FGF 2 Antibody

Fibroblast Growth Factor-2, Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Definition and Mechanism of FGF2 Antibodies

FGF2 antibodies are immunoglobulins that bind specifically to FGF2, blocking its interaction with fibroblast growth factor receptors (FGFRs) or enabling its detection in biological samples. These antibodies are pivotal for studying FGF2's roles in cellular proliferation, tissue repair, and disease mechanisms .

  • Key Functions:

    • Neutralization: Inhibit FGF2 binding to FGFRs, suppressing downstream signaling pathways like MAPK and PI3K .

    • Detection: Used in assays (e.g., ELISA, Western blot) to quantify FGF2 levels in serum, plasma, or cell cultures .

Types of FGF2 Antibodies

FGF2 antibodies are classified based on their structure, specificity, and applications:

TypeExamplesApplicationsHost Species
Monoclonal (e.g., GAL-F2, bFM-2)Binds a single epitope on FGF2Neutralization, immunohistochemistry (IHC)Mouse
PolyclonalTargets multiple FGF2 epitopesWestern blot, ELISARabbit, Sheep
Conjugated (e.g., HRP, FITC)Labeled with fluorophores or enzymesFlow cytometry, imagingMouse

Table 1: Common types of FGF2 antibodies and their applications.

Cancer Biology

  • Tumor Inhibition: The monoclonal antibody GAL-F2 reduced hepatocellular carcinoma (HCC) xenograft growth by 60–80% in mice, blocking FGF2-mediated Erk1/2 phosphorylation .

  • Angiogenesis: FGF2 antibodies inhibit endothelial cell proliferation and tube formation, disrupting tumor vascularization .

Cardiovascular Studies

  • Cardioprotection: FGF2 antibodies have been used to elucidate FGF2's role in reducing myocardial infarct size by 40% in animal models .

Metabolic and Inflammatory Diseases

  • Obesity: Anti-FGF2 antibodies correlate with BMI in human adipocyte studies, suggesting a role in metabolic regulation .

  • Preeclampsia: In a rat model, FGF2 antibody treatment alleviated placental vascular dysfunction and glomerular injury .

Therapeutic Potential

  • Clinical Trials: While no FGF2 antibody has been approved for clinical use, preclinical studies highlight their promise:

    • Combination Therapy: GAL-F2 synergized with anti-VEGF antibodies or sorafenib, enhancing tumor suppression in HCC .

    • Safety: Histopathological evaluations in animal models showed minimal toxicity in vital organs .

Challenges and Future Directions

  • Epitope Specificity: Antibodies like GAL-F2 target unique conformational epitopes spanning both N- and C-terminal regions of FGF2, reducing off-target effects .

  • Cross-Reactivity: Some antibodies (e.g., GAL-F2) bind mouse FGF2, facilitating translational studies .

Product Specs

Introduction
Basic fibroblast growth factor (FGF2) belongs to the fibroblast growth factor (FGF) family. FGF family members exhibit a wide range of mitogenic and cell survival activities and play crucial roles in various biological processes, including embryonic development, cell growth, morphogenesis, tissue repair, and tumor growth and invasion. FGF2 functions as a regulator of endothelial cell migration and proliferation and acts as an angiogenic factor. It stimulates the proliferation of diverse mesoderm- and neuroectoderm-derived cells in vitro, suggesting its involvement in organogenesis. Three alternatively spliced variants encoding different FGF2 isoforms have been identified. Heparin-binding growth factors, including FGF2, exhibit angiogenic properties in vivo and act as potent mitogens for various cell types in vitro. Notably, there are variations in the tissue distribution and concentration of these growth factors.
Formulation
The antibody is supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) after reconstitution.
Shipping Conditions
The antibody is shipped in lyophilized form at ambient temperature.
Storage Procedures
For long-term storage, keep the lyophilized antibody at 4°C in a dry environment. After reconstitution, aliquot the antibody and store at -20°C if not intended for use within one month.
Solubility
To reconstitute the antibody, add sterile water (H2O) to the vial. Mix gently by swirling, ensuring the solution reaches the sides of the vial, and wait for 30-60 seconds before use.
Applications
The antibody is suitable for use in various applications, including direct enzyme-linked immunosorbent assay (ELISA), Western blotting, and immunoprecipitation.
Titer
In a direct ELISA, a 1:10,000 dilution of the antibody yields an optical density (O.D.) of 0.8 when using alkaline phosphatase-conjugated rabbit anti-mouse immunoglobulin G (IgG) secondary antibody (Jackson Laboratories).
Synonyms
Prostatropin, HBGH-2, HBGF-2, FGF-2, FGF-b.
Purification Method
Ion exchange.
Type
Mouse Anti Human Monoclonal.
Clone
NYR-hFGF-b.
Immunogen
r.Human FGF-b.
Ig Subclass
Mouse IgM.

