The antibody has been validated in multiple experimental workflows:
Human Liver: ELF3 expression localized to glandular epithelial nuclei using R&D Systems’ AF5787 (3 µg/mL) .
Prostate Tissue: Mouse monoclonal MAB57871 detects nuclear staining in paraffin-embedded sections .
Cell Lines: ELF3 (~42 kDa) detected in PC-3 (prostate), A549 (lung), and NIH-3T3 (mouse fibroblast) lysates using AF5787 (1 µg/mL) .
Tumor Xenografts: ELF3 overexpression confirmed in A549-derived tumors using MAB57871 .
Cell Signaling’s #31600 antibody isolates ELF3 for downstream analyses, such as chromatin immunoprecipitation (ChIP) .
Ovarian Cancer: ELF3 overexpression inhibits epithelial-mesenchymal transition (EMT) and angiogenesis, correlating with improved patient survival .
Prostate Cancer: Depletion of ELF3 enhances androgen receptor (AR) activity, promoting cell migration and tumor growth .
Genetic Amplification: Focal amplification of the ELF3 locus at 1q32.1 occurs in ~80% of LUAD cases, driving tumor growth .
Functional Studies: ELF3 knockdown reduces viability of LUAD cell lines (HCC827, A549), while rescue experiments restore tumorigenicity .
ELF3 (E74-Like Factor 3), also known as ESE-1, ESX, ERT, EPR-1, and JEN, is a 41-43 kDa member of the ETS family of transcription factors. It plays a crucial role in epithelial cell differentiation during normal homeostasis by repressing genes needed during early differentiation and promoting genes required for terminal differentiation. During inflammatory conditions, ELF3 expression extends beyond epithelial cells to include monocytes, endothelial cells, and chondrocytes, where it regulates the production of molecules such as Ang1 and COX2 . As a transcription factor with context-dependent functions, ELF3 has become an important target in studies of epithelial development, inflammation, and cancer progression.
ELF3 antibodies support numerous experimental applications, with Western Blot (WB), Immunohistochemistry (IHC), and Immunofluorescence (IF) being the most widely validated. Many antibodies also support ELISA, Immunocytochemistry (ICC), Immunoprecipitation (IP), and Chromatin Immunoprecipitation (ChIP) . When selecting an ELF3 antibody, researchers should verify that it has been validated for their specific application, as performance can vary significantly between techniques. For particularly sensitive applications like ChIP-seq, specialized antibodies like Cell Signaling Technology's ELF3 (F8J2G) Rabbit mAb have been specifically validated .
ELF3 antibodies have been validated with various sample types including:
For optimal results, sample preparation protocols should be tailored to both the antibody specifications and the experimental application. Heat-induced epitope retrieval using basic antigen retrieval reagents has been successfully employed for paraffin-embedded tissues prior to IHC with ELF3 antibodies .
For rigorous experimental design with ELF3 antibodies, researchers should include:
Positive controls: Cell lines with known ELF3 expression (e.g., HT-29, PC-3, A549)
Negative controls: Samples where primary antibody is omitted
Specificity controls: Ideally, ELF3 knockout/knockdown samples
Isotype controls: Matching IgG from the same species as the primary antibody
Loading controls: For quantitative Western blot analysis
Some antibodies, such as R&D Systems' Human ELF3 Antibody (MAB57871), have been validated using knockout/knockdown approaches, providing higher confidence in their specificity .
Optimizing ELF3 detection requires consideration of its context-dependent expression patterns:
Epithelial tissues: Use heat-induced epitope retrieval with basic pH buffers (e.g., Antigen Retrieval Reagent-Basic) for paraffin sections . Nuclear staining is expected in glandular epithelial cells.
Inflammatory contexts: When examining ELF3 in non-epithelial cells during inflammation, consider dual staining with cell-type specific markers (e.g., CD68 for macrophages) to confirm identity of ELF3-expressing cells.
