FGFR2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Research Applications

This antibody is validated for multiple techniques:

ApplicationProtocol DetailsSensitivity
Flow CytometryDetects FGFR2 on KATO-III gastric carcinoma cells 1:50–1:100 dilution
ImmunohistochemistryLocalizes FGFR2 in formalin-fixed tissues 1:10,000 dilution
Western BlottingIdentifies FGFR2 isoforms (~92–130 kDa) 1:500 dilution
ImmunofluorescenceVisualizes FGFR2 in fixed cells 1:150 dilution

Cancer Therapeutics

  • 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 .

Mechanism of Action

  • 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 .

Quality Control and Validation

  • 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 .

Limitations and Considerations

  • Isoform Specificity: Some antibodies may not distinguish between IIIb and IIIc isoforms without additional validation .

  • Tissue Penetration: Large antibody size limits efficacy in dense tumor microenvironments .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery details.
Synonyms
FGFR2; BEK; KGFR; KSAM; Fibroblast growth factor receptor 2; FGFR-2; K-sam; Keratinocyte growth factor receptor; CD antigen CD332
Target Names
Uniprot No.

Target Background

Function
Fibroblast Growth Factor Receptor 2 (FGFR2) is a tyrosine-protein kinase that serves as a cell-surface receptor for fibroblast growth factors. It plays a critical role in regulating cell proliferation, differentiation, migration, and apoptosis, and is essential for proper embryonic development. FGFR2 is involved in various developmental processes, including normal embryonic patterning, trophoblast function, limb bud development, lung morphogenesis, osteogenesis, and skin development. It also plays a crucial role in regulating osteoblast differentiation, proliferation, and apoptosis, and is necessary for normal skeletal development. FGFR2 promotes cell proliferation in keratinocytes and immature osteoblasts, but induces apoptosis in differentiated osteoblasts. It phosphorylates PLCG1, FRS2, and PAK4. Ligand binding activates several signaling cascades. Activation of PLCG1 leads to the production of diacylglycerol and inositol 1,4,5-trisphosphate, both of which are important cellular signaling molecules. Phosphorylation of FRS2 triggers the recruitment of GRB2, GAB1, PIK3R1, and SOS1, mediating the activation of RAS, MAPK1/ERK2, MAPK3/ERK1, and the MAP kinase signaling pathway, as well as the AKT1 signaling pathway. FGFR2 signaling is down-regulated through ubiquitination, internalization, and degradation. Mutations that lead to constitutive kinase activation or impair normal FGFR2 maturation, internalization, and degradation result in aberrant signaling. Overexpression of FGFR2 promotes the activation of STAT1.
Gene References Into Functions
  1. A study investigating two single nucleotide polymorphisms (SNPs), rs1219648 and rs2981582, found no association with overall breast cancer risk. However, a stratified analysis revealed a strong association between rs2981582 and premenopausal and ER-positive breast cancer in Chinese patients. PMID: 30480917
  2. The emergence of the mesenchymal FGFR2c variant in an epithelial context is believed to drive early stages of carcinogenesis. PMID: 29068468
  3. FGFR2 has been shown to be significantly overexpressed in gastric cancer tissues and correlated with a high risk of lymph node metastasis and late clinical stage. FGFR2 was negatively associated with TSP4, and FGFR2 activation was found to downregulate TSP4 expression. This downregulation plays a significant role in the proliferation, invasion, and migration of gastric cancer cells. PMID: 30355943
  4. In male breast cancer, smoking habits had a significant effect on overall survival at 10 years. In the same multivariate analysis, a significantly higher overall survival was observed in cases with the FGFR2 rs2981582 variant in the dominant transmission model. Sensitivity analysis with left truncation yielded similar results. PMID: 29709729
  5. Research demonstrated that DDX6 regulates HER2 and FGFR2 at the post-transcriptional level in gastric cancer. PMID: 29987267
  6. High FGFR2 expression has been linked to epithelial ovarian cancer cell proliferation and invasion. PMID: 29970688
  7. A wide range of FGFR2 mutations have been identified as causative for Pfeiffer syndrome. This report describes the first Chinese three-generation family with the FGFR2 mutation c.514_515delGCinsTT (p.Ala172Phe). PMID: 29782338
