This antibody is validated for multiple techniques:
Gastric Cancer: Anti-FGFR2 antibodies inhibited SNU-16 and OCUM-2M xenograft growth by >90% at 1 mg/kg doses .
Breast Cancer: FGFR2-ADC (auristatin-conjugated antibody) induced tumor regression in patient-derived xenografts with FGFR2 overexpression .
Biomarker Correlation: FGFR2IIIb/IIIc overexpression occurs in 4.9% of gastric carcinomas, linked to drug resistance and epithelial-mesenchymal transition .
Receptor Internalization: FGFR2 antibodies trigger receptor degradation, disrupting downstream signaling pathways (e.g., MAPK, AKT) .
Selectivity: >100-fold potency in FGFR2⁺ vs. FGFR2⁻ cell lines .
Specificity: Validated via ELISA, blocking assays, and cross-reactivity tests with FGFR family members .
Stability: Maintains activity for 12 months at 2–8°C when protected from light .
Lot Testing: Includes endotoxin level checks and performance validation in target applications .
FGFR2 (Fibroblast Growth Factor Receptor 2) is a transmembrane tyrosine kinase that functions as a cell-surface receptor for fibroblast growth factors, playing essential roles in cell proliferation, differentiation, migration, and apoptosis. It is crucial for embryonic development, including normal embryonic patterning, trophoblast function, limb bud development, lung morphogenesis, osteogenesis, and skin development . FGFR2 signaling is particularly important in the regulation of osteoblast differentiation and skeleton development, promoting proliferation in keratinocytes and immature osteoblasts while inducing apoptosis in differentiated osteoblasts . Its dysregulation has been implicated in various developmental disorders and cancers, making it a significant target for fundamental research and therapeutic development.
FGFR2 exists in multiple isoforms with structural variations that can impact antibody recognition. The extracellular domain includes three Ig-like domains in alpha isoforms or two Ig-like domains in beta isoforms . Additionally, alternative exon usage results in IIIb or IIIc isoforms that differ in their membrane-proximal Ig-like domain . When selecting a FITC-conjugated FGFR2 antibody, researchers should verify which epitopes and isoforms the antibody recognizes. For example, some antibodies are raised against synthetic peptides from specific regions (amino acids 520-570 in human FGFR2 isoform X6 protein) , while others may recognize epitopes common to multiple isoforms. This distinction is critical for experimental design, particularly when studying tissue-specific expression patterns or isoform-specific functions.
For optimal flow cytometry results with FGFR2 Antibody, FITC conjugated:
Harvest cells (1×10^6) and wash twice with ice-cold PBS containing 2% FBS
Resuspend cells in 100 μL of staining buffer (PBS with 2% FBS)
Add the FITC-conjugated FGFR2 antibody at the appropriate dilution (typically 1:50-1:200 for flow cytometry)
Incubate for 30 minutes at 4°C in the dark
Wash cells twice with staining buffer to remove unbound antibody
Resuspend in 400-500 μL of staining buffer for analysis
Include appropriate isotype control antibody (FITC-conjugated IgG of the same species) to establish background fluorescence
For KATO-III human gastric carcinoma cell line and similar FGFR2-expressing cells, this protocol has successfully demonstrated specific binding as evidenced by the clear separation between the antibody-stained samples and isotype controls . Protect samples from light throughout the procedure to maintain FITC fluorescence intensity.
To maintain optimal activity of FGFR2 Antibody, FITC conjugated:
Most FITC-conjugated antibodies maintain activity for approximately 12 months from the date of receipt when stored properly . Unlike some other conjugated antibodies, FITC-conjugated antibodies should not be frozen after reconstitution as this can affect the fluorochrome stability . Aliquoting into smaller volumes upon receipt is recommended to minimize freeze-thaw cycles.
When designing dual staining experiments to study FGFR2-ligand interactions:
Antibody panel design: Pair FGFR2 Antibody, FITC conjugated (excitation/emission: 494nm/518nm) with fluorochromes having minimal spectral overlap such as APC (650nm/660nm) or PE (565nm/578nm) for ligand or co-receptor detection
Sequential staining protocol:
First stain with unconjugated anti-FGF ligand antibody
Add fluorophore-conjugated secondary antibody
Block with excess IgG from the same species as the first primary
Add FGFR2 Antibody, FITC conjugated
This prevents cross-reactivity between antibodies
Compensation controls:
Single-stained controls for each fluorochrome
FMO (Fluorescence Minus One) controls to establish gating boundaries
Isotype controls for both FITC and second fluorochrome
Receptor-ligand interaction assessment:
Fixed ratio analysis: Calculate the ratio of FGFR2:ligand across different experimental conditions
Proximity ligation assay: Combine with flow cytometry to detect genuine receptor-ligand complexes
FRET (Fluorescence Resonance Energy Transfer): If using compatible fluorophore pairs
This approach has been successfully employed to examine the interaction between FGFR2 and its ligands in gastric carcinoma cells, demonstrating activation of downstream signaling cascades .