Cancer cell lines: Different cancer lines show variable ELF3 expression levels. A549, HT-29, and PC-3 lines are reliable positive controls for human samples , while EL-4 serves as a mouse control.
Antibody concentration optimization: For IHC, starting concentrations around 15 μg/mL have been effective , but titration experiments should be performed for each new tissue type or experimental system.
Detection system selection: For low abundance detection, consider using amplification-based detection systems such as HRP-DAB rather than direct fluorescence methods.
Interpreting ELF3 antibody results presents several challenges:
Multiple protein isoforms: ELF3 can exist in multiple forms, with the main form at approximately 42 kDa . Unexpected bands may represent legitimate isoforms rather than non-specific binding.
Cross-reactivity with ETS family members: The ETS family shares conserved DNA-binding domains. While some antibodies have been tested for cross-reactivity (e.g., no cross-reactivity with ELF5 for certain clones) , comprehensive cross-reactivity profiles are often unavailable.
Context-dependent expression: ELF3 expression changes dramatically during inflammation or disease states, so interpreting expression changes requires careful consideration of the physiological context.
Nuclear vs. cytoplasmic localization: As a transcription factor, ELF3 is typically nuclear, but cytoplasmic localization can occur. Discrepancies in localization patterns between studies may reflect biological differences rather than antibody performance issues.
Signal intensity interpretation: Quantitative comparisons of ELF3 levels between different studies should be approached with caution due to variations in antibody affinity, detection methods, and exposure settings.
The choice between monoclonal and polyclonal ELF3 antibodies significantly impacts experimental outcomes:
For the most critical experiments, researchers may benefit from comparing results with both monoclonal and polyclonal antibodies to ensure robust findings.
When faced with contradictory results between studies using ELF3 antibodies, researchers should consider these resolution approaches:
Antibody validation comparison: Assess whether antibodies were validated by knockout/knockdown controls . Prioritize findings from studies using comprehensively validated antibodies.
Epitope mapping: Different antibodies recognize distinct regions of ELF3 (e.g., N-terminal region vs. full-length protein) . These differences may explain discrepancies if protein processing occurs in certain contexts.
Multi-antibody verification: Confirm key findings using multiple antibodies targeting different ELF3 epitopes. Concordant results with diverse antibodies increase confidence.
Orthogonal techniques: Complement antibody-based detection with non-antibody methods (e.g., mass spectrometry, RNA analysis) to independently verify findings.
Recombinant ELF3 controls: Use recombinant human ELF3 proteins (e.g., Met1-Gly173 fragment) as standards to calibrate detection sensitivity across studies.
Protocol standardization: Implement identical sample preparation, antibody concentration, and detection methods to determine if methodological differences explain contradictory results.
Selection of the optimal ELF3 antibody should follow this systematic approach:
Application compatibility: Verify the antibody has been validated for your specific application (WB, IHC, IF, ChIP, etc.) .
Species reactivity: Confirm reactivity with your species of interest. Some antibodies react with human ELF3 only, while others cross-react with mouse and rat orthologs .
Epitope consideration: For mechanistic studies, select antibodies targeting functional domains relevant to your hypothesis. N-terminal (Met1-Gly173) targeting antibodies are common .
Validation rigor: Prioritize antibodies validated by knockout/knockdown approaches over those validated only by overexpression systems.
Clonality decision: For quantitative studies requiring high reproducibility, monoclonal antibodies offer advantages. For initial detection in complex samples, polyclonal antibodies may provide higher sensitivity.
Format requirements: Consider whether you need unconjugated antibodies or specific conjugates (HRP, fluorophores) based on your detection system.
Literature precedent: Select antibodies with published track records in experimental systems similar to yours.
When encountering issues with ELF3 antibody performance, consider these troubleshooting approaches:
For weak signal:
Antibody concentration: Increase primary antibody concentration incrementally (e.g., from 1 μg/mL to 5-15 μg/mL) .