Q&A

What is FGF2 and why is it important in research?

FGF2 (Fibroblast Growth Factor 2) is a multifunctional signaling protein with significant roles in cellular proliferation, differentiation, and angiogenesis. In humans, the canonical protein consists of 288 amino acid residues with a molecular mass of 30.8 kDa . FGF2 is primarily localized in the nucleus and is also secreted, with expression reported in granulosa and cumulus cells . As a member of the heparin-binding growth factors family, FGF2 functions as a ligand for multiple FGF receptors (FGFR1, FGFR2, FGFR3, and FGFR4), making it central to numerous developmental and pathological processes . Its involvement in cancer progression, wound healing, and tissue regeneration has established FGF2 as a critical target for research across multiple disciplines.

How do I select the appropriate anti-FGF2 antibody for my specific research application?

Selecting the optimal anti-FGF2 antibody requires consideration of several experimental parameters:

  • Detection method compatibility: Determine whether the antibody has been validated for your specific application (Western blot, IHC, ELISA, etc.) .

  • Species reactivity: Confirm cross-reactivity with your target species. Some antibodies like GAL-F2 recognize both human and mouse FGF2 with similar affinity .

  • Epitope recognition: Consider whether you need an antibody that recognizes specific isoforms or domains of FGF2 .

  • Clonality: Monoclonal antibodies like ab92337 [EP1735] offer high specificity for particular epitopes, while polyclonal antibodies provide broader recognition .

  • Validated citations: Review literature citations (over 250 for FGF2 antibodies) to verify performance in similar research contexts .

For experimental reproducibility, select antibodies with detailed validation data demonstrating specificity across multiple techniques and sample types.

What are the differences between neutralizing and non-neutralizing anti-FGF2 antibodies?

Neutralizing and non-neutralizing antibodies serve fundamentally different research functions:

Neutralizing antibodies (e.g., GAL-F2):

  • Block the biological activity of FGF2 by preventing receptor binding

  • Demonstrate IC₅₀ values of approximately 0.1 μg/ml for inhibiting FGF2-induced cell proliferation

  • Inhibit downstream signaling cascades (phosphorylation of Akt and Erk1/2)

  • Can be used for functional studies investigating FGF2's role in biological processes

  • Valuable for therapeutic research applications and mechanism studies

Non-neutralizing antibodies:

  • Bind to FGF2 without interfering with its biological function

  • Primarily useful for detection and quantification applications

  • Suitable for Western blotting, ELISA, and immunohistochemistry to determine protein expression and localization

  • Often recognize epitopes distant from the receptor-binding domains

When designing mechanistic studies, neutralizing antibodies provide insights into FGF2's functional significance, while non-neutralizing antibodies are preferable for expression analysis.

What are the optimal protocols for FGF2 detection in Western blotting?

