ECFP-Tag Monoclonal Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Phosphate Buffered Saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Typically, we are able to ship orders within 1-3 business days of receipt. Delivery times may vary depending on the mode of purchase and destination. For specific delivery timeframes, please consult your local distributors.
Target Names
ECFP-Tag

Q&A

What is an ECFP-Tag and why are monoclonal antibodies against it valuable?

Enhanced Cyan Fluorescent Protein (ECFP) is a genetic mutant of green fluorescent protein (GFP) originally derived from the jellyfish Aequorea victoria . ECFP functions as an epitope tag that can be genetically fused to proteins of interest, allowing for their visualization and detection in various experimental systems. Monoclonal antibodies specifically targeting the ECFP-Tag are valuable because they provide consistent specificity and reproducibility in protein detection experiments. Unlike the direct fluorescence of ECFP itself, anti-ECFP monoclonal antibodies allow for signal amplification through secondary detection systems, enabling visualization of low-abundance tagged proteins that might not generate sufficient direct fluorescence signal. These antibodies maintain consistent binding properties across experimental batches, providing reliable detection of ECFP-tagged proteins through applications such as Western blotting, immunoprecipitation, and immunofluorescence microscopy.

How do ECFP-Tag monoclonal antibodies compare with other epitope tag systems?

ECFP-Tag monoclonal antibodies represent one of several epitope tagging systems available to researchers. The most commonly used and well-characterized alternative epitope tag systems include M2/FLAG-tag (DYKDDDDK), 9E10/c-Myc-tag (EQKLISEEDL), and 12CA5/HA-tag (YPYDVPDYA) . Each system offers distinct advantages depending on the experimental context.

Tag SystemEpitope SequenceSizeKey AdvantagesCommon Applications
ECFP-TagFull ECFP protein~27 kDaDual functionality (fluorescence + antibody detection), versatile for live imagingLive cell imaging, FRET, protein localization studies
FLAG-TagDYKDDDDK8 aaSmall size, minimal interference with protein functionProtein purification, Western blot
c-Myc-TagEQKLISEEDL10 aaWell-characterized, high specificityImmunoprecipitation, ChIP assays
HA-TagYPYDVPDYA9 aaSmall size, high sensitivityImmunofluorescence, protein detection

ECFP-Tag's unique advantage lies in its dual functionality—it serves both as a fluorescent reporter and as an antigenic epitope for antibody detection. This dual-mode detection capability enables flexible experimental design, allowing researchers to either directly visualize the tagged protein through ECFP fluorescence or employ antibody-based detection methods for enhanced sensitivity.

What are the optimal storage and handling conditions for ECFP-Tag monoclonal antibodies?

For maximum longevity and consistent performance, ECFP-Tag monoclonal antibodies should be stored at -20°C where they typically remain stable for up to one year . Most commercial preparations are supplied in PBS (pH 7.4) containing 0.02% sodium azide as a preservative and 50% glycerol to prevent freeze-thaw damage . When working with these antibodies, researchers should:

  • Aliquot stock antibody solutions upon first thawing to minimize freeze-thaw cycles

  • Maintain cold chain during handling (use ice)

  • Avoid contamination by using sterile techniques when accessing the antibody

  • Return antibodies to -20°C promptly after use

  • Maintain records of freeze-thaw cycles for each aliquot

For diluted working solutions, store at 4°C and use within 1-2 weeks. Extended storage of diluted antibodies may lead to decreased activity or increased background signal in applications like Western blotting.

How can researchers optimize Western blot protocols using ECFP-Tag monoclonal antibodies?

Western blot optimization with ECFP-Tag monoclonal antibodies requires careful attention to several parameters to achieve maximum sensitivity and specificity. Based on manufacturer specifications, these antibodies typically perform optimally at dilutions between 1:2000-1:5000 for Western blot applications .

A comprehensive optimization strategy includes:

  • Sample preparation: Include proper controls (positive control with known ECFP-tagged protein, negative control without ECFP tag) alongside experimental samples. Use freshly prepared protein lysates when possible, with protease inhibitors to prevent degradation.

