controls and Validation:
Use recombinant proteins of each isoform as positive controls
Consider generating isoform-specific knockouts or overexpression systems
Perform phosphatase treatments to eliminate post-translational modifications that may complicate isoform identification
Interpretation Guidelines:
Understanding the structural basis of antibody-antigen recognition can significantly enhance DCAF6 antibody applications:
Complementarity Determining Regions (CDRs) and Epitope Recognition:
While the six hypervariable loops (CDRs) within antibody variable domains are traditionally considered responsible for antigen recognition, recent structural analyses suggest this is an oversimplification
Some CDR positions never participate in antigen binding, while some non-CDR residues critically contribute to antigen interaction
Implications for DCAF6 Antibody Design:
Advanced Application Approaches:
Emerging Technologies:
When faced with conflicting DCAF6 antibody results:
Systematic Troubleshooting Protocol:
Document all experimental variables (antibody clone, lot, dilution, incubation time/temperature)
Compare epitope regions of different antibodies yielding conflicting results
Analyze buffer compositions that may affect epitope accessibility
Biological Source Considerations:
DCAF6 expression and localization may vary by cell type - verify baseline expression
Post-translational modifications may differ between cell types, affecting antibody recognition
Consider species-specific differences in DCAF6 sequence and structure
Standardization Approaches:
Advanced Analytical Methods:
Mass spectrometry validation of DCAF6 presence/absence in your samples
Combine antibody-based detection with orthogonal methods (e.g., RNA expression)
Consider using CRISPR-tagged endogenous DCAF6 as ultimate validation
Community Resources:
Designing robust experimental controls:
Positive Control Selection:
Negative Control Strategy:
DCAF6 knockout or knockdown samples are ideal negative controls
Use tissues with minimal DCAF6 expression as relative negative controls
Include isotype-matched irrelevant antibody controls
Tissue-Specific Considerations:
Adjust fixation protocols based on tissue type (e.g., longer fixation for dense tissues)
Optimize antigen retrieval methods for each tissue type
Consider autofluorescence quenching for highly autofluorescent tissues
Quantitative Controls:
Include gradient dilutions of recombinant DCAF6 protein for quantitative applications
Consider spike-in controls of known quantities in complex samples
Use normalized housekeeping proteins appropriate for each tissue type
When investigating DCAF6's role in androgen receptor signaling:
Hormone Treatment Protocol Design:
Include time-course experiments (30 min, 2h, 6h, 24h) to capture dynamic interactions
Test physiologically relevant androgen concentrations (1-10 nM DHT or testosterone)
Include both hormone-depleted and hormone-treated conditions
Nuclear Translocation Assessment:
Use cellular fractionation to track DCAF6 and AR localization changes
Perform co-immunofluorescence to visualize DCAF6-AR co-localization
Consider live-cell imaging with tagged proteins to monitor dynamic interactions
Transcriptional Readouts:
Measure androgen-responsive gene expression (e.g., PSA, TMPRSS2)
Use reporter assays with androgen-responsive elements
Compare transcriptional output with and without DCAF6 modulation
Interaction-Specific Methodology:
Perform sequential chromatin immunoprecipitation (ChIP-reChIP) to verify co-occupancy
Use proximity ligation assays to confirm direct DCAF6-AR interaction
Consider FRET/BRET approaches for real-time interaction monitoring
Functional Validation Approaches:
Use DCAF6 mutants lacking key domains to map interaction regions
Employ AR mutants to identify interaction surfaces
Consider the three DCAF6 isoforms and their potential differential effects
For cancer research applications:
Tissue Microarray Strategy:
Design tissue microarrays including normal, precancerous, and various cancer stages
Include multiple cancer types, particularly hormone-dependent cancers
Correlate DCAF6 expression patterns with clinical parameters and outcomes
Cell Line Selection Framework:
Choose cell lines representing cancers where nuclear receptor signaling is important
Include isogenic cell line pairs differing in DCAF6 expression/mutation status
Consider patient-derived organoids for more physiologically relevant models
Technical Optimization Approaches:
Functional Investigation Methods:
Assess DCAF6's effect on cancer cell proliferation, migration, and invasion
Investigate DCAF6's potential role in treatment resistance
Examine its interaction with cancer-relevant signaling pathways beyond nuclear receptors
Clinical Correlation Approaches:
Correlate DCAF6 expression with response to hormone therapies
Investigate potential associations with cancer subtypes and progression
Consider DCAF6 as a potential prognostic or predictive biomarker
Managing antibody variability:
Proactive Quality Control Protocol:
Test each new antibody lot against a standardized positive control
Document lot-specific optimal dilutions and performance characteristics
Create internal reference standards for comparative analysis
Standardization Approaches:
Technical Redundancy Strategy:
Maintain overlapping supplies of well-characterized antibody lots
Use multiple antibodies targeting different DCAF6 epitopes
Incorporate non-antibody detection methods as complementary approaches
Statistical Considerations:
Include biological and technical replicates spanning different antibody lots
Apply appropriate statistical methods to account for batch effects
Consider normalization strategies to minimize lot-dependent variation
Documentation Practices:
Maintain detailed records of antibody performance by lot number
Report lot-specific information in publications and repositories
Share batch variability experiences through community resources
Innovative applications include:
Proximity-Based Interaction Mapping:
BioID or TurboID fusion with DCAF6 to identify proximal proteins
APEX2 labeling combined with DCAF6 antibody immunoprecipitation
DCAF6 antibody-based proximity ligation assays to verify predicted interactions
Quantitative Interaction Proteomics:
Stable isotope labeling combined with DCAF6 immunoprecipitation
Label-free quantification of DCAF6 interactomes across conditions
DCAF6 antibody-based cross-linking mass spectrometry (XL-MS)
Dynamic Interaction Visualization:
Super-resolution microscopy with DCAF6 antibodies to track nanoscale interactions
Live-cell imaging using split fluorescent protein complementation
FRAP (Fluorescence Recovery After Photobleaching) combined with DCAF6 antibody staining
Structural Studies:
DCAF6 antibody-facilitated cryo-EM of protein complexes
Hydrogen-deuterium exchange mass spectrometry with DCAF6 antibodies
Single-molecule FRET studies of DCAF6-containing complexes
Systems Biology Integration:
DCAF6 ChIP-seq combined with transcriptomics
Pathway analysis incorporating DCAF6 interactome data
Multi-omics integration centered on DCAF6 function
Common background issues and solutions:
Nonspecific Binding Issues:
Use 3-5% BSA or 5-10% normal serum from the same species as the secondary antibody
Implement additional blocking steps with commercial blocking reagents
Increase washing duration and frequency (3-5 washes of 5-10 minutes each)
Tissue-Specific Challenges:
For tissues with high endogenous biotin (liver, kidney), use avidin-biotin blocking kits
Quench autofluorescence with Sudan Black B (0.1-0.3%) or commercial quenchers
Adjust fixation protocols to minimize epitope masking while preserving structure
Antibody Concentration Optimization:
Technical Protocol Refinements:
Optimize antigen retrieval methods (citrate vs. EDTA buffers, pH adjustments)
Reduce incubation temperature (4°C overnight vs. room temperature)
Consider using specialized detection systems with signal amplification
Validation Controls:
Include peptide competition controls to confirm specific binding
Use DCAF6-negative tissues or knockdown samples to assess background levels
Implement isotype control antibodies at the same concentration
When adapting between fixation methods:
Formaldehyde to Cold Methanol Transition:
Reduce antibody concentration by 25-50% for methanol fixation
Eliminate antigen retrieval steps typically needed for formaldehyde