  8. Studies concluded that miR494 inhibited the cancer initiating cells phenotype and reversed resistance to lapatinib by inhibiting FGFR2 in HER2-positive gastric cancer. PMID: 29786108
  9. Patients exhibiting very strong FGFR2 mRNA expression demonstrated more homogeneous FGFR2 mRNA expression compared to patients with lower FGFR2 mRNA expression. PMID: 28852882
  10. A comprehensive update on FGFR2-related syndromic craniosynostosis has been provided. PMID: 29230096
  11. Mutations were not detected in any unaffected family members or unrelated control subjects. These findings expand the mutation spectrum of FGFR2 and are valuable for genetic counseling and prenatal diagnosis in patients with Crouzon syndrome. PMID: 28901406
  12. Fibroblast growth factor receptor 2 (FGFR2) splice site variants were identified in eight patients with Crouzon or Pfeiffer syndrome. PMID: 26841243
  13. Data suggests that the SOX9 transcription factor (SOX9)-fibroblast growth factor receptor 2 (FGFR2b) feed-forward loop plays a lineage-dependent role in pancreatic ductal adenocarcinoma (PDAC). PMID: 28796141
  14. This study reports, for the first time, the p.W290G mutation in patients with Pfeiffer syndrome (PS). This finding, along with clinical features, genetic analysis, computational analysis, and genotype-phenotype association studies, provides valuable insights. PMID: 28815901
  15. Research indicates that switching from fibroblast growth factor receptor 2 (FGFR-2) IIIb to IIIc variants correlates with increased cancer aggressiveness via epithelial-mesenchymal transition. PMID: 28930565
  16. These results highlight the complexity of ER regulation by FGFR2-mediated signaling, which is likely associated with breast cancer resistance to endocrine therapy. PMID: 28869838
  17. The study reports that 5-10% of epidermal nevi harbor embryonic postzygotic FGFR2 activating mutations. PMID: 27103312
  18. This study identified an FGFR2 mutation in two Chinese patients with syndromic craniosynostosis. This finding expands the reported mutation spectrum of FGFR2 and is of significant value for genetic counseling and prenatal diagnosis in families with syndromic craniosynostosis. PMID: 28849010
  19. Through a stratification analysis, 5q11.2/MAP3K1 (rs16886034, rs16886364, rs16886397, rs1017226, rs16886448) and 7q32.3/LINC-PINT (rs4593472) were associated with Luminal A, and 10q26.1/FGFR2 (rs35054928) was associated with Luminal B. PMID: 28408616
  20. SNORD126 activates the PI3K-AKT pathway through upregulation of FGFR2. PMID: 27913571
  21. This research reveals a direct binding event and characterizes the role of TRPA1 ankyrin repeats in regulating FGFR2-driven oncogenic processes, a mechanism that is hindered by miRNA-142-3p. PMID: 29038531
  22. While FGFR2 amplification is linked to poorer overall survival, it does not appear to be an independent prognostic predictor in patients with advanced gastric cancer treated with palliative fluoropyrimidine and platinum chemotherapy. PMID: 27802183
  23. This meta-analysis of case-control studies provides robust evidence that FGFR2 polymorphisms (rs2981582, rs2420946, and rs2981578) were significantly associated with breast cancer risk. PMID: 27966449
  24. The study refines the influence of variants in the FGFR2 locus on molecular characteristics of breast tumors, indicating a stronger association with estrogen receptor status among cancers without amplification of the HER2 gene. PMID: 27764800
  25. The study suggests FGFR2 as a novel acute myeloid leukemia susceptibility gene, with a haplotype TT (rs7090018 and rs2912759) showing significant association with AML. PMID: 27903959
  26. The research demonstrated that high FGFR2 expression was significantly associated with unfavorable prognosis in gastric adenocarcinoma. Additionally, SPRY2 was found to inhibit FGFR2-induced ERK phosphorylation and suppress FGFR2-elicited gastric cancer cell proliferation and invasion. PMID: 28002800
  27. Through structural (X-ray and NMR) and functional characterization of pathogenic gain-of-function mutations affecting the FGF receptor (FGFR) tyrosine kinase domain, researchers elucidated a long-distance allosteric network composed of four interconnected sites termed the 'molecular brake', 'DFG latch', 'A-loop plug', and 'alphaC tether'. The first three sites repress the kinase from adopting an active conformation... PMID: 28166054