  • Transfer optimization: ECFP-tagged proteins (~27 kDa plus fusion partner size) may require adjusted transfer conditions. Use PVDF membranes for better protein retention and signal-to-noise ratio.

  • Blocking optimization: Test different blocking agents (5% non-fat milk, 3-5% BSA) to determine which provides the best signal-to-noise ratio with your specific ECFP-Tag monoclonal antibody.

  • Antibody concentration titration: Perform a dilution series (1:1000, 1:2000, 1:5000, 1:10000) to identify the optimal antibody concentration that provides maximum specific signal with minimal background.

  • Incubation conditions: Test both room temperature (1-2 hours) and 4°C overnight incubations to determine optimal primary antibody binding conditions.

  • Detection system selection: Compare chemiluminescence, fluorescence, and colorimetric detection methods to identify the most suitable approach for your specific experimental needs and equipment.

In troubleshooting weak signals, consider longer exposure times, higher antibody concentrations, different secondary antibodies, or signal enhancement systems. For high background issues, more stringent washing protocols, higher dilutions of primary antibody, or different blocking reagents may improve results.

What considerations are important when using ECFP-Tag monoclonal antibodies for immunoprecipitation studies?

When using ECFP-Tag monoclonal antibodies for immunoprecipitation (IP) of tagged proteins, researchers should consider several critical factors:

  • Antibody binding capacity: Determine the binding capacity of the selected ECFP-Tag monoclonal antibody to ensure sufficient antibody is used to capture the target protein.

  • Buffer optimization: The IP buffer composition significantly impacts success:

    • Use buffers containing 150-300 mM NaCl, 20-50 mM Tris-HCl (pH 7.4-8.0)

    • Include 0.1-1% non-ionic detergents (NP-40, Triton X-100) to reduce non-specific binding

    • Add protease inhibitors to prevent target degradation during the procedure

  • Pre-clearing: Pre-clear lysates with protein A/G beads without antibody to reduce non-specific binding.

  • Antibody immobilization: For optimal results, pre-couple the ECFP-Tag monoclonal antibody to protein A/G beads or use commercially available conjugated beads.

  • Elution strategies: Consider gentle elution methods (competitive elution with ECFP peptide) versus denaturing conditions (boiling in SDS sample buffer), depending on downstream applications.

  • Controls: Always include:

    • Negative control IP (non-specific IgG of same species as ECFP-Tag antibody)

    • Lysate from cells not expressing ECFP-tagged proteins

    • Input sample (pre-IP lysate) to assess IP efficiency

The reproducibility and specificity of monoclonal antibodies make them excellent choices for IP experiments compared to polyclonal alternatives, offering consistent performance across experiments and reduced batch-to-batch variation.

How can ECFP-Tag monoclonal antibodies be integrated into advanced microscopy techniques?

ECFP-Tag monoclonal antibodies can significantly enhance the capabilities of advanced microscopy techniques beyond direct visualization of ECFP fluorescence. These applications include:

  • Super-resolution microscopy: ECFP-Tag monoclonal antibodies conjugated with bright and photostable fluorophores can overcome the relatively lower brightness of direct ECFP fluorescence, enabling techniques such as STORM, PALM, or STED microscopy for nanoscale visualization of protein localization and dynamics.

  • Expansion microscopy: Immunolabeling with ECFP-Tag monoclonal antibodies allows for detection of ECFP-tagged proteins in expanded samples, combining the specificity of antibody detection with physical sample expansion for improved resolution.

  • Correlative light and electron microscopy (CLEM): ECFP-Tag monoclonal antibodies conjugated to gold particles or peroxidase enable correlation between fluorescence microscopy and electron microscopy, bridging the resolution gap between these techniques.

  • Multi-color imaging: By using antibody-based detection of ECFP-tagged proteins, researchers can free up fluorescence channels for other targets, enabling more complex experimental designs. This strategy works particularly well in systems where:

    • The direct ECFP fluorescence signal is too weak

    • Spectral overlap with other fluorophores presents challenges

    • Post-fixation detection is preferred

  • Multiplexed imaging: Through sequential staining protocols, ECFP-Tag monoclonal antibodies can be incorporated into cyclic immunofluorescence or multiplexed ion beam imaging (MIBI) workflows for highly multiplexed protein detection.