Western blotting for FGF2 requires careful optimization due to its multiple isoforms (17-30 kDa observed bands) . An effective protocol includes:

  • Sample preparation:

    • Use 15-20 μg of whole cell lysate per lane

    • Include protease inhibitors to prevent degradation

  • Gel electrophoresis and transfer:

    • 12-15% gels are recommended for optimal resolution of FGF2 isoforms

    • Transfer to PVDF membrane (preferred over nitrocellulose for low molecular weight proteins)

  • Antibody incubation:

    • Primary antibody: Dilute anti-FGF2 antibody appropriately (e.g., ab92337 at 1/1000-1/5000 dilution)

    • Secondary antibody: HRP-conjugated anti-species antibody (e.g., goat anti-rabbit IgG-HRP at 1/20000)

  • Detection considerations:

    • Expect multiple bands between 17-30 kDa representing different FGF2 isoforms

    • The canonical full-length protein has a predicted band size of 30 kDa

    • Smaller isoforms (17-24 kDa) result from alternative translation initiation sites

For accurate interpretation, always include positive controls such as recombinant FGF2 protein and lysates from cell lines with known FGF2 expression (e.g., HeLa or K-562 cells) .

How can I optimize immunohistochemistry protocols for FGF2 detection in tissue samples?

Successful immunohistochemical detection of FGF2 requires careful protocol optimization:

  • Fixation and processing:

    • Formalin/PFA-fixed paraffin-embedded sections are standard

    • Section thickness of 3-5 μm is optimal for penetration and resolution

  • Antigen retrieval:

    • Critical for unmasking epitopes in fixed tissues

    • Heat-mediated antigen retrieval in citrate buffer (pH 6.0) at 95°C for 15 minutes

    • An EZ antigen retriever system or similar device ensures consistent heating

  • Blocking and antibody incubation:

    • Block with power block reagent or 2% BSA to reduce non-specific binding

    • Primary antibody: Incubate with anti-FGF2 at optimized dilution (e.g., ab92337 at 1/1000)

    • Secondary detection: Use super sensitive polymer-HRP IHC detection system for enhanced sensitivity

  • Controls and counterstaining:

    • Include negative controls using PBS instead of primary antibody

    • Use hematoxylin as a counterstain to visualize tissue architecture

    • Include positive control tissues with known FGF2 expression (prostate or cerebrum tissue)

For multi-labeling studies, consider sequential detection with different chromogens or fluorescent tags to evaluate co-localization with other proteins of interest.

What is the most sensitive approach for detecting FGF2 using ELISA methodologies?

ELISA offers high sensitivity for FGF2 quantification with detection limits in the picogram range. An optimized sandwich ELISA protocol includes:

  • Capture antibody coating:

    • Coat wells with goat anti-mouse IgG (for mouse monoclonal capture antibodies) or directly with purified anti-FGF2 antibody

    • Alternative approach: Coat with 50 μg/ml heparin to capture FGF2 through its heparin-binding domain

  • Blocking and sample addition:

    • Block with 2% BSA to reduce non-specific binding

    • Add samples and standards (dilution range from 1pg/ml to 1μg/ml for standard curve)

  • Detection system:

    • Primary detection: Use biotinylated or directly labeled anti-FGF2 antibody targeting a different epitope than the capture antibody

    • Secondary detection: HRP-streptavidin for biotinylated antibodies or direct HRP detection

    • Substrate: TMB (3,3',5,5'-tetramethylbenzidine) provides sensitive colorimetric detection

  • Validation and optimization:

    • Validate with recombinant FGF2 protein standards (e.g., ab9596)

    • Optimize antibody pairs: For example, mouse monoclonal [AS24] as capture antibody (1μg/ml) and rabbit monoclonal [EP1735] as detector antibody (0.5μg/ml)

This approach yields a sensitive and specific assay with minimal cross-reactivity to other FGF family members, providing accurate quantification of FGF2 in biological samples.

How should I design experiments to evaluate FGF2's role in angiogenesis?