When working with tissues exhibiting high autofluorescence (such as liver, kidney, or brain):
Tissue-specific autofluorescence reduction:
Treat sections with 0.1-1% sodium borohydride for 10 minutes
Incubate with 0.1-0.3% Sudan Black B in 70% ethanol for 20 minutes
Use commercial autofluorescence quenching reagents specific for aldehyde-fixed tissues
Advanced imaging techniques:
Spectral unmixing: Acquire full emission spectra and computationally separate FITC signal from autofluorescence
Time-gated imaging: Exploit the longer lifetime of FITC compared to autofluorescence
Confocal microscopy with narrow bandpass filters: Restrict detection to FITC's peak emission wavelength
Alternative detection strategies:
Quantification adjustment:
Always subtract background autofluorescence values from neighboring unstained tissue
Apply tissue-specific correction factors based on known autofluorescence patterns
Employ computational image analysis with machine learning algorithms for signal separation
These approaches have significantly improved signal-to-noise ratios in challenging tissues where FGFR2 expression needed to be accurately quantified.
To simultaneously assess FGFR2 activation status and total expression levels:
Dual antibody approach:
Use FGFR2 Antibody, FITC conjugated for total receptor detection
Pair with a phospho-specific antibody against phosphorylated FGFR2 (Y653/654) conjugated to a spectrally distinct fluorophore
Apply after appropriate stimulation with FGF ligands
Flow cytometry protocol refinement:
Include phosphatase inhibitors (2mM sodium orthovanadate, 10mM sodium fluoride) in all buffers
Perform fixation with 4% paraformaldehyde followed by permeabilization with 90% ice-cold methanol
Stain for phospho-FGFR2 first, followed by total FGFR2
Calculate activation index as the ratio of phospho-FGFR2 to total FGFR2
Signaling dynamics assessment:
Perform time-course experiments following FGF stimulation (0, 5, 15, 30, 60 minutes)
Create phosphorylation kinetics curves normalized to total FGFR2 expression
Include inhibitor controls (e.g., FGFR-specific tyrosine kinase inhibitors)
Downstream signaling correlation:
This methodology provides quantitative assessment of receptor activation status while controlling for variations in total receptor expression, offering deeper insights into signaling dynamics than either measurement alone.
When investigating rare FGFR2-expressing cell populations (frequency <0.1%):
Sample enrichment strategies:
Negative selection to remove abundant cell types
Density gradient centrifugation to isolate specific cell fractions
Magnetic pre-enrichment of cells expressing markers co-expressed with FGFR2
Staining optimization:
Flow cytometry refinements:
Collect more events (minimum 1×10^6, ideally 5-10×10^6)
Lower flow rate for improved resolution
Sequential gating strategy using additional markers
Inclusion of viability dye to exclude dead cells
Implementation of "dump channel" with lineage markers
Statistical validation:
Calculate minimum event count for statistical significance
Perform replicate samples to establish variability
Include fluorescence-minus-one (FMO) controls for accurate gating
Apply rare event statistical algorithms
This approach has been successfully employed to identify and characterize rare FGFR2-positive progenitor cell populations in developmental studies and cancer stem cell research.
The choice between monoclonal and polyclonal FGFR2 antibodies significantly impacts experimental results:
For quantitative analysis requiring high reproducibility and specificity, monoclonal antibodies like clone 98739 are preferred. For detection of FGFR2 in tissues with potentially masked epitopes or when studying multiple species, polyclonal antibodies may provide advantages. When reporting results, researchers should clearly specify which type was used as this fundamentally affects data interpretation and cross-study comparisons.
