GFP Antibody

Green Fluorescent Protein, Mouse Antibody
Shipped with Ice Packs
In Stock

Description

Introduction to GFP Antibodies

GFP antibodies are immunoglobulins designed to specifically recognize and bind Green Fluorescent Protein (GFP), a 27 kDa monomeric protein derived from the jellyfish Aequorea victoria. GFP’s intrinsic fluorescence (emission peak at 508 nm) makes it a cornerstone tool for studying protein localization, gene expression, and real-time cellular dynamics in fixed or live cells . Anti-GFP antibodies enhance detection sensitivity, circumvent GFP’s limitations in denatured conditions, and enable applications like immunoprecipitation or Western blotting where native fluorescence is insufficient . These antibodies are available in polyclonal or monoclonal forms, with diverse host sources (e.g., goat, rabbit, mouse) and conjugates (e.g., fluorophores, enzymes) tailored to specific experimental needs .

Types and Hosts of GFP Antibodies

GFP antibodies vary in clonality, host species, and conjugation, influencing their specificity, stability, and application suitability:

FeaturePolyclonalMonoclonal
HostGoat, rabbit, chicken (IgY)Mouse, rabbit
Epitope BindingMultiple epitopesSingle epitope
ApplicationsWestern blot, ELISA, IF, IPIF, IHC, FACS, WB (selective)
AdvantagesBroad reactivity, signal amplificationHigh specificity, consistency

Host-Specific Characteristics:

  • Goat: High affinity, validated for ELISA and WB .

  • Rabbit: Polyclonal antibodies enable signal amplification via multiple epitope binding .

  • Mouse (e.g., GFP-G1): Monoclonal antibodies suited for IF/IHC but ineffective in WB .

  • Chicken (IgY): Larger IgY antibodies resist proteolysis and avoid human Fc receptor interference .

Key Applications of GFP Antibodies

GFP antibodies are pivotal in diverse methodologies:

Detection and Localization

  • Immunofluorescence (IF): Amplifies GFP signals in fixed cells, compensating for photobleaching or low expression .

  • Western Blot (WB): Detects denatured GFP in lysates; varies by antibody efficacy (e.g., Rockland’s goat antibody vs. DSHB’s mouse GFP-G1) .

  • Flow Cytometry (FACS): Identifies GFP-expressing cells using conjugated antibodies (e.g., Alexa Fluor® 488) .

Functional Studies

  • Immunoprecipitation (IP): Isolates GFP-tagged proteins for interaction studies .

  • Chromogenic IHC: Enables tissue analysis in transgenic models using HRP/alkaline phosphatase conjugates .

  • Nanobody Applications: Camelid-derived anti-GFP nanobodies (e.g., cAbGFP4) enable live-cell imaging and super-resolution microscopy due to their small size and stability .

Nanobodies for Advanced Imaging

Camelid nanobodies, like cAbGFP4, exhibit:

  • High Affinity: Binding constants as low as 0.23 nM .

  • Stability: Function in intracellular environments without aggregation .

  • Versatility: Used in X-ray crystallography and isothermal titration calorimetry to map GFP interactions .

Signal Amplification and Fluorophore Options

Jackson ImmunoResearch offers conjugates with distinct emission profiles to bypass autofluorescence or equipment limitations:

ConjugateExcitation (nm)Emission (nm)Application
Alexa Fluor® 488493519Standard IF/FACS
Alexa Fluor® 647651667Long-wavelength imaging
R-Phycoerythrin488580High-sensitivity FACS
HRP/Alkaline PhosphataseN/AN/AChromogenic IHC

Data adapted from Jackson ImmunoResearch .

Antibody Performance in Transgenic Models

  • Tissue Detection: Rabbit Anti-GFP conjugates enable chromogenic staining of GFP fusion proteins in formalin-fixed, paraffin-embedded (FFPE) tissues .

  • Flow Cytometry Validation: Mouse Anti-GFP (MAB42401R) distinguishes eGFP-transfected HEK293 cells from untransfected controls .

Autofluorescence Mitigation

  • Strategic Conjugates: Use far-red fluorophores (e.g., Alexa Fluor® 647) to avoid overlap with cellular autofluorescence .

  • Signal Amplification: Polyclonal antibodies enhance detection in low-expression scenarios .