Increase primary antibody incubation time (overnight at 4°C recommended)
Be aware that methanol may disrupt some conformational epitopes
Paraffin to Frozen Section Adaptation:
Decrease antibody concentration by 30-50% for frozen sections
Implement additional blocking steps to reduce background
Use shorter incubation times for both primary and secondary antibodies
Optimize fixation time post-sectioning (typically 10-20 minutes)
Native to Cross-linked Protein Transition (for WB/IP):
For formaldehyde cross-linked samples, include reversal step (95°C for 5-10 min)
Use different extraction buffers optimized for cross-linked material
Consider specialized detergents for cross-linked nuclear extractions
Adjust gel running conditions for cross-linked complexes
General Considerations Across Methods:
Integrating mass spectrometry approaches:
Antibody Validation Framework:
Novel Interaction Discovery Pipeline:
Perform quantitative IP-MS across different conditions
Compare DCAF6 interactome changes with and without stimulus
Validate top candidates through reciprocal IP and other methods
Post-translational Modification Analysis:
Use DCAF6 antibodies to enrich for the protein prior to MS analysis
Identify phosphorylation, ubiquitination, or other modifications
Correlate modifications with functional changes
Cross-linking Mass Spectrometry Applications:
Apply formaldehyde or specialized cross-linkers before immunoprecipitation
Map interaction interfaces between DCAF6 and binding partners
Develop structural models based on cross-linking constraints
Absolute Quantification Strategy:
Develop absolute quantification methods using isotope-labeled peptides
Compare antibody-based quantification with MS absolute quantification
Establish calibration curves for more accurate protein measurements
Optimizing epitope retrieval:
Heat-Induced Epitope Retrieval (HIER) Optimization:
Compare citrate buffer (pH 6.0) vs. EDTA buffer (pH 9.0)
Test retrieval times (10, 20, 30 minutes)
Evaluate different heating methods (microwave, pressure cooker, water bath)
For DCAF6, EDTA-based buffers at pH 8-9 often perform well with nuclear proteins
Enzymatic Retrieval Considerations:
Test proteinase K treatment (1-10 μg/mL, 5-20 minutes)
Evaluate trypsin digestion (0.05-0.1%, 10-30 minutes at 37°C)
Consider pepsin for highly fixed tissues (0.05-0.1%, 5-15 minutes at 37°C)
Often less effective for nuclear proteins like DCAF6
Combination Approaches:
Sequential protease treatment followed by HIER
Dual buffer systems with pH shifts
Variable temperature cycling protocols
Tissue-Specific Adaptations:
Increase retrieval time for dense tissues
Use gentler conditions for delicate tissue architecture
Adjust protocols based on fixation duration of tissues
Validation Metrics:
Leveraging deep learning for improved analysis:
Automated Localization and Quantification:
Train convolutional neural networks (CNNs) to identify DCAF6-positive nuclei
Implement instance segmentation for individual cell analysis
Develop algorithms for quantifying nuclear vs. cytoplasmic signal ratios
Pattern Recognition Applications:
Use deep learning to identify distinct DCAF6 distribution patterns
Correlate patterns with cell cycle phases or differentiation states
Develop classifiers for normal vs. pathological DCAF6 localization
Multiplexed Analysis Enhancement:
Apply neural networks for spectral unmixing in multiplexed images
Develop co-localization algorithms for DCAF6 and interaction partners
Create spatial relationship maps in tissue contexts
Quality Control Implementation:
Train models to identify technical artifacts vs. true signal
Develop automated background correction algorithms
Create consistency scores across batches and experiments
Integration with Other Data Modalities:
Combine imaging with genomic or transcriptomic data using multimodal networks
Develop predictive models for DCAF6 function based on localization patterns
Create integrated visualization and analysis platforms
These approaches build upon methods similar to those used for antibody repertoire analysis in immunology research , where deep learning has successfully revealed patterns not detectable through conventional analysis.
