The E2-Tag Monoclonal Antibody (Clone 5E11, Catalog ab977) is a mouse-derived IgG1 antibody that specifically recognizes the SSTSSDFRDR epitope sequence . This 10-amino acid tag serves as a molecular handle for detecting recombinant proteins in laboratory settings.
The E2-Tag antibody has been validated in multiple experimental paradigms:
Compared to FLAG, HA, or Myc tags, the E2 system offers:
Smaller size (10 vs. 8-25 amino acids in other tags), minimizing steric interference
Reduced immunogenicity in mammalian expression systems
High specificity due to unique SSTSSDFRDR sequence absent in natural proteomes
While highly effective, users should note:
The E2-Tag is a peptide sequence derived from the Bovine Papillomavirus type-1 transactivator protein E2. It consists of 10 amino acids (SSTSSDFRDR) and serves as an epitope tag for protein labeling and detection in research applications . Epitope tags like E2 are valuable molecular tools that allow researchers to track and analyze proteins of interest without interfering with their native function, particularly when specific antibodies against the protein of interest are unavailable.
The tag's relatively small size minimizes interference with protein folding or interactions while still providing a unique recognition site for highly specific antibodies. E2-Tag functions as a molecular handle that can be genetically engineered to various positions within a recombinant protein.
E2-Tag monoclonal antibodies specifically recognize the E2 epitope sequence when it is fused to a protein of interest. The detection mechanism relies on the binding of mouse monoclonal antibodies (such as clone 4F8 or B2-E2) that have high specificity for the E2-Tag sequence . These antibodies can recognize the E2-Tag regardless of its position within the recombinant protein (C-terminal, internal, or N-terminal), providing versatility in experimental design .
The antibodies function through standard immunological principles:
Primary binding occurs between the anti-E2-Tag monoclonal antibody and the epitope tag on the fusion protein
Detection systems (enzyme-conjugated secondary antibodies, fluorophores, etc.) then recognize the primary antibody
Signal generation allows visualization or quantification of the tagged protein
This system enables researchers to detect, isolate, or visualize the tagged protein in various experimental contexts without needing antibodies against the protein itself.
Different E2-Tag monoclonal antibody clones exhibit varying characteristics that can significantly impact experimental outcomes:
Clone | Isotype | Advantages | Optimal Applications |
---|---|---|---|
4F8 | IgG | High specificity, works in multiple applications | WB, ELISA, IP |
B2-E2 | IgG | Good signal-to-noise ratio in cellular applications | IF-Cell, WB, IP, ELISA |
When selecting an E2-Tag antibody clone, researchers should consider:
Binding affinity - higher affinity clones provide better sensitivity
Background signal - some clones may produce lower non-specific binding
Application compatibility - certain clones perform better in specific techniques
Recognition efficiency - quantitative studies show some antibodies generate high signals even at low concentrations (50 ng/mL), placing them in the "good" antibody category
The selection of an appropriate clone should be guided by the specific experimental requirements and validated for each application.
For optimal Western blot results with E2-Tag monoclonal antibodies, follow this methodological approach:
Sample Preparation:
Lyse cells in an appropriate buffer (RIPA buffer for most applications) containing protease inhibitors
Quantify protein concentration (Bradford or BCA assay)
Prepare samples in Laemmli buffer with reducing agent (β-mercaptoethanol)
Heat samples at 95°C for 5 minutes
Gel Electrophoresis and Transfer:
Load 10-30 μg total protein per lane (adjust based on expression level)
Separate proteins using SDS-PAGE (select gel percentage based on target protein size)
Transfer to PVDF or nitrocellulose membrane (PVDF recommended for higher protein retention)
Immunodetection:
Block membrane with 5% non-fat milk or 3-5% BSA in TBS-T (1 hour, room temperature)
Incubate with anti-E2-Tag antibody at 1:5000 dilution in blocking buffer (overnight, 4°C)
Wash 3-5 times with TBS-T (5-10 minutes each)
Incubate with HRP-conjugated secondary antibody (1:5000-1:10000, 1 hour, room temperature)
Wash thoroughly and develop using enhanced chemiluminescence (ECL) substrate
Controls:
Include positive control (known E2-tagged protein)
Include negative control (non-tagged version of protein)
Use loading control (β-actin, GAPDH) to normalize protein amounts
This protocol typically yields specific detection of E2-tagged proteins with minimal background signal.
