The FLAG PAT3G6AT antibody binds to the DYKDDDDK sequence, which is typically appended to recombinant proteins at the N-terminus, C-terminus, or internal sites . Unlike calcium-dependent antibodies like M1 (which bind only N-terminal FLAG tags), this antibody does not require divalent cations for binding . Its specificity allows detection of FLAG-tagged proteins in diverse contexts, including bacterial, mammalian, and plant expression systems .
Antibody Clone | Binding Sites | Calcium Dependency | Primary Applications |
---|---|---|---|
PAT3G6AT | N-terminal, C-terminal, internal FLAG tags | No | Western blot, immunoprecipitation |
M2 | N-terminal, C-terminal, internal tags | No | Western blot, IP, ELISA |
M1 | N-terminal FLAG tags only | Yes (Ca²⁺ required) | Affinity purification |
FG4R | N-terminal, C-terminal, internal tags | No | Western blot, immunohistochemistry |
Data highlights PAT3G6AT’s versatility compared to clones with restricted binding profiles (e.g., M1) .
Western Blotting: Detects FLAG-tagged proteins in denatured lysates. No specific titer is provided, but typical FLAG antibody dilutions range from 1:1,000 to 1:5,000 .
Immunoprecipitation (IP): Purifies FLAG-tagged proteins from complex mixtures. Protocols often involve immobilizing the antibody on Protein A/G beads or agarose resins .
Protein Localization: Used in immunocytochemistry or immunofluorescence to track FLAG-tagged proteins in subcellular compartments .
Monoclonal Specificity: Eliminates cross-reactivity with endogenous proteins, reducing background noise .
Consistent Performance: Standardized production ensures reproducibility across experiments .
Factor | Impact | Mitigation Strategy |
---|---|---|
Tag Position | C-terminal or internal tags may reduce accessibility for antibody binding | Optimize tag placement to N-terminus or exposed regions |
Protein Expression | Poorly expressed FLAG-tagged proteins may limit detection sensitivity | Validate expression via parallel Coomassie staining |
Cross-Reactivity | Potential binding to non-target proteins (e.g., endogenous KLH-related epitopes) | Pre-block with non-specific IgG; use negative controls |
While the antibody exhibits high specificity, users should validate its performance in their experimental system. For example, in bacterial lysates, FLAG-tagged proteins may be prone to proteolytic degradation, necessitating protease inhibitor cocktails .
The FLAG tagging system remains a cornerstone of protein research, with antibodies like PAT3G6AT enabling precise detection and purification. Emerging trends include:
Multiplexing: Combining FLAG with other epitope tags (e.g., HA, His) for simultaneous detection .
High-Throughput Screening: Using FLAG-coated plates for ELISA-based binding assays .
Structural Studies: Leveraging FLAG tags for protein crystallization or cryo-EM workflows .
While specific studies using the PAT3G6AT antibody are not cited in the literature, its design aligns with established protocols for FLAG-based immunoassays . Researchers should prioritize validation in their specific experimental context to optimize performance.
The FLAG tag is an eight amino acid peptide sequence (AspTyrLysAspAspAspAspLys) that includes an enterokinase-cleavage site. It is specifically designed for immunoaffinity chromatography, enabling elution under non-denaturing conditions. Several antibodies have been developed against the FLAG peptide, including M1, which requires the presence of bivalent metal cations, preferably Ca(+), for binding. Chelating agents are used for elution with the M1 antibody. Another strategy employs competitive elution using an excess of free FLAG Peptide. The FLAG peptide is a versatile tool for purifying and detecting recombinant fusion proteins, proving useful in various applications like Western blotting, immunocytochemistry, immunoprecipitation, flow cytometry, and protein purification. Moreover, it aids in studying protein-protein interactions, cell ultrastructure, and protein localization. As a hydrophilic tag, the FLAG peptide significantly enhances the detection and purification of recombinant fusion proteins.
The solution is at a concentration of 1mg/ml and contains PBS at a pH of 7.4, 10% Glycerol, and 0.02% Sodium Azide.
FLAG antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT3G6AT.
Anti-human FLAG mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a Synthetic peptide (DYKDDDDKC)-KLH.
Mouse IgG1 heavy chain and k light chain.
The FLAG tag is an artificial fusion tag consisting of eight amino acids (AspTyrLysAspAspAspAspLys, or DYKDDDDK) that includes an enterokinase-cleavage site. It was specifically designed for immunoaffinity chromatography, enabling protein elution under non-denaturing conditions . The PAT3G6AT antibody is a mouse monoclonal antibody developed through hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice that were immunized with a synthetic DYKDDDDKC-KLH peptide . This antibody belongs to the IgG class with a heavy chain and kappa light chain, and is purified from mouse ascitic fluids by protein-A affinity chromatography . PAT3G6AT specifically recognizes the FLAG epitope, making it valuable for detecting, localizing, and purifying FLAG-tagged recombinant proteins.
