Applications : Western Blot (WB)
Sample type: Cells
Sample dilution: 1:500
Review: Antibody specificity is very good.
The Myc tag is a short synthetic polypeptide sequence derived from the human c-myc protein, specifically corresponding to amino acids 410-419 with the sequence EQKLISEEDL. This tag is widely used in molecular biology research when an antibody against the protein of interest is not available. By fusing the Myc tag to a protein of interest through recombinant DNA technology, researchers can track, purify, and study the tagged protein using commercially available anti-Myc tag antibodies . This approach is particularly valuable for newly discovered proteins or when generating specific antibodies is challenging or time-consuming.
Myc tag monoclonal antibodies have diverse applications in molecular and cellular biology research:
Application | Description | Typical Dilution |
---|---|---|
Western Blot (WB) | Detection of Myc-tagged proteins after gel electrophoresis | 1:1000-1:8000 |
Immunoprecipitation (IP) | Isolation of Myc-tagged proteins from complex mixtures | 0.5-4.0 μg for 1-3 mg lysate |
Immunofluorescence (IF) | Visualization of Myc-tagged proteins in fixed cells | 1:200-1:800 |
Flow Cytometry | Quantification of Myc-tagged proteins in cell populations | 1:200 |
Immunohistochemistry (IHC) | Detection of Myc-tagged proteins in tissue sections | 1:200-1:400 |
ELISA | Quantitative measurement of Myc-tagged proteins | 1:1000 |
Chromatin Immunoprecipitation (ChIP) | Study of protein-DNA interactions | Variable |
These applications enable researchers to study protein expression, localization, interaction, and function in various experimental systems .
The Myc tag has a molecular weight of approximately 1203 Da, comprising the ten amino acid sequence EQKLISEEDL . When designing experiments, researchers should account for this additional mass when analyzing tagged proteins. While the small size of the Myc tag generally minimizes interference with protein structure and function, it's advisable to validate that the tag does not affect the biological activity of your protein of interest through appropriate controls. The tag can be fused to either the N-terminus or C-terminus of the target protein, depending on which arrangement is less likely to interfere with protein function .
A comprehensive analysis of six antibodies recognizing the c-Myc epitope tag revealed significant differences in context-dependent detection capabilities:
Antibody Clone | Type | Context Sensitivity | Optimal Applications | Notes |
---|---|---|---|---|
9E10 | Monoclonal | High | Traditionally used, but variable performance | Most cited in literature but shows high context-dependent variability |
4A6 | Monoclonal | Low | Robust detection in diverse contexts | Recommended for consistent detection across different tag positions |
9B11 | Monoclonal | Low | Robust detection in diverse contexts | Recommended for consistent detection across different tag positions |
71D10 | Monoclonal | Moderate | Multiple applications | Good general-purpose antibody |
9106 | Polyclonal | Variable | Multiple applications | Higher background but potentially better sensitivity |
A-14 | Polyclonal | Variable | Multiple applications | Higher background but potentially better sensitivity |
Research has demonstrated that recently developed, purpose-made monoclonal antibodies (particularly 4A6 and 9B11) have much more uniform reactivity in diverse assays and are much less context-sensitive than the legacy antibody 9E10 . This finding is important when selecting antibodies for experiments where the Myc tag may be positioned in different contexts.
Epitope tag position and neighboring sequences can significantly affect recognition by antibodies. Context-dependent differences in tag binding may have wide-ranging effects on data interpretation. Western blotting tests have shown that all Myc tag antibodies display some level of context-dependent differences in their ability to detect N- or C-terminal Myc-tagged proteins, though to varying degrees .
For optimal detection:
Consider testing multiple anti-Myc antibody clones if you observe weak or inconsistent signals
Be aware that clone 9E10, despite being widely cited, shows high context-dependent variability
For applications requiring consistent detection regardless of tag position, consider clones 4A6 or 9B11
When possible, standardize the position of the tag (N- or C-terminal) across all constructs in a study
This context sensitivity was confirmed through peptide microarray analyses, demonstrating that epitope accessibility plays a crucial role in antibody binding efficiency .