When implementing these advanced microscopy applications, researchers should optimize fixation protocols, antibody concentrations, and detection systems specifically for their imaging platform of choice.

What are the key considerations for validating ECFP-Tag monoclonal antibody specificity?

Thorough validation of ECFP-Tag monoclonal antibody specificity is essential for generating reliable research data. A comprehensive validation strategy should include:

  • Positive and negative controls: Test the antibody on samples with known ECFP-tagged proteins and completely negative samples to confirm specific recognition.

  • Competitive inhibition: Pre-incubate the antibody with purified ECFP protein before immunostaining or Western blotting to demonstrate binding specificity.

  • Cross-reactivity assessment: Test the antibody against cells expressing other fluorescent proteins (GFP, YFP, RFP) to ensure specificity for ECFP.

  • Knockout/knockdown verification: Compare antibody staining in wild-type samples versus those where the ECFP-tagged protein has been depleted or eliminated.

  • Molecular weight confirmation: Verify that the detected band in Western blots matches the expected molecular weight of your ECFP-tagged protein (ECFP ~27 kDa plus the target protein size).

  • Cross-application validation: Compare results across multiple detection methods (Western blot, immunofluorescence, flow cytometry) to ensure consistent specificity.

  • Batch-to-batch consistency: When obtaining new lots of antibody, perform side-by-side validation with previously validated lots.

Monoclonal antibodies generally offer superior specificity compared to polyclonal alternatives due to their recognition of a single epitope, making them particularly valuable for detecting specific tagged proteins in complex biological samples.

How should researchers troubleshoot non-specific binding issues with ECFP-Tag monoclonal antibodies?

Non-specific binding can significantly compromise experimental results when using ECFP-Tag monoclonal antibodies. A systematic troubleshooting approach includes:

  • Optimize blocking conditions:

    • Test different blocking agents (BSA, non-fat dry milk, normal serum, commercial blockers)

    • Increase blocking duration (1-3 hours at room temperature or overnight at 4°C)

    • Add 0.1-0.3% Tween-20 to blocking buffer to reduce hydrophobic interactions

  • Adjust antibody concentration:

    • Perform a dilution series to identify the optimal concentration

    • For Western blot applications, titrate from 1:1000 to 1:10000

    • For immunofluorescence, test dilutions from 1:100 to 1:1000

  • Modify washing protocols:

    • Increase the number of wash steps (5-6 washes instead of 3)

    • Extend wash durations (10-15 minutes per wash)

    • Use buffers with higher salt concentration (up to 500 mM NaCl) to disrupt weak non-specific interactions

  • Pre-adsorb the antibody:

    • Incubate diluted antibody with cell/tissue lysates lacking ECFP-tagged proteins

    • Remove antibodies that bind non-specifically before using in the actual experiment

  • Consider fixation impact:

    • Different fixation methods (paraformaldehyde, methanol, acetone) can affect epitope accessibility

    • Test multiple fixation protocols to determine optimal conditions

  • Evaluate secondary antibody specificity:

    • Test secondary antibody alone (without primary) to identify potential direct non-specific binding

    • Consider using secondary antibodies pre-adsorbed against potential cross-reactive species

  • Implement additional controls:

    • Include isotype control antibodies to distinguish between specific binding and Fc receptor interactions

    • Use peptide competition to confirm specificity

Systematic implementation of these strategies can significantly improve signal-to-noise ratio and experimental reliability.

What methodological approaches enable multiplexed detection using ECFP-Tag monoclonal antibodies?