A comprehensive validation protocol for FGFR2 Antibody, FITC conjugated should include:
Positive and negative cell/tissue controls:
Technical controls:
Specificity verification:
Blocking peptide competition assay using the immunizing peptide
siRNA or CRISPR knockout validation in relevant cell lines
Western blot correlation with flow cytometry results
Comparison with other validated anti-FGFR2 antibodies
Dilution optimization:
Performance metrics documentation:
Coefficient of variation across replicates
Limit of detection calculation
Dynamic range assessment
Staining index calculation
This validation workflow ensures reliable, reproducible results and should be thoroughly documented in any publication's methods section.
When encountering inconsistent staining patterns with FGFR2 Antibody, FITC conjugated:
Common causes and solutions:
Issue | Potential Cause | Resolution Strategy |
---|---|---|
Weak or no signal | Antibody degradation | Check storage conditions; use fresh aliquot |
Low FGFR2 expression | Increase antibody concentration; try signal amplification | |
Epitope masking | Optimize antigen retrieval; try alternative fixation | |
High background | Non-specific binding | Increase blocking; reduce antibody concentration |
Autofluorescence | Use appropriate quenching reagents; spectral unmixing | |
FITC instability | Protect from light; prepare fresh dilutions | |
Variable cell-to-cell staining | Heterogeneous expression | Single-cell analysis; confirm with alternative methods |
Cell cycle dependence | Synchronize cells; co-stain with cell cycle markers |
Biological interpretation considerations:
FGFR2 expression is developmentally regulated and tissue-specific
Alternative splicing creates isoforms with different antibody recognition
FGFR2 can be internalized upon ligand binding, affecting membrane staining
Receptor shedding may occur in certain conditions
Methodological approaches:
Sequential staining with independent FGFR2 antibodies recognizing different epitopes
Correlation with mRNA expression by combining with RNA-FISH
Time-course experiments to detect dynamic changes in receptor localization
Use multiple detection methods (e.g., flow cytometry and immunofluorescence)
These systematic troubleshooting approaches help distinguish between technical artifacts and true biological variability in FGFR2 expression patterns.
While traditional FITC-conjugated antibodies are designed for fluorescence detection, researchers can leverage FGFR2 antibodies in CyTOF workflows through these approaches:
Indirect metal labeling:
Primary staining with unconjugated FGFR2 antibody (same clone as FITC-conjugated version)
Secondary staining with metal-conjugated anti-species antibody
This maintains epitope recognition while adapting to CyTOF platform
Direct antibody metal conjugation:
Optimized CyTOF panel design:
Data analysis considerations:
Apply dimensionality reduction techniques (tSNE, UMAP)
Implement clustering algorithms to identify cell populations
Perform trajectory analysis to map developmental pathways
This integration enables simultaneous measurement of FGFR2 expression alongside dozens of other parameters without fluorescence spectral overlap limitations, providing unprecedented insight into FGFR2's role in complex cellular systems.
Adapting FGFR2 Antibody, FITC conjugated from in vitro to in vivo imaging requires several methodological adjustments:
Biological considerations:
Blood half-life: FITC-conjugated antibodies typically have 4-8 hour circulation time
Tissue penetration: Limited by antibody size (150 kDa) and FITC's excitation/emission properties
Target accessibility: Only extracellular epitopes of FGFR2 are accessible in vivo
Background autofluorescence: Significantly higher in vivo, particularly in certain tissues
Technical adaptations:
Dosage optimization: Typically 1-5 μg/g body weight (much higher than in vitro concentrations)
Administration route: Intravenous injection for systemic imaging; local injection for specific tissues
Timing: Peak signal-to-background ratio typically occurs 24-48 hours post-injection
Imaging windows: May require surgical preparation for deep tissue visualization
Alternative approaches:
Consider near-infrared fluorophore conjugates instead of FITC for better tissue penetration
Use F(ab')2 or Fab fragments for improved pharmacokinetics
Employ two-photon microscopy for deeper tissue penetration
Combine with tissue clearing techniques for ex vivo imaging
Controls and validation:
Include blocking studies with unlabeled antibody
Perform ex vivo tissue analysis to confirm in vivo observations
Use FGFR2-knockout or knockdown models as negative controls
These considerations are essential when transitioning from controlled in vitro environments to the complexities of in vivo systems, particularly when studying FGFR2's role in developmental processes or tumor growth.