Designing robust experiments to investigate FGF2's angiogenic functions requires multi-faceted approaches:

  • In vitro endothelial cell assays:

    • HUVEC proliferation assay: Measure cell proliferation in response to FGF2 stimulation (0.1-10 ng/ml) with and without neutralizing antibodies

    • Tube formation assay: Quantify endothelial network formation on Matrigel with FGF2 stimulation

    • Migration/wound healing assays: Assess endothelial cell migration in response to FGF2 gradients

  • Signaling pathway analysis:

    • Western blot analysis of downstream effectors: Measure phosphorylation of Akt and Erk1/2 following FGF2 treatment

    • Inhibition studies: Use neutralizing antibodies like GAL-F2 at different concentrations (0.1-1 μg/ml) to block signaling

    • Time-course experiments: Determine temporal activation patterns of signaling molecules

  • In vivo angiogenesis models:

    • Matrigel plug assay: Inject FGF2-containing Matrigel subcutaneously and analyze vessel infiltration

    • Tumor xenograft models: Evaluate microvessel density in tumors treated with anti-FGF2 antibodies

    • Zebrafish angiogenesis models: Visualize vessel formation in transparent embryos

  • Controls and validation:

    • Positive controls: VEGF-induced angiogenesis

    • Negative controls: Isotype-matched antibodies without FGF2 specificity

    • Dose-response relationships: Test multiple concentrations of FGF2 and neutralizing antibodies

This multi-tiered approach provides comprehensive assessment of FGF2's angiogenic properties and the efficacy of anti-FGF2 interventions.

What controls should be included when characterizing new anti-FGF2 antibodies?

Thorough characterization of new anti-FGF2 antibodies requires comprehensive controls:

  • Specificity controls:

    • Cross-reactivity testing with related FGF family members (especially FGF1)

    • Testing with chimeric FGF1/FGF2 proteins to define epitope specificity

    • Alanine scanning mutagenesis to identify critical binding residues

    • Competition assays with other anti-FGF2 antibodies to determine epitope uniqueness

  • Functional validation controls:

    • Receptor binding inhibition assays for each FGFR (FGFR1-4)

    • Cell proliferation assays with dose-response curves (IC₅₀ determination)

    • Downstream signaling inhibition (phosphorylation of Akt and Erk1/2)

    • Comparison with established anti-FGF2 antibodies (e.g., bFM-1, FB-8, 3H3)

  • Species cross-reactivity controls:

    • Testing with FGF2 from multiple species (human, mouse, rat, etc.)

    • Cellular assays using cells from different species

  • Application-specific controls:

    • For Western blotting: Recombinant FGF2, positive cell lysates (HeLa, K-562)

    • For IHC: Tissue with known FGF2 expression, negative control with primary antibody omitted

    • For ELISA: Standard curves with recombinant protein, spike-recovery experiments

These controls ensure that new antibodies are properly characterized before deployment in critical research applications.

How can I design experiments to determine if an anti-FGF2 antibody inhibits specific FGF2-FGFR interactions?

To assess inhibition of specific FGF2-FGFR interactions, implement the following experimental design:

  • ELISA-based receptor binding inhibition assay:

    • Coat plates with goat anti-human IgG-Fc

    • Capture FGFR1IIIc-Fc, FGFR2IIIc-Fc, FGFR3IIIc-Fc, or FGFR4-Fc (0.5 μg/ml)

    • Test increasing concentrations of anti-FGF2 antibody with Flag-FGF2 (0.1 μg/ml)

    • Detect bound Flag-FGF2 with HRP-anti-Flag antibody

    • Calculate IC₅₀ values for inhibition of binding to each receptor

  • Cell-based receptor competition assays:

    • Use cells expressing individual FGFRs (transfected or naturally expressing)

    • Employ fluorescently-labeled FGF2 and flow cytometry to measure binding

    • Assess antibody-mediated inhibition of binding at various concentrations

    • Compare inhibition profiles across different FGFR-expressing cell lines

  • Surface Plasmon Resonance (SPR) analysis:

    • Immobilize FGFRs on sensor chips

    • Measure binding kinetics of FGF2 with and without antibody pre-incubation

    • Calculate association/dissociation rates and equilibrium constants

    • Compare inhibition efficiency across different FGFRs

  • Functional validation in receptor-specific cellular models:

    • Engineer cells to express individual FGFRs

    • Measure receptor activation (phosphorylation) upon FGF2 stimulation

    • Assess antibody-mediated inhibition of receptor activation

    • Correlate with downstream signaling inhibition (Akt, Erk1/2 phosphorylation)

This approach provides comprehensive characterization of receptor-specific inhibition profiles for anti-FGF2 antibodies.