Multiplexed detection involving ECFP-Tag monoclonal antibodies enables researchers to simultaneously analyze multiple proteins or parameters in the same sample. Several effective methodological approaches include:

  • Antibody conjugation strategies:

    • Direct conjugation of ECFP-Tag monoclonal antibodies to spectrally distinct fluorophores

    • Use of zenon labeling technology for temporary fluorophore attachment without purification

    • Selection of fluorophore combinations with minimal spectral overlap for multicolor imaging

  • Sequential staining protocols:

    • Perform serial rounds of staining, imaging, and signal removal

    • Use mild elution buffers (glycine-HCl, pH 2.5-3.0) to remove antibodies between rounds

    • Implement careful controls to ensure complete signal removal before subsequent staining

  • Species-specific secondary antibody strategy:

    • Use ECFP-Tag monoclonal antibodies from different host species (mouse, rabbit)

    • Detect with species-specific secondary antibodies conjugated to different fluorophores

    • Ensure secondary antibodies show no cross-reactivity between species

  • Isotype-specific detection:

    • Utilize ECFP-Tag monoclonal antibodies of different isotypes (IgG1, IgG2a, IgG2b)

    • Employ isotype-specific secondary antibodies for differential detection

  • Integration with microfluidic-based screening:

    • Incorporate ECFP-Tag monoclonal antibodies into microfluidic encapsulation systems

    • Combined with antigen bait sorting by conventional flow cytometry for high-throughput analysis

    • Enable multiplexed detection through various fluorescent channels

  • Spectral unmixing approaches:

    • Collect full emission spectra rather than discrete channels

    • Apply computational unmixing algorithms to separate overlapping signals

    • Particularly valuable when combining ECFP direct fluorescence with antibody-based detection

These multiplexed detection strategies are particularly valuable for complex experimental designs requiring simultaneous analysis of multiple proteins or cellular parameters.

How do monoclonal and polyclonal ECFP-Tag antibodies compare in research applications?

The choice between monoclonal and polyclonal ECFP-Tag antibodies significantly impacts experimental outcomes. Both options offer distinct advantages and limitations that researchers should consider:

CharacteristicMonoclonal ECFP-Tag AntibodiesPolyclonal ECFP-Tag Antibodies
SpecificityHigh specificity to single epitopeRecognize multiple epitopes on ECFP
Batch consistencyExcellent lot-to-lot reproducibilityBatch variation can be significant
Development timeline6-8 months for production 4 months for production
Signal strengthPotentially lower signal per moleculeHigher signal due to multiple binding sites
BackgroundGenerally lower backgroundMay exhibit higher background
ApplicationsExcellent for quantitative applicationsSuperior for detection of denatured proteins
Cost considerationsHigher initial development costGenerally more cost-effective for large quantities
Storage stabilityTypically stable at -20°C for 1 year Similar stability profile

Some research groups implement a hybrid approach, using polyclonal ECFP-Tag antibodies for capture (e.g., in immunoprecipitation) followed by monoclonal detection (e.g., in Western blot), combining the sensitivity of polyclonals with the specificity of monoclonals .

What factors should researchers consider when selecting between ECFP and other fluorescent protein tags?

Selecting between ECFP and alternative fluorescent protein tags requires careful consideration of several key factors that influence experimental success:

  • Spectral properties:

    • ECFP has excitation/emission maxima at approximately 439/476 nm

    • Consider potential spectral overlap with other fluorophores in multi-color experiments

    • Newer CFP variants (Cerulean, mTurquoise2) offer improved brightness and photostability

  • Brightness and photostability:

    • ECFP has lower intrinsic brightness compared to EGFP or YFP variants

    • Consider photobleaching characteristics for long-term imaging experiments

    • Balance detection sensitivity requirements with signal strength

  • Maturation and folding efficiency:

    • ECFP has intermediate maturation rates compared to other fluorescent proteins

    • Temperature and cellular environment affect maturation efficiency

    • Consider experimental timeframe and expression system

  • pH sensitivity:

    • ECFP exhibits moderate pH sensitivity

    • Critical for experiments in compartments with varying pH (endosomes, lysosomes)

    • Alternative tags may be preferable for acidic environments

  • Oligomerization potential:

    • Original ECFP has weak dimerization tendency at high concentrations

    • Monomeric versions (mCFP) minimize interference with fusion protein function

    • Critical for membrane proteins and other oligomerization-sensitive applications

  • FRET compatibility:

    • ECFP pairs effectively with YFP variants for FRET applications

    • Consider Förster radius and spectral overlap for optimal FRET efficiency

    • Newer CFP variants offer improved FRET performance

  • Availability of validated detection antibodies:

    • Commercial availability of high-quality ECFP-Tag monoclonal antibodies

    • Validated applications across multiple experimental platforms

    • Consistency and reliability of detection reagents

The selection decision should integrate these factors with specific experimental requirements and available detection systems.