  28. Moreover, the study found that miR-628-5p targeted and down-regulated the expression of fibroblast growth factor receptor 2 (FGFR2). FGFR2 was expressed at higher levels in ovarian cancer tissues and was correlated with worse prognosis. These findings suggest that miR-628-5p plays a critical role in ovarian cancer stem cell-driven tumorigenesis. PMID: 29229394
  29. Mandibular growth in children with FGFR2 mutations is not normal, with impairments observed in forward sagittal growth and skull base widening. PMID: 28468153
  30. Inhibition of FGF-R partially reversed alphavbeta3 integrin activity in Mll-Ell+ progenitor cells. PMID: 27340869
  31. In gastric cancer, FGFR2 protein overexpression predicts gene amplification and poor survival. PMID: 27230412
  32. Her2, cMet, and FGFR2 statuses were profiled in gastric cancer (GC) patients and the derived tumor xenograft (PDX) models. PMID: 28292264
  33. The research demonstrates that the bent bone dysplasia syndrome mutations in FGFR2 p.M391R and p.Y381D enhance FGFR2's ability to epigenetically activate rDNA. Mutations p.M391R and p.Y381D activate the p53 nucleolar stress response pathway and alter FGFR2-mediated activation of ribosome biogenesis. PMID: 28595297
  34. Polyclonal secondary FGFR2 mutations represent a significant clinical resistance mechanism to protein kinase inhibitors in patients with FGFR2 fusion-positive cholangiocarcinoma. PMID: 28034880
  35. CD44 and FGFR2 maintain stemness in gastric cancer by differentially regulating c-Myc transcription. PMID: 27107424
  36. A novel identical postzygotic activating FGFR2 mutation was identified in two unrelated fetuses with papillomatous pedunculated sebaceous naevus. PMID: 27095246
  37. Findings suggest fibroblast growth factor receptor 2 (FGFR2) as a potential therapeutic target for esophagogastric junction (EGJ) adenocarcinoma. PMID: 26933914
  38. The description of these patients expands the prenatal and postnatal findings of Bent Bone Dysplasia-FGFR2 type and contributes to the phenotypic spectrum observed in all FGFR2 disorders. PMID: 27240702
  39. The results indicate that the same FGFR2 mutations can result in diverse phenotypes. Genetic studies are recommended not only for individuals with obvious manifestations but also for family members with seemingly normal phenotypes or non-specific subtle abnormalities. PMID: 27683237
  40. Liensinine inhibits FGFR2 activity. PMID: 28132898
  41. FGFR2-ACSL5 fusion is associated with resistance to LY2874455 in FGFR2-amplified gastric cancer. PMID: 28122360
  42. FGFR2 mutation is associated with endometrial carcinoma progression and abdominopelvic metastasis. PMID: 27348297
  43. FGFR inhibitors, particularly BGJ398, are therapeutic options for cholangiocarcinoma patients harboring FGFR2-CCDC6 fusions. PMID: 27216979
  44. High FGFR2 expression is associated with Gastric Cancers. PMID: 27197184
  45. Two novel FGFR 2 gene missense mutations were identified in Chinese patients with Crouzon syndrome. PMID: 27430617
  46. The incidence of progression (progressed, recurred, or died from disease) of endometrioid endometrial cancer was significantly more prevalent (32/125, 26%) among patients with FGFR2 mutation versus wild type (120/848, 14%; p<0.001, Chi-square test). PMID: 28314589
  47. The study demonstrates that this stabilizes the tyrosine and primes it for the catalytic phosphotransfer, potentially lowering the activation barrier of the phosphotransfer reaction. This work highlights the value of incorporating dynamic information from computer simulation in deciphering RTK regulatory function. PMID: 28151998
  48. The research reveals that a decrease in PI(4,5)P2 levels under non-stimulated conditions inhibits PTEN activity, leading to aberrant activation of the oncoprotein Akt. In addition to defining a novel mechanism of Akt phosphorylation with important therapeutic implications, the study also demonstrates that differential expression levels of FGFR2, Plc11, and Grb2 correlate with patient survival. PMID: 26212011
  49. High FGFR2 expression was significantly associated with the depth of invasion, lymph-node metastasis, pathological stage, and distant metastasis or recurrent disease in gastric cancer. PMID: 28056982
  50. Fibroblast growth factor receptor 2 overexpression is correlated with decreased survival in most solid tumors, suggesting that FGFR2 expression status is a valuable prognostic biomarker and a novel therapeutic target in human solid tumors. PMID: 28618942