Leveraging single-cell approaches:
Single-Cell Protein Analysis:
Adapt DCAF6 antibodies for mass cytometry (CyTOF) protocols
Implement single-cell Western blotting for protein quantification
Develop microfluidic antibody capture systems for rare cell analysis
Spatial Transcriptomics Integration:
Combine DCAF6 immunostaining with spatial transcriptomics
Correlate protein localization with mRNA expression patterns
Create multimodal maps of DCAF6 function in tissue contexts
High-Content Imaging Applications:
Apply DCAF6 antibodies in high-content screening platforms
Develop single-cell tracking of DCAF6 dynamics
Implement machine learning classification of cellular responses
Heterogeneity Characterization:
Identify DCAF6 expression/localization subtypes within tissues
Correlate DCAF6 patterns with cell state markers
Develop computational methods to detect rare cell populations with distinct DCAF6 features
Technical Considerations:
Optimize fixation and permeabilization for single-cell suspension compatibility
Develop multiplexed panels including DCAF6 and key interaction partners
Implement quality control metrics specific to single-cell applications
Enhancing specificity:
Epitope Engineering Strategy:
Design immunogens targeting unique regions of DCAF6 not present in related proteins
Use bioinformatic approaches to identify minimally conserved sequences
Develop antibodies against DCAF6-specific post-translational modifications
Advanced Selection Techniques:
Implement negative selection against related proteins during antibody development
Use phage display with stringent selection parameters
Apply deep sequencing to identify rare highly specific antibody clones
Validation Framework:
Test against panels of related WD-repeat proteins
Perform cross-adsorption experiments to remove cross-reactive antibodies
Validate in systems expressing only specific family members
Application-Specific Optimizations:
Use higher stringency washing conditions in immunoblotting
Implement competition assays with related proteins
Develop dual-labeling approaches requiring two epitopes for signal generation
Computational Approaches:
Apply structural modeling to predict cross-reactivity
Use machine learning to optimize antibody selection
Develop specificity prediction algorithms based on epitope characteristics
Specialized methodological approaches:
Complex-Specific Detection Strategy:
Develop proximity ligation assays specific for DCAF6-CUL4-DDB1 association
Use sequential immunoprecipitation to isolate intact complexes
Implement split-luciferase complementation assays for dynamic interaction monitoring
Functional Assessment Approaches:
Develop in vitro ubiquitination assays with immunopurified complexes
Create fluorescent ubiquitin sensors for live-cell imaging
Establish proteasome activity correlations with complex formation
Structural Investigation Methods:
Use antibodies as tools for cryo-EM sample preparation
Develop conformation-specific antibodies that recognize complex-bound DCAF6
Implement hydrogen-deuterium exchange MS to map interaction surfaces
Substrate Identification Pipeline:
Combine DCAF6 antibody immunoprecipitation with ubiquitin remnant profiling
Develop quantitative proteomics workflows for substrate discovery
Create validation systems for candidate substrates
Technical Optimization Approaches:
Adjust lysis conditions to preserve complex integrity
Develop extraction methods that maintain ubiquitination status
Optimize immunoprecipitation protocols for large complex isolation
Ethical and practical framework:
Transparency Requirements:
Reproducibility Practices:
Resource Sharing Guidelines:
Consider depositing validated antibodies in community repositories
Share cell lines or tissues used for validation
Make validation data openly accessible
Quality Assurance Practices:
Implement blinded analysis when possible
Use quantitative metrics for antibody performance
Develop standard operating procedures for your laboratory
Ethical Considerations:
Report negative findings to prevent resource waste
Acknowledge limitations of antibody-based approaches
Consider the environmental and animal welfare impacts of antibody production
These guidelines align with recent initiatives to improve research reproducibility in antibody-based research .
Emerging technologies and their implications:
Recombinant Antibody Development:
Shift toward fully sequenced recombinant antibodies for DCAF6
Implementation of renewable expression systems
Development of humanized antibodies for potential therapeutic applications
Nanobody and Single-Domain Antibody Applications:
Creation of DCAF6-specific nanobodies for super-resolution microscopy
Development of intrabodies for live-cell tracking of DCAF6
Implementation of nanobody-based biosensors for conformational changes
Multispecific Antibody Approaches:
Design of bispecific antibodies targeting DCAF6 and interaction partners
Development of antibody-drug conjugates for targeted functional studies
Creation of antibody-based proximity inducers for controlled interactions
Computational Design Advancements:
Machine learning-guided antibody optimization
Structure-based antibody engineering for improved affinity and specificity
In silico prediction of optimal epitopes and binding characteristics
Integration with Gene Editing Technologies:
CRISPR-based endogenous tagging for validated antibody epitopes
Development of antibodies recognizing edited forms of DCAF6
Creation of synthetic biology systems combining antibody detection with genetic modifications