How can researchers implement ECFP-Tag monoclonal antibodies in microfluidic-based high-throughput screening?

Integrating ECFP-Tag monoclonal antibodies into microfluidic-based high-throughput screening represents an advanced application that can significantly accelerate protein function and interaction studies. Implementation involves several key methodological considerations:

  • Microfluidic encapsulation system setup:

    • Single cells expressing ECFP-tagged proteins can be encapsulated into antibody capture hydrogels using droplet microfluidics

    • This approach enables processing rates of up to 10^7 cells per hour

    • The hydrogel matrix creates a stable capture environment around the cell, allowing for concentration of secreted proteins or surface-expressed ECFP-tagged proteins

  • Antibody detection system optimization:

    • ECFP-Tag monoclonal antibodies can be fluorescently labeled for direct detection

    • Alternatively, unlabeled primary antibodies can be detected with fluorescent secondary antibodies

    • Detection reagents must be carefully selected to maintain compatibility with the microfluidic system

  • Flow cytometry sorting parameters:

    • Encapsulated cells can be analyzed and sorted using conventional FACS instrumentation

    • Gates should be established using appropriate positive and negative controls

    • Index sorting capabilities allow correlation between cell phenotype and ECFP-tagged protein characteristics

  • Post-sorting analysis workflow:

    • Sorted cells can be subjected to single-cell sequencing

    • Alternatively, the ECFP-tagged proteins can be recombinantly expressed for further characterization

    • This approach enables the high-throughput study of protein-protein interactions, enzymatic activities, or ligand binding properties

  • Data integration and analysis:

    • Correlate ECFP-tag detection with other cellular parameters

    • Implement machine learning algorithms for pattern recognition

    • Establish quantitative relationships between ECFP-tagged protein expression and biological function

This microfluidic approach significantly improves throughput compared to traditional screening methods, enabling researchers to study ECFP-tagged proteins in a rapid, high-content format.

What are the best practices for using ECFP-Tag monoclonal antibodies in FRET experiments?

FRET (Förster Resonance Energy Transfer) experiments involving ECFP-tagged proteins require careful experimental design and rigorous controls. When incorporating ECFP-Tag monoclonal antibodies into these studies, researchers should follow these best practices:

  • Experimental design considerations:

    • ECFP commonly serves as the donor fluorophore in FRET pairs (typically with YFP variants as acceptors)

    • Antibody-based detection can be used to verify ECFP-tagged protein expression independently of direct fluorescence

    • Consider using ECFP variants with improved properties (mTurquoise2) for enhanced FRET efficiency

  • Antibody labeling strategies:

    • For indirect FRET measurements, label ECFP-Tag monoclonal antibodies with appropriate acceptor fluorophores

    • Optimize degree of labeling (DOL) to prevent self-quenching

    • Verify that antibody binding doesn't disrupt the native ECFP fluorescence properties

  • Controls for antibody-based FRET:

    • Donor-only samples (ECFP-tagged protein with unlabeled antibody)

    • Acceptor-only samples (untagged protein with labeled antibody)

    • Negative FRET controls (ECFP-tagged proteins with non-interacting partners)

    • Positive FRET controls (ECFP directly fused to acceptor fluorophore)

  • Data acquisition protocols:

    • Implement appropriate correction for spectral bleed-through

    • Collect complete emission spectra when possible

    • Use acceptor photobleaching to confirm FRET occurrence

  • Quantitative analysis approaches:

    • Calculate FRET efficiency using established equations

    • Implement pixel-by-pixel FRET analysis for spatial resolution

    • Consider fluorescence lifetime measurements (FLIM) for more robust FRET quantification

  • Validation strategies:

    • Compare antibody-based FRET results with direct fluorescence measurements

    • Use multiple analytical approaches to confirm interactions

    • Implement appropriate statistical analysis to establish significance

By following these methodological guidelines, researchers can effectively integrate ECFP-Tag monoclonal antibodies into FRET studies to investigate protein-protein interactions with high specificity and spatial resolution.