Epitope retrieval optimization: For IHC/IF, test different antigen retrieval methods and durations. Basic pH buffers have proven effective for some ELF3 antibodies .
Incubation conditions: Extend primary antibody incubation time (overnight at 4°C) or adjust temperature.
Detection system enhancement: Switch to more sensitive detection systems (e.g., tyramide signal amplification).
Sample preparation review: Ensure sample processing maintains ELF3 integrity (avoid excessive fixation for tissues).
For non-specific signal:
Blocking optimization: Increase blocking buffer concentration or duration.
Antibody specificity verification: Test the antibody on known negative controls or ELF3 knockout samples .
Wash stringency increase: Add additional wash steps or detergents to reduce non-specific binding.
Secondary antibody cross-reactivity check: Test secondary antibody alone to rule out non-specific binding.
Cross-adsorption: For polyclonal antibodies with cross-reactivity issues, consider custom cross-adsorption against potential cross-reacting proteins.
For rigorous quantitative analysis of ELF3 expression:
Western blot quantification:
Use recombinant ELF3 standards to create a standard curve
Ensure linear dynamic range of detection
Normalize to appropriate loading controls
Apply consistent image acquisition settings across all samples
Use technical replicates (minimum triplicate) for statistical analysis
IHC/IF quantification:
Implement standardized scoring systems (H-score, Allred score)
Use digital image analysis with consistent thresholds
Include reference standards in each batch
Blind scorers to experimental conditions
Quantify multiple fields per sample (minimum 5-10)
ELISA-based quantification:
Develop sandwich ELISA using validated ELF3 antibody pairs
Include recombinant ELF3 standard curves
Process all samples simultaneously when possible
Account for matrix effects in complex samples
Flow cytometry:
Use isotype controls and fluorescence-minus-one controls
Report relative fluorescence intensity or molecules of equivalent soluble fluorochrome
Standardize using calibration beads between experiments
Controls for normalization:
Include identical reference samples across all experimental batches
Consider normalizing to total protein rather than single housekeeping proteins
Document and report all normalization methods in detail
ELF3's subcellular localization provides critical functional insights:
Nuclear localization: As a transcription factor, nuclear ELF3 typically indicates active transcriptional regulation . Predominantly observed in:
Cytoplasmic localization: May indicate:
Sequestration as a regulatory mechanism
Post-translational modifications affecting nuclear import
Potential non-transcriptional functions
Pathological states in certain disease contexts
Dual localization patterns: Often observed during dynamic cellular processes:
Developmental transitions
Inflammatory responses
Early stages of malignant transformation
Methodological considerations for localization studies:
Fixation methods significantly impact observed localization
Cell fractionation coupled with Western blotting provides quantitative assessment
Co-localization with compartment markers enhances interpretative value
Live-cell imaging with fluorescently-tagged ELF3 can reveal dynamic localization changes
Experimental controls for localization studies:
Include known nuclear proteins (e.g., HDAC1) and cytoplasmic proteins (e.g., GAPDH)
Verify antibody access to different compartments using permeabilization controls
Consider dual staining approaches with organelle-specific markers
ELF3 antibodies have become valuable tools in cancer research across multiple applications:
Diagnostic biomarker development:
Mechanistic studies:
Therapeutic target validation:
Antibody-based detection confirms target engagement in drug development
Monitoring ELF3 levels during treatment response
Validation of ELF3 knockdown/knockout models
Cancer cell line characterization:
Cancer-inflammation interface:
Dual staining with inflammatory markers reveals ELF3's role at the intersection of inflammation and cancer
Recent technological advances are enhancing ELF3 antibody applications:
Antibody engineering improvements:
Validation enhancements:
Application-specific optimizations:
Novel formats and conjugates:
Bifunctional antibodies for proximity ligation assays
Site-specific conjugation methods preserving antigen-binding capacity