Optimization Considerations

  • Epitope Accessibility: Fixation methods (e.g., paraformaldehyde vs. methanol) impact antibody binding .

  • Cross-Reactivity: Ensure antibodies recognize GFP variants (eGFP, Venus) or wild-type GFP .

Product Specs

Introduction
Green Fluorescent Protein (GFP), derived from the jellyfish Aequorea Victoria, emits bioluminescence in the green spectrum. This versatile protein serves as a fluorescent tag for creating chimeric proteins. Widely expressed in various cell types, GFP acts as a non-invasive fluorescent marker in living cells and organisms. Its applications encompass cell lineage tracing, gene expression reporting, and protein-protein interaction studies.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
1 mg/ml in PBS, pH 7.4, with 0.1% Sodium Azide.
Storage Procedures
Store at 4°C for up to 1 month. For extended storage, keep at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
Stable for 12 months at -20°C and 1 month at 4°C.
Applications
This GFP antibody has undergone ELISA and Western blot validation to ensure specificity and reactivity. However, optimization is recommended for individual applications. For Western blot analysis, a dilution range of 1:1000 to 1:2000 is suggested, with an initial dilution of 1:1000.
Synonyms
Green fluorescent protein, GFP.
Purification Method
GFP antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Antibody Monoclonal.
Clone
PAT2G5AT.
Immunogen
Anti GFP mAb is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant GFP amino acids 1-238 purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and κ light chain.

Q&A

What is GFP and why are GFP antibodies needed in research?

GFP is a 27 kDa cytoplasmic protein originally isolated from the jellyfish Aequorea victoria. While GFP itself is fluorescent (excitation at 488nm, emission at 509nm), GFP antibodies provide several advantages:

  • Enhanced detection sensitivity when natural GFP fluorescence is weak

  • Ability to amplify signals for quantitative applications

  • Detection of GFP in fixed samples where native fluorescence may be compromised

  • Compatibility with various applications beyond fluorescence microscopy

  • Capacity to detect GFP fusion proteins in applications like western blotting and immunoprecipitation

GFP antibodies are particularly valuable for tracking protein expression and localization in both living and fixed cells .

What are the major variants of GFP and how do they affect antibody selection?

Several GFP variants have been developed with improved properties:

VariantKey CharacteristicsAntibody Considerations
Wild-type GFPOriginal form, lower brightnessDetected by most GFP antibodies
Enhanced GFP (eGFP)6× brighter, faster maturation, reduced temperature sensitivityWidely recognized by commercial antibodies
S65T-GFPSimplified excitation spectrum with single peak at ~490 nmGenerally recognized by GFP antibodies
RS-GFPRed-shifted spectrum, valuable for FRET experimentsCheck specific cross-reactivity
YFPYellow emission instead of greenMay require specific YFP antibodies

What are the primary applications for GFP antibodies in research?

GFP antibodies are versatile tools with multiple applications:

  • Western blotting: For detecting and quantifying GFP fusion proteins

  • Immunofluorescence (IF): To enhance visualization of GFP-tagged proteins in fixed cells

  • Immunohistochemistry (IHC): For detection in tissue sections

  • Flow cytometry (FC): For cell sorting based on GFP expression

  • Immunoprecipitation (IP): To isolate GFP-tagged protein complexes

  • Electron microscopy (EM): For ultrastructural localization studies

  • Fluorescence in situ hybridization (FISH): For combined protein-nucleic acid detection

  • Chromatin immunoprecipitation (ChIP): For studying DNA-protein interactions

The versatility of these antibodies makes them indispensable for a wide range of molecular and cellular biology experiments .

What controls should be included when working with GFP antibodies?

Proper experimental controls are critical for reliable results:

  • Negative control: Non-transfected cells to establish background signal

  • Positive control: Cells known to express GFP at detectable levels

  • Antibody controls:

    • GFP-negative cells + antibody (to assess non-specific binding)

    • GFP-positive cells + antibody (to confirm specific detection)

  • Secondary antibody only: To identify background from secondary antibodies

  • Isotype control: To assess non-specific binding when using monoclonal antibodies

These controls help with accurate data interpretation, especially for flow cytometry gating and immunofluorescence specificity assessment .

How do I optimize GFP antibody dilution for different applications?