The FLAG peptide system has multiple applications in contemporary molecular biology research, including:
Western blotting for protein detection and quantification
Immunocytochemistry for cellular localization studies
Immunoprecipitation for protein-protein interaction analysis
Flow cytometry for cell surface protein detection
Protein purification via affinity chromatography
Investigation of protein-protein interactions
Analysis of cell ultrastructure
The hydrophilic nature of the FLAG tag significantly enhances detection and purification efficiency of recombinant fusion proteins without substantially altering their structure or function .
FLAG-based detection differs fundamentally from direct protein detection in that it targets a standardized epitope tag rather than variable protein-specific epitopes. This approach offers several methodological advantages:
Standardization: The FLAG system uses a universal tag recognized by well-characterized antibodies, enabling consistent detection protocols across different target proteins .
Circumvention of antibody limitations: When quality antibodies against specific proteins are unavailable, the FLAG system provides an alternative detection method by recognizing the tag rather than the protein itself .
Expression control: In knock-in mice with FLAG-tagged proteins, expression remains under control of the gene's endogenous regulatory elements, preserving normal expression patterns and physiological relevance—unlike transgenic approaches that may lead to overexpression artifacts .
Improved signal-to-noise ratio: Some FLAG antibodies demonstrate exceptional sensitivity with minimal background staining, particularly valuable when detecting proteins expressed at low levels .
Different FLAG antibody clones have distinct properties that affect their performance in various experimental contexts:
Calcium dependency: The M1 antibody clone binds FLAG peptide only in the presence of bivalent metal cations (preferably Ca²⁺), which enables specific elution strategies using chelating agents .
Detection sensitivity: Some clones, such as the 2H8 antibody, demonstrate significantly higher affinity, enabling detection of proteins expressed at low levels that might be missed by other FLAG antibodies. For example, 2H8 antibody has been shown to stain FLAG-tagged G-protein-coupled receptors (GPCRs) more strongly than commercially available antibodies in both flow cytometry and immunostaining experiments .
Background signal: High-affinity clones like 2H8 can produce cleaner results with minimal non-specific background staining, which is particularly important for immunostaining applications in vitro and in vivo .
Immunoprecipitation efficiency: Some high-sensitivity antibodies require substantially less material (as little as 10 ng) to effectively immunoprecipitate FLAG-tagged proteins from cell lysates .
Host species compatibility: For mouse tissue immunohistochemistry, antibodies from goat (like anti-DDDDK polyclonal) can reduce background associated with using mouse primary antibodies on mouse tissues .
For optimal immunohistochemical detection of FLAG-tagged proteins in tissue samples, researchers should consider the following validated methodology:
Tissue preparation:
Antigen retrieval technique:
Blocking strategy:
Antibody selection and concentration:
Detection system:
Counterstaining and mounting:
Controls:
Two primary strategies have been established for eluting FLAG-tagged proteins while maintaining protein structure and function:
Calcium-dependent elution:
Competitive elution:
Uses excess free FLAG peptide to compete for antibody binding sites
Tagged proteins are displaced from antibody without harsh conditions
Advantage: Versatile approach that works with most FLAG antibodies
Consideration: Requires synthesis or purchase of free FLAG peptide
Typical concentration: 100-200 μg/ml of FLAG peptide in elution buffer
Both strategies preserve protein integrity and function better than traditional harsh elution methods such as extreme pH or denaturing agents, making them particularly valuable for functional studies of purified proteins.