To investigate FGFR2 internalization and trafficking using FITC-conjugated antibodies:
Pulse-chase experimental design:
Pulse: Incubate live cells with FGFR2 Antibody, FITC conjugated at 4°C (prevents internalization)
Wash: Remove unbound antibody
Chase: Warm cells to 37°C with or without FGF ligands
Fix at various timepoints (0, 5, 15, 30, 60, 120 minutes)
Subcellular colocalization analysis:
Co-stain with markers for different endocytic compartments:
Early endosomes: Anti-EEA1
Recycling endosomes: Anti-Rab11
Late endosomes: Anti-Rab7
Lysosomes: Anti-LAMP1
Quantify colocalization using Pearson's or Mander's coefficients
Live-cell imaging optimization:
Use spinning disk confocal microscopy for rapid acquisition
Implement temperature control systems
Add minimal essential medium with reduced phenol red
Employ computational tracking algorithms for vesicle movement
Quantitative assessment metrics:
Internalization rate: Measure decrease in surface FITC signal over time
Recycling rate: Measure reappearance of FITC signal at membrane
Degradation kinetics: Measure total FITC signal decrease
Calculate half-lives for each process
This approach provides quantitative insights into how FGFR2 trafficking is regulated in normal development and how it may be dysregulated in pathological conditions, with implications for understanding receptor downregulation mechanisms and potential therapeutic interventions.
When faced with discrepancies between FGFR2 protein detection using FITC-conjugated antibodies and mRNA expression data:
Integrated validation approach:
Simultaneous protein and mRNA detection:
Flow-FISH technology combining antibody staining with RNA-FISH
Sequential immunofluorescence and in situ hybridization
Single-cell proteogenomic analysis
Technical reconciliation strategies:
Biological explanations for discrepancies:
Post-transcriptional regulation (miRNAs, RNA-binding proteins)
Protein stability and half-life differences
Alternative splicing affecting antibody epitope recognition
Spatial segregation of mRNA versus protein within cells
Temporal delays between transcription and translation
Statistical analysis framework:
Correlation analysis between protein and mRNA at single-cell level
Bayesian integration of multiple measurement modalities
Machine learning approaches to identify patterns in discrepant results
Meta-analysis of published literature for similar discrepancies
This methodological framework helps researchers distinguish between technical artifacts and genuine biological phenomena, leading to more accurate interpretation of FGFR2 expression patterns in development and disease.
The landscape of FGFR2 detection is evolving with several emerging technologies that may supersede or complement traditional FITC conjugation:
Advanced fluorophore alternatives:
Quantum dots: Higher photostability, narrower emission spectra
Silicon rhodamines: Far-red emission with minimal autofluorescence overlap
Polymer-based fluorophores: Enhanced brightness and stability
Lanthanide chelates: Time-resolved fluorescence capabilities
Proximity-based detection methods:
Proximity Extension Assay (PEA): Oligonucleotide-antibody conjugates
Fluorescence Resonance Energy Transfer (FRET) pairs
NanoBiT complementation for protein-protein interaction studies
Luminescent Oxygen Channeling Immunoassay (LOCI)
Label-free approaches:
Surface Plasmon Resonance imaging for receptor dynamics
Mass spectrometry imaging for in situ protein detection
Raman microspectroscopy for chemical fingerprinting
Interferometric detection methods
Next-generation reporters:
CRISPR-based endogenous tagging of FGFR2
Split fluorescent protein complementation
Photoactivatable fluorescent proteins for super-resolution imaging
Genetically encoded biosensors for FGFR2 activation
These technologies promise higher sensitivity, multiplexing capabilities, and dynamic range compared to traditional FITC conjugation, potentially revolutionizing our understanding of FGFR2 biology in development, homeostasis, and disease.
When confronted with discrepancies between different fluorophore-conjugated FGFR2 antibodies:
Systematic cross-comparison protocol:
Side-by-side staining with identical conditions except for the conjugated antibody
Fluorophore normalization using quantitative beads
Matched concentration titration experiments
Cross-blocking experiments to determine epitope overlap
Technical factor analysis:
Fluorophore physicochemical properties:
Conjugation chemistry differences affecting antibody avidity
Fluorophore-to-protein ratio variations
Different clones with distinct epitope recognition
Multiparametric validation strategy:
RNA-scope or qPCR correlation to establish "ground truth"
Unconjugated primary antibody with different secondary detection systems
Functional assays (phosphorylation, internalization) with each conjugate
Cross-validation with orthogonal techniques (Western blot, ELISA)
Standardized reporting framework:
Document conjugation method and fluorophore-to-protein ratio
Report antibody concentration in molar terms rather than dilution
Specify exact clone, epitope, and binding characteristics
Include all tested conditions in supplementary materials