Optimal antibody dilution varies by application, sample type, and specific antibody. A methodical approach includes:

  • Start with manufacturer's recommended range

  • Perform a titration experiment using multiple dilutions (e.g., 1:500, 1:1000, 1:2000, 1:5000)

  • Include positive and negative controls at each dilution

  • Evaluate signal-to-noise ratio

  • Select the dilution giving the strongest specific signal with minimal background

For example, some immunofluorescence applications may use GFP antibody at approximately 1.7 μg/mL, while western blots might require 1 μg/mL .

How can GFP antibodies be used in FACS for isolating GFP-expressing cells?

Fluorescence-Activated Cell Sorting (FACS) with GFP antibodies is useful for isolating transfected cells, particularly when endogenous GFP signal is weak:

  • Sample preparation: Harvest cells according to standard protocols

  • Fixation/permeabilization: For intracellular GFP, fix cells with paraformaldehyde and permeabilize with saponin or similar agent

  • Antibody staining: Incubate with anti-GFP primary antibody followed by fluorophore-conjugated secondary antibody

  • Controls: Set up compensation controls and establish gating parameters using:

    • GFP-negative cells

    • GFP-positive cells

    • GFP-negative cells + antibody

    • GFP-positive cells + antibody

  • Cell sorting: Gate cells based on fluorescence intensity and collect desired populations

This approach is particularly valuable in CRISPR experiments where transfected cells express GFP alongside sgRNA, although researchers should consider the trade-off between additional antibody staining and cell stress/viability .

What are the differences between polyclonal and monoclonal GFP antibodies?

The choice between polyclonal and monoclonal GFP antibodies depends on specific research needs:

FeaturePolyclonal GFP AntibodiesMonoclonal GFP Antibodies
Target epitopesMultiple epitopes on GFPSingle epitope on GFP
SensitivityGenerally higher (multiple binding sites)Generally lower (single binding site)
SpecificityModerate (potential cross-reactivity)High (epitope-specific)
Lot-to-lot variabilityHigherLower
Best applicationsSignal amplification, IP, IHCPrecise epitope detection, quantitative analysis
Example productsGoat anti-GFP polyclonal (Rockland 600-101-215)Various mouse monoclonals

Polyclonal antibodies provide superior signal amplification and epitope recognition after various fixation methods, while monoclonals offer consistency and specificity for standardized protocols .

How do I troubleshoot weak or non-specific signals when using GFP antibodies?

Common issues with GFP antibody staining can be systematically addressed:

For weak signals:

  • Increase antibody concentration or incubation time

  • Optimize fixation to better preserve epitopes

  • Use signal amplification systems (e.g., tyramide signal amplification)

  • Try alternative antibody clones with higher affinity

  • Use a more sensitive detection system

For non-specific signals:

  • Increase blocking time/concentration

  • Use different blocking agents (BSA, serum, commercial blockers)

  • Increase washing stringency (time, detergent concentration)

  • Reduce antibody concentration

  • Pre-absorb antibodies against non-specific targets

  • Use more specific antibodies (consider monoclonals)

For high background:

  • Include autofluorescence quenching steps

  • Adjust imaging parameters to minimize autofluorescence

  • Use fluorophores with emission spectra distinct from cellular autofluorescence

  • Try different fixation methods that minimize background

Systematic optimization of these parameters can significantly improve signal quality .

How do fixation and permeabilization methods affect GFP antibody staining?

Different fixation and permeabilization approaches significantly impact GFP antibody staining:

MethodEffect on Native GFP FluorescenceEffect on Antibody AccessibilityBest For
Paraformaldehyde (PFA)Preserves wellModerate epitope maskingBalancing native GFP and antibody staining
MethanolTypically quenchesGood epitope exposureApplications where only antibody signal is needed
GlutaraldehydePreserves well but causes autofluorescenceSignificant epitope maskingElectron microscopy
Triton X-100 (permeabilization)Minimal effectEffective for intracellular accessCytoplasmic proteins
Saponin (permeabilization)Minimal effectGentler, reversibleMembrane proteins, flow cytometry

As demonstrated in search result , a common approach for intracellular GFP staining in flow cytometry involves fixation with paraformaldehyde followed by permeabilization with saponin .

What are the considerations for using GFP antibodies in live versus fixed cells?