The sensitivity of FLAG detection varies significantly between western blotting and immunohistochemistry, with important technical considerations for each:
Western blotting:
Typically demonstrates higher sensitivity for detecting low abundance proteins
FLAG-HRP conjugated antibodies can achieve detection at dilutions of 1:4000, allowing efficient protein detection with minimal antibody consumption
In comparative studies, detection with anti-FLAG antibodies often provides superior sensitivity compared to antibodies against the native protein, as demonstrated in studies with RIPK3.3×FLAG
Denatured proteins may expose the FLAG epitope more consistently
Immunohistochemistry:
Sensitivity varies significantly based on tissue fixation, processing, and antigen retrieval
Proteins expressed at very low levels may be undetectable by IHC but detectable by western blotting
Spatial resolution allows detection in specific cell types that might be diluted in whole tissue lysates
Studies with RIPK3.3×FLAG demonstrated that certain tissues (bone marrow) showed few positive cells by IHC but fell below detection limits in western blotting of whole tissue lysates
Certain high-affinity antibodies like 2H8 have demonstrated sufficient sensitivity to detect FLAG-tagged GPCRs expressed in vivo in the small intestine of mice under control of tissue-specific promoters
When working with FLAG-tagged proteins, researchers commonly encounter the following challenges with corresponding solutions:
High background staining:
Weak signal detection:
Solution: Consider high-affinity antibody clones like 2H8 that demonstrate superior sensitivity for detecting proteins expressed at low levels
Optimize antigen retrieval methods for immunohistochemistry applications
For western blotting, using FLAG-HRP conjugated antibodies can improve sensitivity with optimal dilutions (e.g., 1:4000)
Inconsistent detection across tissue types:
Difficulties in FLAG-tagged GPCR detection:
Validation of FLAG antibody specificity is critical for experimental rigor. Multiple complementary approaches can be employed:
Parallel detection methods:
Negative controls:
Comparative analysis:
Molecular weight verification:
In western blotting, the FLAG-tagged protein should show the expected molecular weight shift compared to the untagged version
FLAG tag adds approximately 1 kDa to the protein molecular weight
When designing FLAG-tagged knock-in mice for protein expression studies, researchers should consider these critical factors:
Tag insertion strategy:
C-terminal tagging (inserting FLAG sequence into the last coding exon ahead of the translation termination codon) preserves most protein functions while allowing detection
N-terminal tagging may interfere with signal peptides or cellular localization
Internal tagging requires careful domain analysis to avoid disrupting functional regions
Tag configuration:
Expression control:
Homozygosity considerations:
Detection strategy:
The PAT3G6AT FLAG antibody demonstrates distinct performance characteristics compared to other commonly used FLAG antibodies:
This comparison demonstrates that researchers should select FLAG antibodies based on specific application requirements, with PAT3G6AT offering reliable performance for standard western blotting and immunoprecipitation applications.
The FLAG tag system offers distinct advantages and limitations compared to other common epitope tagging systems:
Small size (8 amino acids): Minimally disrupts protein structure and function compared to larger tags
Hydrophilic nature: Improves accessibility for antibody recognition and reduces interference with protein folding
Flexible elution options: Enables both competitive elution with FLAG peptide and calcium-dependent elution strategies
High sensitivity detection: Some FLAG antibodies (e.g., 2H8) demonstrate exceptional sensitivity for detecting proteins expressed at low levels
Validated in multiple systems: Extensively characterized in cell culture, tissues, and in vivo models including knock-in mice
Potential proteolytic cleavage: Contains an enterokinase-cleavage site that could lead to tag removal in certain contexts
Multiple antibody clones: Different clones have varying properties, requiring careful selection for specific applications
Calcium-dependency considerations: Some antibodies (M1) require calcium for binding, limiting their use in calcium-free buffers
Background concerns in mouse tissues: Mouse-derived antibodies may produce higher background when used on mouse tissues, requiring alternative host antibodies
Several emerging technologies promise to expand the research applications of FLAG-tagged proteins:
Super-resolution microscopy:
Enhanced spatial resolution will allow more precise localization of FLAG-tagged proteins within subcellular compartments
Multi-color imaging with orthogonal epitope tags can reveal protein complex formation at nanoscale resolution
Proximity labeling approaches:
Combining FLAG tags with proximity labeling enzymes (BioID, APEX) could enable mapping of protein interaction networks in living cells
FLAG-based purification complements proximity labeling for proteomic analysis
CRISPR-Cas9 genome editing:
Streamlined generation of FLAG knock-in models in diverse organisms beyond mice
Multiplex tagging of protein families to study related proteins simultaneously
Single-cell proteomics:
FLAG-based detection could help quantify tagged proteins in individual cells to understand cellular heterogeneity
Combining with single-cell transcriptomics could reveal post-transcriptional regulation mechanisms
Antibody engineering:
Development of even higher affinity FLAG antibodies for detection of extremely low abundance proteins
Novel recombinant antibody formats optimized for specific applications (nanobodies, single-chain antibodies)
These technological advances will likely expand the utility of the FLAG tag system beyond its current applications, particularly for in vivo studies and low-abundance protein detection.
The FLAG peptide is a widely used fusion tag in molecular biology and biochemistry. It consists of eight amino acids (Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys) and includes an enterokinase-cleavage site. This peptide is specifically designed for immunoaffinity chromatography, allowing elution under non-denaturing conditions .
The FLAG peptide and its corresponding antibodies, such as Clone PAT3G6AT, are highly versatile and are used in various applications, including:
The FLAG antibody is formulated at a concentration of 1 mg/ml in PBS (pH 7.4) containing 10% glycerol and 0.02% sodium azide. For optimal stability, it should be stored at -20°C for long-term storage and at 4°C for short-term storage (up to 1 month). It is important to prevent freeze-thaw cycles to maintain the antibody’s integrity .