Show More

Hide All

Database Links

HGNC: 3689

OMIM: 101200

KEGG: hsa:2263

STRING: 9606.ENSP00000410294

UniGene: Hs.533683

Involvement In Disease
Crouzon syndrome (CS); Jackson-Weiss syndrome (JWS); Apert syndrome (APRS); Pfeiffer syndrome (PS); Beare-Stevenson cutis gyrata syndrome (BSTVS); Familial scaphocephaly syndrome (FSPC); Lacrimo-auriculo-dento-digital syndrome (LADDS); Antley-Bixler syndrome, without genital anomalies or disordered steroidogenesis (ABS2); Bent bone dysplasia syndrome (BBDS); Saethre-Chotzen syndrome (SCS)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, Fibroblast growth factor receptor subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Golgi apparatus. Cytoplasmic vesicle. Note=Detected on osteoblast plasma membrane lipid rafts. After ligand binding, the activated receptor is rapidly internalized and degraded.; [Isoform 1]: Cell membrane; Single-pass type I membrane protein. Note=After ligand binding, the activated receptor is rapidly internalized and degraded.; [Isoform 3]: Cell membrane; Single-pass type I membrane protein. Note=After ligand binding, the activated receptor is rapidly internalized and degraded.; [Isoform 8]: Secreted.; [Isoform 13]: Secreted.

Q&A

What is FGFR2 and why is it an important research target?

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.

What are the different isoforms of FGFR2 and how do they affect antibody selection?

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.

What is the optimal protocol for flow cytometry using FGFR2 Antibody, FITC conjugated?

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.

How should FGFR2 Antibody, FITC conjugated be stored to maintain optimal activity?

To maintain optimal activity of FGFR2 Antibody, FITC conjugated:

Storage ParameterRecommendationRationale
Long-term storage-20°C Preserves antibody structure and FITC conjugation
Working solution2-8°C for up to 1 monthMinimizes freeze-thaw cycles
Light exposureProtect from light FITC is photosensitive and fluorescence decreases with light exposure
Freeze-thaw cyclesAvoid repeated cyclesMultiple freeze-thaw cycles can denature antibody proteins
Shipping condition4°C with cold packsMaintains antibody integrity during transport

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.

How can dual staining with FGFR2 Antibody, FITC conjugated be optimized to study receptor-ligand interactions?

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 .

What strategies can overcome signal interference when using FGFR2 Antibody, FITC conjugated in tissues with high autofluorescence?

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:

    • Consider using FGFR2 antibodies conjugated to fluorophores in the far-red spectrum (e.g., Alexa Fluor 647)

    • Sequential immunohistochemistry with FITC quenching between rounds

    • Use of CyTOF technology for mass cytometry detection

  • 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.

How can phosphorylation-specific activation of FGFR2 be measured in conjunction with total FGFR2 using FITC-conjugated antibodies?

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:

    • Include detection of phosphorylated downstream targets like PLCG1, FRS2, and PAK4

    • Develop a hierarchical signaling profile to understand sequential activation

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.

What are the key considerations when using FGFR2 Antibody, FITC conjugated for detecting rare cell populations?

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:

    • Higher antibody concentrations may be necessary (1:50 dilution)

    • Extended incubation times (45-60 minutes at 4°C)

    • Addition of blocking reagents to reduce non-specific binding

    • Use of biotin-streptavidin amplification systems for enhanced sensitivity

  • 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.

How do differences between monoclonal and polyclonal FGFR2 Antibody, FITC conjugated affect experimental outcomes?