E2-Tag monoclonal antibodies can be effectively employed in immunofluorescence microscopy using the following methodological approach:
Cell Preparation:
Grow cells on glass coverslips or appropriate imaging chambers
Transfect or transduce cells with constructs expressing E2-tagged proteins
Fix cells using 4% paraformaldehyde (10 minutes, room temperature) for most applications
Alternative fixation methods (methanol, acetone) may be tested if PFA affects epitope accessibility
Immunostaining:
Permeabilize cells with 0.1-0.3% Triton X-100 in PBS (5-10 minutes)
Block with 3-5% BSA or 5-10% normal serum in PBS (30-60 minutes)
Incubate with anti-E2-Tag primary antibody at 1:100-1:1000 dilution (overnight, 4°C)
Wash 3-5 times with PBS containing 0.05% Tween-20
Incubate with fluorophore-conjugated secondary antibody (1:500-1:1000, 1 hour, room temperature)
Wash thoroughly and counterstain nuclei with DAPI if desired
Mount slides with anti-fade mounting medium
Optimization Strategies:
Titrate antibody concentration to determine optimal signal-to-noise ratio
Test different blocking agents to minimize background
Include detergents in antibody diluents to reduce non-specific binding
Compare multiple fixation protocols to maximize epitope accessibility
Consider signal amplification systems for low-abundance targets
Comparative studies have shown that high-quality E2-Tag antibodies can generate significant signals even at lower concentrations (50 ng/mL), placing them among the more efficient epitope tag detection systems .
To enhance the sensitivity of E2-Tag detection in ELISA applications, implement these methodological improvements:
Antibody Selection and Preparation:
Use high-affinity monoclonal antibodies specific to E2-Tag
Purify antibodies to remove contaminants that may interfere with binding
Determine optimal working concentration through titration experiments
Signal Amplification Techniques:
Implement streptavidin-biotin detection systems (4-8× signal enhancement)
Consider polymer-based detection systems with multiple enzyme molecules per antibody
Utilize chemiluminescent or fluorescent substrates instead of colorimetric detection
Extend substrate development time for increased sensitivity
Assay Format Optimization:
Test different plate coating buffers (carbonate buffer pH 9.6 vs. PBS pH 7.4)
Evaluate various blocking agents (BSA, casein, commercial blockers) for lowest background
Optimize incubation times and temperatures:
Extended primary antibody incubation (overnight at 4°C)
Secondary antibody incubation (2 hours at room temperature)
Implement sandwich ELISA format with capture and detection antibodies for maximum sensitivity
Sample Processing:
Pre-clear samples to remove interfering substances
Concentrate samples when target protein is in low abundance
Optimize sample diluent composition to enhance specific binding
These optimizations can significantly improve detection limits, potentially reaching sensitivity in the low pg/mL range. Researchers should systematically test these parameters in their specific experimental context to determine the optimal conditions.
E2-Tag monoclonal antibodies offer valuable tools for epitope mapping through several methodological approaches:
Fusion Protein Strategy:
Generate a series of fusion constructs with E2-Tag positioned at various locations relative to the target epitope
Express and purify these fusion proteins
Perform binding assays (ELISA, Western blot) to assess accessibility of the E2-Tag
Map regions where tag insertion disrupts antibody binding to identify critical epitope residues
Peptide Array Analysis:
Create overlapping peptide arrays spanning the protein of interest
Include E2-Tag as a control epitope with known antibody reactivity
Compare binding patterns between anti-E2-Tag antibodies and antibodies against the target protein
Identify peptides with similar binding characteristics to map epitopes
Domain Swapping:
Engineer chimeric proteins containing domains from different proteins with E2-Tags
Assess antibody binding to identify which domains contain the epitope of interest
Further refine mapping by creating subdomain constructs with E2-Tags
This approach has been successfully applied in studies with viral proteins, where researchers identified linear B-cell epitopes using monoclonal antibodies . For example, with CSFV E2 protein, researchers identified specific epitopes (25GLTTTWKEYSHDLQL39 and 259GNTTVKVHASDERGP273) using similar methodologies . The same principles can be applied using E2-Tag as a control or reference point for mapping epitopes in other proteins.