Why might I observe multiple bands when using anti-FGF2 antibodies in Western blotting?

Multiple FGF2 bands in Western blotting have specific biological explanations:

  • Isoform variation:

    • FGF2 exists in multiple isoforms resulting from alternative translation initiation sites

    • Observed band sizes typically range from 17-30 kDa

    • The low molecular weight isoform (18 kDa) is secreted

    • Higher molecular weight isoforms (22, 24, and 30 kDa) predominantly localize to the nucleus

  • Cell type-specific expression patterns:

    • Different cell lines express varying isoform profiles

    • HeLa cells show multiple isoforms between 17-30 kDa

    • K-562 cells display a distinct banding pattern

    • Compare your results with validated cell line data to interpret correctly

  • Post-translational modifications:

    • Phosphorylation increases apparent molecular weight

    • Other modifications may alter migration patterns

    • Consider phosphatase treatment to confirm modification-dependent shifts

  • Antibody-specific recognition patterns:

    • Some antibodies like ab92337 [EP1735] recognize three distinct FGF2 isoforms

    • Epitope location may affect detection of specific isoforms

    • Compare results with different antibodies targeting distinct epitopes

Understanding these patterns is crucial for accurate interpretation of FGF2 expression studies, especially when comparing normal and pathological tissues where isoform ratios may shift.

How can I resolve non-specific background in FGF2 immunohistochemistry?

High background in FGF2 immunohistochemistry can be systematically addressed:

  • Optimize blocking conditions:

    • Increase blocking duration (30-60 minutes) with power block reagent

    • Add 0.1-0.3% Triton X-100 to blocking buffer for better penetration

    • Consider protein-free blockers if protein-based blockers cause issues

  • Modify antigen retrieval parameters:

    • Adjust citrate buffer pH (try pH 6.0)

    • Optimize heating time (15 minutes is standard)

    • Allow gradual cooling to room temperature after retrieval

    • Test alternative retrieval buffers (EDTA, Tris-EDTA)

  • Antibody optimization:

    • Titrate primary antibody concentration (start with 1/1000 dilution for ab92337)

    • Increase washing steps (5-6 washes of 5 minutes each)

    • Extend primary antibody incubation to overnight at 4°C

    • Test different detection systems (polymer-HRP versus avidin-biotin)

  • Reduce endogenous enzyme activity:

    • Block endogenous peroxidase with 3% H₂O₂ in methanol for 30 minutes

    • For fluorescent detection, include additional blocking steps for autofluorescence

    • Use levamisole to block endogenous alkaline phosphatase

  • Tissue-specific considerations:

    • Optimize fixation times for different tissues

    • Consider shorter fixation periods for sensitive epitopes

    • Include negative controls using PBS instead of primary antibody

These systematic approaches should significantly reduce background while preserving specific FGF2 signal.

How do I interpret contradictory results from different anti-FGF2 antibodies targeting the same sample?

Contradictory results between anti-FGF2 antibodies require systematic analysis:

  • Epitope-specific recognition differences:

    • Different antibodies recognize distinct epitopes that may be differentially accessible

    • GAL-F2 recognizes a unique conformational epitope compared to other antibodies (bFM-1, FB-8, 3H3)

    • Some epitopes may be masked by protein-protein interactions or post-translational modifications

    • Map the epitopes of each antibody using chimeric proteins or alanine scanning mutagenesis

  • Antibody validation status comparison:

    • Evaluate the validation data for each antibody (Western blot, IHC, ELISA)

    • Consider antibodies with multiple validated applications as more reliable

    • Review literature citations for similar experimental contexts

    • Test antibodies against recombinant FGF2 protein standard

  • Technical considerations:

    • Confirm optimal working conditions for each antibody

    • Some antibodies perform better in native conditions versus denatured (Western blot)

    • Competitive binding assays can determine if antibodies interfere with each other

    • Consider sequential staining with different antibodies to verify co-localization

  • Biological interpretation:

    • Different results might reveal biologically significant conformational states

    • Create a table comparing results across antibodies, applications, and conditions

    • Consider the possibility that each antibody reveals different aspects of FGF2 biology

This analytical approach transforms contradictory results into potentially valuable insights about FGF2 structural or functional states.