The choice between monoclonal and polyclonal FGFR2 antibodies significantly impacts experimental results:

CharacteristicMonoclonal FGFR2 Antibody, FITC Polyclonal FGFR2 Antibody, FITC Experimental Implications
Epitope recognitionSingle epitopeMultiple epitopesPolyclonals offer stronger signal but potentially more background
Batch-to-batch consistencyHighModerate to lowMonoclonals provide better reproducibility across experiments
Isoform specificityMay recognize specific isoformsGenerally recognizes multiple isoformsCritical for studies focusing on particular FGFR2 isoforms
Cross-reactivityMinimalPotentially higherImportant for multi-species studies
Signal strengthModerateTypically strongerInfluences detection of low-expression samples
ApplicationsExcellent for quantitative analysisBetter for detection in difficult samplesSelect based on experimental priorities

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.

What controls should be included when validating FGFR2 Antibody, FITC conjugated for a new application?

A comprehensive validation protocol for FGFR2 Antibody, FITC conjugated should include:

  • Positive and negative cell/tissue controls:

    • Positive: KATO-III human gastric carcinoma cell line (known FGFR2 expression)

    • Negative: Cell lines with confirmed absence of FGFR2 expression

    • Gradient controls: Cell lines with varying FGFR2 expression levels

  • Technical controls:

    • Isotype control: FITC-conjugated IgG from same species (rabbit or mouse)

    • Fluorescence-minus-one (FMO) controls

    • Secondary antibody-only control (for indirect methods)

    • Unstained control for autofluorescence assessment

  • 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:

    • Titration series (1:10, 1:50, 1:100, 1:200, 1:500, 1:1000)

    • Signal-to-noise ratio calculation for each dilution

    • Determination of optimal concentration (typically 1:50-1:200 for flow cytometry)

  • 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.

How can inconsistent staining patterns with FGFR2 Antibody, FITC conjugated be resolved and interpreted?

When encountering inconsistent staining patterns with FGFR2 Antibody, FITC conjugated:

  • Common causes and solutions:

    IssuePotential CauseResolution Strategy
    Weak or no signalAntibody degradationCheck storage conditions; use fresh aliquot
    Low FGFR2 expressionIncrease antibody concentration; try signal amplification
    Epitope maskingOptimize antigen retrieval; try alternative fixation
    High backgroundNon-specific bindingIncrease blocking; reduce antibody concentration
    AutofluorescenceUse appropriate quenching reagents; spectral unmixing
    FITC instabilityProtect from light; prepare fresh dilutions
    Variable cell-to-cell stainingHeterogeneous expressionSingle-cell analysis; confirm with alternative methods
    Cell cycle dependenceSynchronize 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.

How can FGFR2 Antibody, FITC conjugated be integrated into CyTOF (mass cytometry) workflows?

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:

    • Custom metal conjugation of the same FGFR2 antibody clone (e.g., clone 98739)

    • Use of commercial conjugation kits for metal attachment

    • Validation against standard FITC results to ensure comparable binding properties

  • Optimized CyTOF panel design:

    • Include FGFR2 in panels with signaling molecules like phosphorylated PLCG1, FRS2, and MAPK pathway components

    • Combine with markers for cell differentiation status

    • Include proliferation markers to correlate with FGFR2's role in cell proliferation

  • 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.

What are the methodological differences when using FGFR2 Antibody, FITC conjugated for in vivo imaging compared to in vitro applications?

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.

How can FGFR2 Antibody, FITC conjugated be used to study receptor internalization and trafficking dynamics?

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.

What methodological approaches can resolve contradictory FGFR2 expression data between antibody-based detection and mRNA analysis?

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:

    • Antibody validation with multiple clones recognizing different FGFR2 epitopes

    • Confirmation with non-antibody methods (e.g., receptor-ligand binding assays)

    • Western blot correlation with different antibody clones

    • Genetic validation using CRISPR-Cas9 knockout controls

  • 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.

What emerging technologies might replace or complement FITC conjugation for FGFR2 detection in future research?

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.

How can conflicting results from different conjugated FGFR2 antibodies (FITC vs. APC vs. Alexa Fluor 647) be systematically resolved?

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:

      • FITC: pH sensitive, moderate photostability

      • APC: Larger protein, potential steric hindrance

      • Alexa Fluor 647: Higher photostability, less pH sensitive

    • 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

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.