Nanobody and aptamer alternatives for applications requiring smaller binding molecules
Reproducibility initiatives:
Standardized reporting of validation parameters
Antibody registry entries with unique identifiers
Independent validation through antibody validation initiatives
ELF3 antibodies provide valuable insights into inflammatory mechanisms:
Cell type-specific expression analysis:
Signaling pathway investigations:
Combine with phospho-specific antibodies against inflammatory mediators
ChIP analysis of ELF3 binding to inflammatory gene promoters
Co-localization with nuclear translocation of NF-κB and other inflammatory transcription factors
Inflammatory mediator regulation:
Tissue-specific inflammatory responses:
Compare ELF3 patterns across multiple inflamed tissues
Evaluate epithelial-specific vs. immune cell-specific ELF3 functions
Study interface between epithelial damage and inflammatory cell recruitment
Chronic inflammation models:
Track ELF3 expression during transition from acute to chronic inflammation
Correlate with tissue remodeling and fibrosis markers
Evaluate as potential biomarker for inflammatory disease progression
Longitudinal studies using ELF3 antibodies require special considerations:
Antibody stability and storage:
Protocol standardization:
Instrument calibration:
Regular calibration of imaging systems and readers
Include calibration standards in each experimental batch
Document all instrument settings for reproducibility
Reference standards:
Prepare master aliquots of positive control samples
Include recombinant ELF3 standards at multiple concentrations
Create standard curves for quantitative analyses
Data normalization strategies:
Establish baseline ELF3 levels for each subject/sample
Use relative rather than absolute quantification when comparing across time points
Include time-matched controls for each experimental time point
Account for batch effects in statistical analyses
Optimal experimental designs to study ELF3's dual functions include:
Temporal monitoring systems:
Time-course analysis of ELF3 expression during epithelial differentiation
Parallel tracking of differentiation markers and inflammatory mediators
Sequential sampling during inflammatory challenge and resolution
Cell type-specific approaches:
Co-staining with epithelial markers (E-cadherin, cytokeratins) and inflammatory cell markers
Cell sorting followed by ELF3 protein quantification
Single-cell analysis correlating ELF3 levels with cell phenotypes
Conditional expression systems:
Inducible ELF3 expression in epithelial and non-epithelial cells
Tissue-specific knockout models evaluated under homeostatic and inflammatory conditions
Domain mutant expression to separate differentiation vs. inflammatory functions
3D culture models:
Epithelial organoids with inflammatory cell co-culture
Air-liquid interface cultures with inflammatory stimulation
Wound healing models to capture transition states
Integrative analysis approaches:
Combined ChIP-seq and RNA-seq to identify context-dependent gene regulation
Proteomics to identify interaction partners in different cellular contexts
Multi-omics integration to map ELF3 function across biological states
Integrated experimental approaches combining ELF3 antibodies with complementary tools:
Multi-omic strategies:
ChIP-seq with ELF3 antibodies combined with RNA-seq and ATAC-seq
Proteomics following ELF3 immunoprecipitation
Integration with DNA methylation and histone modification data
Functional genomics integration:
CRISPR-Cas9 modification of ELF3 binding sites
Correlation of ELF3 binding (ChIP) with functional enhancer assays
ELF3 knockout paired with rescue experiments using domain mutants
Live-cell dynamics:
Combine fixed-cell antibody staining with live-cell ELF3-fluorescent protein fusions
FRAP (Fluorescence Recovery After Photobleaching) to assess dynamics, validated by antibody staining
Optogenetic ELF3 control systems with antibody-based outcome measurement
Protein interaction networks:
Proximity labeling (BioID, APEX) with ELF3 fusions, validated by co-immunoprecipitation
Protein complementation assays combined with antibody-based localization
Mammalian two-hybrid screens with ELF3 domains, confirmed by co-immunoprecipitation
Translational approaches:
Patient-derived xenografts with human-specific ELF3 antibody detection
Ex vivo tissue culture with pharmacological manipulation and ELF3 monitoring
Correlation of genomic alterations with antibody-detected ELF3 protein levels