Proper storage of E2-Tag monoclonal antibodies is critical for maintaining their activity and specificity over time. Based on manufacturer recommendations and research practices:
Long-term Storage:
Most commercial preparations contain 50% glycerol and 0.02% sodium azide as preservatives
Divide antibody into small aliquots before freezing to avoid repeated freeze-thaw cycles
Label aliquots with date, concentration, and number of freeze-thaw cycles
Working Solution Storage:
Diluted antibody solutions can be stored at 4°C for 1-2 weeks
Add preservatives (0.02% sodium azide) to prevent microbial growth
For prolonged storage of working dilutions, return to -20°C
Stability Factors:
Avoid repeated freeze-thaw cycles, which can cause protein denaturation and aggregation
Protect from extended exposure to room temperature
Shield from direct light, especially if conjugated to fluorophores
Maintain sterile conditions to prevent contamination
Monitoring Antibody Activity:
Include positive controls when using antibodies that have been stored for extended periods
Record antibody performance over time to detect any degradation
Consider refreshing antibody stocks if signal quality diminishes
Following these storage guidelines will help maintain antibody activity and ensure consistent experimental results across multiple studies.
Non-specific binding is a common challenge when using E2-Tag antibodies that can compromise experimental results. Implement these methodological approaches to minimize background:
Blocking Optimization:
Test different blocking agents:
3-5% BSA in PBS or TBS
5% non-fat dry milk (not suitable for phospho-specific applications)
Commercial blocking solutions
Extend blocking time to 1-2 hours at room temperature or overnight at 4°C
Include blocking agent in antibody diluent solutions
Antibody Preparation:
Pre-adsorb antibodies against tissues or cells lacking the E2-Tag
Centrifuge antibody solutions (10,000 × g, 5 minutes) before use to remove aggregates
Titrate antibody concentration to determine the minimal effective concentration
Use high-quality, affinity-purified antibodies
Washing Procedures:
Increase number of washes (5-6 washes instead of standard 3)
Extend washing time (10-15 minutes per wash)
Add detergents to wash buffers (0.05-0.1% Tween-20 or 0.1% Triton X-100)
Consider higher salt concentration in wash buffers (up to 500 mM NaCl) to disrupt non-specific ionic interactions
Additional Techniques:
Add 0.1-0.3% Triton X-100 to antibody diluents to reduce hydrophobic interactions
Include 5-10% normal serum from the species of the secondary antibody
For Western blots, consider using PVDF membranes instead of nitrocellulose
For immunofluorescence, include an additional permeabilization step before blocking
These optimizations should be systematically tested and adapted to specific experimental conditions, as the sources of non-specific binding can vary between applications and sample types.
Researchers frequently encounter several challenges when working with E2-Tag antibodies. Here are common pitfalls and their methodological solutions:
Low Signal Intensity:
Problem: Weak or undetectable signal despite proper expression
Solutions:
Increase antibody concentration or incubation time
Try alternative antibody clones with higher affinity
Implement signal amplification systems
Verify tag accessibility in protein structure
Check if fixation methods have altered epitope conformation
High Background:
2. Problem: Excessive non-specific binding obscuring specific signal
Solutions:
Optimize blocking conditions (agent, time, temperature)
Increase washing stringency (more washes, longer duration)
Reduce antibody concentration
Pre-adsorb antibody against non-specific binding sites
Use more specific secondary antibodies
Multiple Bands in Western Blot:
3. Problem: Detection of unexpected bands beyond target protein
Solutions:
Verify sample integrity (add protease inhibitors)
Optimize lysis conditions to prevent degradation
Increase gel percentage for better resolution
Perform peptide competition assays to identify specific bands
Validate with alternative detection methods
Tag Inaccessibility:
4. Problem: E2-Tag is masked by protein folding
Solutions:
Redesign construct with tag at different position (N-terminal, C-terminal)
Add flexible linker sequences between protein and tag
Use denaturing conditions for Western blot
Try alternative fixation methods for immunofluorescence
Consider mild denaturation steps before antibody incubation
Cross-Reactivity:
5. Problem: Antibody binds to endogenous proteins
Solutions:
Include non-transfected controls
Perform immunoprecipitation with tagged proteins
Use siRNA to knock down target protein and confirm specificity
Try alternative antibody clones with higher specificity
Validate with orthogonal methods
Systematic troubleshooting using these approaches can significantly improve the reliability and specificity of E2-Tag detection in various experimental applications.