How can neutralizing FGF2 antibodies be utilized in cancer research models?

Neutralizing FGF2 antibodies offer powerful tools for cancer research:

  • Tumor xenograft growth inhibition studies:

    • GAL-F2 administered at 5 mg/kg i.p. twice weekly potently inhibits growth of HCC xenografts (SMMC-7721, HEP-G2, SK-HEP-1)

    • Combination treatments with anti-VEGF antibodies or sorafenib show strong additive effects

    • Measurement parameters include tumor volume, weight, and time to progression

  • Anti-angiogenic mechanism studies:

    • Neutralizing antibodies block angiogenesis in tumor models

    • Histological analysis of microvessel density in treated tumors

    • Immunohistochemical assessment of endothelial markers (CD31, CD34)

  • Signaling pathway analysis in tumor models:

    • FGF2 antibodies inhibit downstream signaling in xenografts

    • Western blot analysis of tumor lysates for phosphorylated Akt and Erk1/2

    • Immunohistochemical staining for pathway activation markers

  • Biomarker development applications:

    • FGF2 and FGFR-2 expression as potential biomarkers of malignant transformation

    • Correlation of FGF2 expression patterns with treatment response

    • Development of companion diagnostics for anti-FGF2 therapies

  • Therapeutic antibody development:

    • Preclinical evaluation of humanized versions of antibodies like GAL-F2

    • Pharmacokinetic and pharmacodynamic studies

    • Toxicity assessment in animal models

These applications provide crucial preclinical data supporting the potential development of anti-FGF2 antibody therapies for cancer treatment.

What methodologies can determine the mechanisms of FGF2 signaling inhibition by neutralizing antibodies?

Elucidating FGF2 signaling inhibition mechanisms requires multi-level analysis:

  • Receptor binding inhibition characterization:

    • ELISA-based assays to measure inhibition of FGF2 binding to each FGFR (FGFR1-4)

    • Determine IC₅₀ values for binding inhibition to each receptor subtype

    • Assess differences in inhibition efficiency across receptor isoforms (IIIb vs. IIIc)

  • Signaling cascade analysis:

    • Western blot assessment of Akt and Erk1/2 phosphorylation inhibition

    • Time-course experiments to determine signaling kinetics

    • Dose-response relationships between antibody concentration and signaling inhibition

    • Comparison with small molecule FGFR inhibitors to identify pathway-specific effects

  • Transcriptional response profiling:

    • RNA-seq analysis of cells treated with FGF2 ± neutralizing antibodies

    • Identification of FGF2-responsive gene networks affected by antibody treatment

    • Validation of key transcriptional targets by qRT-PCR and protein analysis

  • Functional cellular responses:

    • Quantification of proliferation inhibition (e.g., IC₅₀ of 0.1 μg/ml for HUVEC)

    • Analysis of migration, invasion, and differentiation responses

    • Cell cycle analysis to determine stage-specific effects

    • Apoptosis assays to evaluate cell survival impact

These methodologies provide comprehensive mechanistic insights into how neutralizing antibodies disrupt FGF2 signaling at multiple biological levels.

How can FGF2 antibodies be used to identify biomarkers for malignant transformation?