Using GFP antibodies in different sample preparations involves distinct considerations:

Live cells:

  • Standard antibodies cannot penetrate intact cell membranes, limiting detection to cell surface GFP

  • Cell viability may be compromised by permeabilization methods

  • Temperature sensitivity requires physiological conditions during staining

  • Consider non-toxic, membrane-permeable nanobodies for intracellular GFP detection in live cells

Fixed cells:

  • Fixation method affects both GFP fluorescence and epitope accessibility

  • Permeabilization is required for intracellular GFP detection

  • Higher signal-to-noise ratio is generally achievable

  • More flexibility in staining conditions (time, temperature, buffer composition)

Researchers should carefully evaluate their experimental requirements and prioritize cell viability or signal strength accordingly .

How can GFP antibodies enhance CRISPR/Cas9 experiments for isolating successfully edited cells?

In CRISPR/Cas9 experiments, GFP is often used as a reporter for transfection or editing. GFP antibodies can play a crucial role in enriching for successfully edited cells:

  • Experimental design strategies:

    • CRISPR vectors often include GFP as a reporter linked to Cas9 or as a separate expression cassette

    • GFP expression serves as a proxy for successful transfection

  • Sorting methodology:

    • Use FACS with anti-GFP antibodies to enhance detection and isolation

    • Sort based on GFP fluorescence intensity (higher intensity may correlate with better sgRNA expression)

    • Consider antibody-based enhancement when endogenous GFP signal is weak

  • Critical considerations:

    • Balance between antibody enhancement and cell stress

    • Correlation between GFP signal strength and actual editing efficiency

    • Viability of sorted cells for downstream applications

    • Need for aseptic conditions and mycoplasma testing after sorting

  • Validation strategies:

    • Confirm editing in sorted population using sequencing or functional assays

    • Consider expanding cells post-sorting to recover from stress

This approach is particularly valuable when the GFP signal from CRISPR vectors is weaker than optimal for direct sorting .

What methodologies exist for using GFP antibodies in multiplex imaging?

Multiplex imaging with GFP antibodies allows simultaneous visualization of multiple targets:

  • Fluorophore selection strategy:

    • Choose secondary antibody fluorophores with minimal spectral overlap

    • Consider brightness hierarchy (assign brighter fluorophores to less abundant targets)

    • Account for potential bleed-through from GFP's natural fluorescence

    • Utilize narrow bandwidth filters to minimize overlap

  • Staining optimization:

    • Sequential staining may reduce cross-reactivity compared to simultaneous staining

    • Determine optimal antibody order for sequential protocols

    • Use antibodies from different species to enable species-specific secondaries

  • Essential controls:

    • Single-stained controls for spectral unmixing

    • Fluorescence Minus One (FMO) controls for accurate discrimination

    • Absorption controls to assess potential energy transfer between fluorophores

  • Technical approaches:

    • Linear unmixing algorithms for overlapping spectra

    • Multi-round imaging with antibody stripping between rounds

    • Indirect immunofluorescence with species-specific secondaries

    • Direct conjugated antibodies to eliminate secondary cross-reactivity

Many GFP antibodies are specifically validated for multiplex applications, making them valuable components of complex imaging panels .

How can GFP antibodies be utilized in super-resolution microscopy techniques?

Super-resolution microscopy with GFP antibodies enables visualization beyond the diffraction limit:

  • STORM/PALM applications:

    • Use GFP antibodies conjugated to photoswitchable fluorophores

    • Alternatively, use unconjugated primary GFP antibodies with appropriate secondary antibodies

    • Optimize buffer conditions for photoswitching behavior

    • Collect thousands of frames with different subsets of molecules in the "on" state

  • STED microscopy considerations:

    • Select secondary antibodies with fluorophores optimized for STED (e.g., ATTO or Abberior dyes)

    • Consider photobleaching resistance and depletion efficiency

    • Optimize depletion laser power to balance resolution and signal strength

  • SIM methodology:

    • Less demanding on fluorophore properties than STORM or STED

    • Ensure sufficient signal-to-noise ratio

    • Use bright and photostable fluorophores

    • Requires high-quality sample preparation to minimize artifacts

  • Critical parameters across techniques:

    • Linkage error: Distance between GFP and fluorophore affects achievable resolution

    • Labeling density: Critical for single-molecule localization techniques

    • Sample drift: Must be minimized during extended acquisitions

    • Fixation quality: Sample preparation is crucial for structural preservation

The variety of available GFP antibody conjugates makes them particularly suitable for super-resolution applications .