How are ECFP-Tag monoclonal antibodies being integrated with emerging single-cell analysis technologies?

The integration of ECFP-Tag monoclonal antibodies with cutting-edge single-cell technologies represents an exciting frontier in molecular biology research. Several emerging applications demonstrate the potential of this integration:

  • Single-cell proteomics:

    • ECFP-Tag monoclonal antibodies enable specific protein detection in mass cytometry (CyTOF) workflows

    • Metal-conjugated anti-ECFP antibodies allow quantification of tagged proteins alongside dozens of other cellular markers

    • Integration with microfluidic platforms enables high-throughput, multi-parameter analysis of ECFP-tagged proteins at single-cell resolution

  • Spatial transcriptomics integration:

    • Combining ECFP-Tag protein detection with in situ transcriptomics

    • Correlation between spatial protein distribution and gene expression patterns

    • Enhanced understanding of protein-RNA relationships in tissue context

  • Live-cell dynamics analysis:

    • ECFP-Tag monoclonal antibodies conjugated to cell-permeable fluorophores

    • Real-time monitoring of protein dynamics in living cells

    • Integration with advanced microscopy for 4D visualization of protein behavior

  • Single-cell multi-omics:

    • CITE-seq-like approaches incorporating ECFP-Tag detection

    • Simultaneous analysis of transcriptome and ECFP-tagged proteins

    • Correlation between gene expression and protein abundance/localization

  • Microfluidic encapsulation systems:

    • Single-cell encapsulation in antibody capture hydrogels

    • High-throughput sorting by conventional flow cytometry (10^7 cells per hour)

    • Integration with single-cell sequencing for genotype-phenotype correlations

These emerging applications highlight the versatility of ECFP-Tag monoclonal antibodies in advanced single-cell analysis workflows, enabling researchers to gain unprecedented insights into protein function and dynamics at the individual cell level.

What methodological approaches can enhance the sensitivity of ECFP-Tag detection in challenging samples?

Detecting ECFP-tagged proteins in challenging biological samples—such as those with high autofluorescence, low target abundance, or complex matrices—requires specialized methodological approaches. Researchers can implement several advanced strategies to enhance detection sensitivity:

  • Signal amplification methods:

    • Tyramide signal amplification (TSA) using ECFP-Tag monoclonal antibodies

    • Rolling circle amplification (RCA) for exponential signal enhancement

    • Proximity ligation assay (PLA) for detecting protein-protein interactions involving ECFP-tagged proteins

  • Background reduction strategies:

    • Spectral unmixing to separate ECFP signal from autofluorescence

    • Time-gated detection leveraging the relatively long fluorescence lifetime of ECFP

    • Photobleaching of endogenous fluorophores prior to antibody-based detection

  • Advanced microscopy techniques:

    • Structured illumination microscopy (SIM) for improved signal-to-noise ratio

    • Light sheet microscopy for reduced phototoxicity and background

    • Two-photon excitation for deeper tissue penetration and reduced photodamage

  • Sample preparation optimization:

    • Tissue clearing techniques compatible with immunofluorescence

    • Antigen retrieval methods to enhance epitope accessibility

    • Hydrogel embedding to preserve spatial relationships while allowing antibody penetration

  • Antibody engineering approaches:

    • Nanobody-based detection for improved tissue penetration

    • Fab fragments for reduced background in specific applications

    • Recombinant antibodies with enhanced affinity and specificity

  • Multiplexed detection strategies:

    • Sequential immunofluorescence with signal removal between rounds

    • Multispectral imaging to separate overlapping fluorescence signals

    • Mass cytometry (CyTOF) using metal-conjugated anti-ECFP antibodies

By implementing these advanced methodological approaches, researchers can significantly enhance the sensitivity and specificity of ECFP-Tag detection even in the most challenging biological samples, enabling new insights into protein function and localization.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.