E2-Tag monoclonal antibodies can be utilized in sophisticated single B-cell isolation protocols, particularly for developing new monoclonal antibodies against specific targets. The methodology involves:
Protein Preparation:
Express and purify E2-tagged target protein using appropriate expression systems
Biotinylate the E2-tagged protein using NHS-LC-biotin reagents
Validate biotinylation by Western blot analysis
Quantify the degree of biotinylation to ensure optimal labeling
Immunization Strategy:
Immunize mice with purified E2-tagged protein (typically 25 μg per dose)
Use appropriate adjuvants (e.g., Freund's) to enhance immune response
Administer 2-3 booster immunizations at 2-week intervals
Validate antibody response using ELISA or other serological assays
Single B-Cell Isolation:
Harvest splenocytes from immunized mice
Block Fc receptors with mouse Fc block reagent
Stain cells with:
Sort E2-specific plasmablasts (E2-APC+ and IgM-FITC−) using flow cytometry
Distribute single cells into 96-well plates containing lysis buffer
Antibody Gene Amplification:
Perform reverse transcription using random primers
Amplify immunoglobulin genes using nested PCR with specific primers
Sequence VH and VL regions to identify unique antibody clones
Clone into expression vectors for recombinant antibody production
This methodology has been successfully applied for generating monoclonal antibodies against viral proteins like CSFV E2 and can be adapted for various research targets using E2-Tag as a selection marker or reference point.
When utilizing E2-Tag for structural biology studies, researchers must address several methodological considerations:
Tag Positioning Effects:
N-terminal tagging may affect signal peptide processing or disrupt functional domains
C-terminal tagging generally offers less interference with protein folding
Internal tagging requires careful placement in flexible loop regions
Consider using removable tags with protease cleavage sites for post-purification tag removal
Structural Impact Assessment:
Compare activity of tagged vs. untagged proteins to evaluate functional consequences
Analyze thermal stability using differential scanning fluorimetry
Assess oligomerization state using size-exclusion chromatography
Verify correct folding using circular dichroism spectroscopy
Conduct limited proteolysis to identify potential structural perturbations
Crystallization Considerations:
E2-Tag (10 amino acids) may introduce flexibility that hinders crystallization
Include constructs with and without tags in crystallization trials
Consider tag removal before crystallization attempts
Use engineered crystallization chaperones as alternative to traditional tags
Test different linker lengths between protein and tag
Cryo-EM Applications:
E2-Tag can serve as a localization marker for subunit identification in complexes
Position tags away from important interaction surfaces
Tag visibility may be enhanced by fusion to larger domains for improved particle picking
Compare structures with tags in different positions to verify structural integrity
NMR Considerations:
By systematically addressing these considerations, researchers can effectively use E2-Tag in structural biology applications while minimizing potential artifacts or interference with native protein structure.