To maintain antibody activity and stability:
Store unopened antibodies at -20°C for up to one year
Make small aliquots to avoid repeated freeze-thaw cycles
Prior to opening, briefly spin tubes to ensure liquid collection at the bottom
For working solutions, store at 4°C for short-term use (2-4 weeks)
Protect conjugated antibodies (HRP, FITC) from light exposure
Follow manufacturer's recommendations for storage buffer composition (typically PBS, pH 7.4, with 0.05% sodium azide and sometimes glycerol or BSA)
Proper storage significantly impacts experimental reproducibility and antibody shelf-life. Degradation can lead to reduced sensitivity, increased background, and experiment failure.
Optimal dilution factors vary by application and specific antibody clone. While manufacturers provide recommended ranges, optimization for your specific experimental system is crucial:
Western Blot optimization:
Start with the recommended dilution (typically 1:1000)
Perform a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000)
Evaluate signal-to-noise ratio at each dilution
Select the dilution that provides clear specific bands with minimal background
Immunofluorescence optimization:
Begin with manufacturer's recommendation (typically 1:200-1:800)
Test on cells with known expression levels of Myc-tagged proteins
Assess specificity by comparing transfected vs. non-transfected cells
Minimize autofluorescence with appropriate controls and blocking reagents
Immunoprecipitation optimization:
Remember that exact working dilution needs to be determined by the end user for each specific experimental setup .
Despite its utility, Myc tagging can potentially introduce artifacts that require careful consideration:
Protein mislocalization:
Compare localization with alternative tags (FLAG, HA)
Use both N- and C-terminal tagged versions
Validate with endogenous protein localization when possible
Consider disruption of localization signals (NLS, NES, transmembrane domains)
Altered protein function:
Perform functional assays comparing tagged and untagged versions
If available, compare with endogenous protein activity
Consider using a cleavable linker between tag and protein of interest
Test both N- and C-terminal tags to identify optimal configuration
Expression level artifacts:
Use inducible expression systems to control expression levels
Compare multiple clones with varying expression levels
Include dose-response experiments in functional assays
Consider single-cell analyses to account for expression heterogeneity
Aggregation or stability issues:
Assess protein solubility and stability through biochemical fractionation
Examine protein half-life with cyclohexamide chase experiments
Consider native PAGE alongside denaturing conditions
Evaluate oligomerization state with size exclusion chromatography
The small size of the Myc tag (10 amino acids) generally minimizes interference with protein function, but validation is essential for each new protein under investigation.
When different antibody clones yield conflicting results, systematic troubleshooting is necessary:
Context-dependent epitope accessibility:
The high context sensitivity of some clones (particularly 9E10) may explain detection discrepancies
Try multiple antibody clones with different epitope recognition characteristics
Clone 4A6 or 9B11 may provide more consistent detection across contexts
Consider using a dual-tagging approach (e.g., Myc+FLAG) for validation
Methodological approach:
Compare native versus denaturing conditions
Assess fixation effects in microscopy applications
Optimize blocking conditions to reduce non-specific binding
Consider epitope retrieval methods for fixed samples
Systematic comparison:
Create a standardized experimental system with known positive controls
Test all antibody clones side-by-side under identical conditions
Quantify detection efficiency using recombinant standards
Document batch-to-batch variation from the same manufacturer
Independent validation:
Verify results with orthogonal methods (mass spectrometry, alternate tags)
Use CRISPR/Cas9 knock-in to tag endogenous proteins
Consider proximity labeling approaches (BioID, APEX) as complementary methods
Validate with functional assays that don't rely on antibody detection
Understanding the specific binding characteristics of each antibody clone can help reconcile apparently contradictory results and guide experimental design.
Simultaneous detection of multiple tagged proteins requires careful experimental design:
Compatible tag selection:
Choose tags with minimal cross-reactivity (Myc, FLAG, HA, V5)
Ensure antibodies are raised in different host species to enable secondary antibody discrimination
Consider size differences between tags for distinguishing proteins of similar molecular weight
Test for potential tag-tag interactions in your experimental system
Multi-color immunofluorescence optimization:
Select fluorophores with minimal spectral overlap
Perform single-color controls to establish detection parameters
Use sequential staining protocols for antibodies from the same species
Apply appropriate controls for background autofluorescence and bleed-through
Sequential immunoprecipitation strategies:
Perform tandem IP using different tags to verify protein-protein interactions
Elute specifically using competitor peptides rather than denaturing conditions
Consider using magnetic beads with different properties for separation
Validate recovery efficiency at each step with known standards
Multiplexed Western blotting:
Use differentially labeled secondary antibodies for simultaneous detection
Strip and reprobe membranes sequentially with documentation between rounds
Consider size differences to avoid signal overlap
Use dual-color detection systems with appropriate controls
These strategies enable complex experimental designs investigating interactions between multiple tagged proteins within the same experimental system.