How can GFP antibodies be optimized for ChIP-seq experiments with GFP-tagged transcription factors?

ChIP-seq with GFP-tagged transcription factors requires careful optimization:

  • Experimental design:

    • Express GFP-tagged transcription factor in target cells

    • Verify expression and nuclear localization by microscopy before proceeding

    • Consider the impact of GFP tag on transcription factor function

  • Chromatin preparation:

    • Optimize crosslinking time (typically 10-15 minutes with 1% formaldehyde)

    • Adjust sonication parameters to achieve ideal fragment size (200-500 bp)

    • Verify fragmentation efficiency by agarose gel electrophoresis

  • Immunoprecipitation optimization:

    • Test different GFP antibodies for IP efficiency

    • Determine optimal antibody:chromatin ratio

    • Adjust wash stringency to balance signal and noise

    • Include appropriate controls (IgG, input DNA)

  • Quality control measures:

    • qPCR validation at known binding sites before sequencing

    • Assessment of enrichment over background

    • Library quality evaluation before high-throughput sequencing

  • Advantages over epitope tag ChIP:

    • Visualization of expression and localization via GFP fluorescence

    • Potential for combining with live-cell imaging prior to ChIP

    • Well-characterized antibodies with high specificity

This approach provides a powerful way to identify genome-wide binding sites of transcription factors without requiring factor-specific antibodies .

What strategies exist for using GFP antibodies in quantitative proteomics of GFP-tagged proteins?

GFP antibodies enable several quantitative proteomics approaches:

  • Co-immunoprecipitation coupled to mass spectrometry:

    • Use GFP antibodies to pull down GFP-tagged proteins and interacting partners

    • Elute under native or denaturing conditions depending on interaction strength

    • Analyze by LC-MS/MS to identify and quantify interaction partners

    • Compare to control immunoprecipitations to identify specific interactions

  • Targeted proteomics approaches:

    • Develop selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) assays

    • Use GFP antibody enrichment prior to targeted MS analysis

    • Quantify GFP-tagged proteins and interaction partners with high sensitivity

  • Absolute quantification strategies:

    • Use isotope-labeled peptide standards for absolute quantification

    • Calculate stoichiometry of protein complexes

    • Determine copy number of GFP-tagged proteins per cell

  • Proximity-dependent labeling:

    • Fuse GFP-tagged protein with BioID or APEX2

    • Perform proximity labeling followed by GFP antibody purification

    • Identify proximal proteins by mass spectrometry

    • Quantify relative proximity through label-free or labeled quantification

These approaches leverage the specificity of GFP antibodies for precise protein complex analysis .

Product Science Overview

Introduction to Green Fluorescent Protein (GFP)

Green Fluorescent Protein (GFP) is a protein that exhibits bright green fluorescence when exposed to light in the blue to ultraviolet range. Originally isolated from the jellyfish Aequorea victoria, GFP has become a crucial tool in molecular and cellular biology due to its unique properties .

Discovery and Development

GFP was first purified in the 1960s and 1970s by Osamu Shimomura, who studied its properties along with the luminescent protein aequorin . The gene encoding GFP was cloned in 1992 by Douglas Prasher, and Martin Chalfie’s lab successfully expressed the sequence in vivo . Roger Tsien’s lab later improved GFP, making it a widely used research tool . In recognition of their contributions, Shimomura, Chalfie, and Tsien were awarded the Nobel Prize in Chemistry in 2008 .

Properties and Applications

GFP has a major excitation peak at a wavelength of 395 nm and a minor one at 475 nm, with an emission peak at 509 nm . This makes it an excellent tool for various biological applications, including:

  • Gene Expression Monitoring: GFP can be used as a reporter gene to monitor gene expression in living organisms .
  • Protein Localization and Dynamics: GFP-tagged proteins can be visualized to study their localization and movement within cells .
  • Biosensors: Modified forms of GFP have been used to create biosensors for detecting various cellular conditions .
Mouse Antibody for GFP

Monoclonal antibodies against GFP, such as those produced in mice, are essential tools for detecting and studying GFP-tagged proteins. These antibodies are typically derived from hybridoma cells produced by fusing mouse myeloma cells with splenocytes from immunized mice . Mouse monoclonal antibodies against GFP are highly specific and can be used in various techniques, including Western blotting, immunoprecipitation, and ELISA .

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.