E2-Tag antibodies offer powerful tools for developing sensitive and specific diagnostic assays with numerous methodological advantages:
Assay Design Principles:
Blocking ELISA (bELISA) format:
Sandwich ELISA format:
Capture antibody binds target protein
E2-tagged detection antibody binds secondary epitope
Anti-E2-Tag antibody (enzyme-labeled) provides detection signal
Enhances specificity through dual-epitope recognition
Optimization Strategies:
Antigen design:
Engineer recombinant antigens with strategically placed E2-Tags
Express multiple antigens with identical E2-Tags for multiplexed detection
Validate epitope accessibility in assay conditions
Signal amplification:
Implement enzyme cascades for signal enhancement
Use nanoparticle-conjugated anti-E2-Tag antibodies
Explore electrochemical detection methods for quantitative results
Validation protocols:
Test against panels of known positive and negative samples
Calculate sensitivity, specificity, and predictive values
Perform cross-reactivity studies with related pathogens
Establish reproducibility through inter-laboratory testing
Practical Applications:
Infectious disease diagnostics:
Viral hepatitis serology
Arbovirus antibody detection
Parasitic disease diagnostics
Veterinary diagnostics:
Biomarker detection:
Cancer biomarkers
Autoimmune disease antibodies
Therapeutic drug monitoring
By standardizing the detection component of assays through E2-Tag technology, researchers can develop highly consistent diagnostic platforms with reduced development time and improved performance characteristics.
E2-Tag technology is expanding into sophisticated cellular imaging applications with several promising methodological developments:
Super-Resolution Microscopy:
Site-specific labeling with small E2-Tags offers advantages over larger fluorescent protein fusions
Combining E2-Tag with click chemistry approaches for orthogonal labeling strategies
Development of high-affinity, photoswitchable fluorophore-conjugated anti-E2-Tag antibodies for STORM/PALM microscopy
Implementation in multi-color super-resolution imaging for protein interaction studies
Live-Cell Imaging:
Engineering of cell-permeable anti-E2-Tag antibody fragments (Fabs, nanobodies)
Development of split-tag systems where E2-Tag complementation induces fluorescence
Temporal control of E2-Tag expression for pulse-chase experiments
Combination with optogenetic tools for spatiotemporal protein function analysis
Multiplexed Imaging:
Cyclic immunofluorescence using E2-Tag as one of multiple epitope tags
Mass cytometry (CyTOF) applications using metal-conjugated anti-E2-Tag antibodies
Spatial transcriptomics integration with E2-Tag protein localization
Correlated light and electron microscopy using E2-Tag for protein localization
Intrabody Applications:
Engineering anti-E2-Tag antibody fragments for expression inside living cells
Targeting specific subcellular compartments with E2-tagged proteins
Modulating protein function through intrabody binding to E2-Tag
Real-time monitoring of protein dynamics in living systems
These emerging applications represent the cutting edge of E2-Tag technology in cellular imaging, offering researchers powerful new tools for investigating protein function, localization, and dynamics at unprecedented resolution.
E2-Tag technology offers several methodological advantages in therapeutic protein development pipelines:
Manufacturing Process Development:
Consistent purification protocols across diverse protein therapeutics
High-affinity anti-E2-Tag antibody resins for efficient capture steps
Site-specific tag placement to minimize impact on protein function
Incorporation of protease cleavage sites for tag removal after purification
Real-time monitoring of expression levels using anti-E2-Tag detection systems
Analytical Characterization:
Standardized quantification assays across different therapeutic modalities
Structural integrity assessment using conformation-sensitive anti-E2-Tag antibodies
Batch-to-batch consistency evaluation with sensitive E2-Tag detection systems
Development of reference standards with defined E2-Tag content
Improved comparability studies between originator and biosimilar products
Functional Analysis:
Receptor binding assays using E2-tagged ligands
Cell-based potency assays with standardized detection systems
Biodistribution studies using anti-E2-Tag antibodies for tissue localization
Pharmacokinetic analysis with sensitive E2-Tag immunoassays
Target engagement studies in preclinical models
Safety Assessment:
Immunogenicity evaluation of E2-Tag in various contexts
Development of anti-drug antibody assays based on E2-Tag detection
Assessment of E2-Tag removal efficiency during manufacturing
Cross-reactivity screening against human tissues
The strategic implementation of E2-Tag technology in therapeutic protein development can streamline manufacturing processes, enhance analytical capabilities, and provide consistent methodologies across diverse protein modalities, ultimately accelerating the path to clinical applications.