Accurate interpretation of experimental results requires awareness of potential artifacts:
Implementing rigorous controls and validation steps significantly improves data reliability and reproducibility.
Thorough validation ensures experimental reliability:
Essential controls:
Non-transfected/non-transformed negative controls
Cells expressing known Myc-tagged proteins as positive controls
Competitive inhibition with excess Myc peptide (EQKLISEEDL)
Secondary antibody-only controls to assess non-specific binding
Cross-validation approaches:
Compare detection with multiple anti-Myc antibody clones
Validate with alternative detection methods (e.g., fluorescent proteins)
Use alternative tagging systems (FLAG, HA) on the same protein
Perform mass spectrometry analysis of immunoprecipitated material
Specificity testing:
Examine size-appropriate bands on Western blots
Perform siRNA/shRNA knockdown of tagged construct to confirm signal reduction
Use CRISPR/Cas9 to tag endogenous proteins for physiological expression level controls
Compare subcellular localization with known distribution patterns or GFP fusion proteins
Advanced validation:
Use proximity ligation assays to verify co-localization specificity
Employ super-resolution microscopy to assess spatial distribution
Perform epitope mapping to confirm precise antibody binding sites
Conduct functional rescue experiments to verify biological activity of tagged proteins
Thorough validation increases confidence in experimental outcomes and facilitates meaningful interpretation of results.
Recent technological advances are expanding the utility of Myc-tagged proteins:
Single-molecule detection:
Super-resolution microscopy enables visualization of individual Myc-tagged proteins
Single-molecule pull-down techniques allow analysis of protein complexes at endogenous levels
Nanobody-based detection improves spatial resolution and penetration
Improved signal amplification methods enable detection of low-abundance proteins
Live-cell applications:
Split-tag complementation systems for studying protein-protein interactions
FRET/BRET-based approaches for real-time interaction monitoring
Development of cell-permeable anti-Myc antibody fragments
Integration with optogenetic tools for spatiotemporal control
High-throughput methodologies:
Microfluidic antibody validation platforms for systematic comparison
Automated image analysis algorithms for quantitative assessment
Barcode-enabled multiplexing for simultaneous detection of multiple Myc-tagged proteins
Integration with single-cell technologies for heterogeneity analysis
Next-generation antibodies:
Recombinant antibody technologies improving batch-to-batch consistency
Engineering of high-affinity variants with reduced context sensitivity
Development of bispecific antibodies for multiplexed detection
Nanobody and single-domain antibody alternatives with improved tissue penetration
These advances are expanding the experimental possibilities for researchers using Myc-tagged proteins and improving data quality and reproducibility.
As research techniques evolve, integrating Myc tag detection with advanced methodologies requires careful consideration:
Integration with CRISPR/Cas9 genome editing:
Design appropriate homology-directed repair templates for endogenous tagging
Consider tag position effects on protein function and antibody accessibility
Validate edited clones with multiple detection methods
Account for potential heterogeneity in edited populations
Compatibility with advanced imaging techniques:
Optimize fixation and permeabilization protocols for super-resolution microscopy
Consider probe size limitations for techniques like STORM or PALM
Validate spatial resolution with known structural features
Implement appropriate controls for colocalization studies
Adaptation for proteomics applications:
Optimize elution conditions to maintain complex integrity
Consider on-bead digestion protocols to improve peptide recovery
Implement isotopic labeling strategies for quantitative comparisons
Validate results with alternative purification strategies
Single-cell analysis considerations:
Account for expression level heterogeneity in cell populations
Optimize fixation and permeabilization for intracellular epitope access
Implement appropriate compensation controls for multi-parameter analysis
Consider cell cycle effects on tagged protein expression and localization
Careful integration of Myc tag detection with these advanced methodologies enables sophisticated experimental designs while maintaining data reliability.