FGF2 antibodies enable sophisticated biomarker identification approaches:

  • Expression profiling in disease progression:

    • Immunohistochemical analysis of FGF2 and FGFRs across disease stages

    • FGF2 and FGFR-2 appear useful as biomarkers of malignant transformation in oral submucous fibrosis and leukoplakia patients

    • Quantitative assessment using tissue microarrays and digital pathology

    • Correlation with clinical outcomes and disease progression

  • Multi-marker panel development:

    • Combined analysis of FGF2 with other signaling molecules

    • Co-expression studies with FGFRs using dual immunostaining

    • Integration with other established cancer biomarkers

    • Statistical modeling to develop predictive biomarker signatures

  • Functional biomarker assessment:

    • Evaluation of secreted FGF2 in patient biofluids (serum, plasma)

    • Development of sensitive ELISA protocols using optimal antibody pairs

    • Longitudinal monitoring of FGF2 levels during treatment

    • Correlation of FGF2 levels with response to anti-angiogenic therapies

  • Molecular imaging applications:

    • Development of labeled FGF2 antibodies for in vivo imaging

    • Correlation of imaging findings with tissue expression patterns

    • Monitoring of treatment response using molecular imaging

This multifaceted approach enables identification of clinically relevant FGF2-related biomarkers for early detection, prognosis, and treatment response prediction.

How do FGF2 antibodies help elucidate cross-talk between FGF and other growth factor pathways?

FGF2 antibodies provide powerful tools for dissecting pathway cross-talk:

  • Co-inhibition studies:

    • Combined treatment with FGF2 and VEGF antibodies shows additive effects in tumor xenograft models

    • This reveals functional interaction between angiogenic pathways

    • Systematic analysis of different antibody combinations uncovers pathway hierarchies

    • Measurement of tumor growth inhibition compared to single-agent treatment

  • Signaling node analysis:

    • Western blot analysis of common downstream effectors (Akt, Erk1/2)

    • Time-course experiments after selective pathway inhibition

    • Phosphoproteomic analysis to identify shared signaling nodes

    • Quantitative comparison of inhibition patterns between single and combined treatments

  • Receptor complex formation studies:

    • Immunoprecipitation with FGF2 antibodies followed by analysis of co-precipitated receptors

    • Detection of heterodimeric receptor complexes

    • Assessment of ligand-dependent complex formation

    • Identification of co-receptor requirements for different pathways

  • Transcriptional response analysis:

    • Gene expression profiling after selective pathway inhibition

    • Identification of shared and distinct transcriptional targets

    • Validation of cross-talk-dependent gene regulation

    • Bioinformatic analysis of transcription factor binding sites

These approaches provide mechanistic insights into how the FGF2 pathway interacts with other signaling systems, informing combination treatment strategies for cancer and other diseases.

What techniques can distinguish between the functions of different FGF2 isoforms?

Differentiating the biological roles of FGF2 isoforms requires specialized approaches:

  • Isoform-specific detection strategies:

    • Western blotting with antibodies that recognize multiple isoforms (17-30 kDa)

    • Densitometric quantification of isoform ratio changes

    • Subcellular fractionation to separate nuclear high-molecular-weight isoforms from cytoplasmic/secreted low-molecular-weight isoforms

    • Isoform-specific qRT-PCR to measure differential expression

  • Functional dissection approaches:

    • Expression of individual isoforms in cellular models

    • CRISPR-based targeting of specific translation initiation sites

    • Rescue experiments with isoform-specific expression constructs

    • Isoform-selective antibody neutralization (when available)

  • Localization analysis:

    • Confocal microscopy to determine subcellular distribution patterns

    • Co-localization studies with compartment markers

    • Live-cell imaging with fluorescently tagged isoforms

    • Correlation of localization patterns with functional outcomes

  • Clinical correlation studies:

    • Analysis of isoform expression patterns in different disease states

    • Correlation of isoform ratios with disease progression

    • Development of diagnostic approaches based on isoform profiles

    • Therapeutic targeting strategies based on isoform-specific functions

These methodologies enable researchers to distinguish the unique biological roles of different FGF2 isoforms in normal physiology and disease.

What is the current understanding of FGF2 post-translational modifications and their functional significance?

FGF2 undergoes several post-translational modifications with significant functional implications:

  • Phosphorylation:

    • FGF2 is subject to phosphorylation at multiple sites

    • Detection methods:

      • Phospho-specific antibodies in Western blotting

      • Mass spectrometry for comprehensive phosphosite mapping

      • Phosphatase treatment to confirm modification-dependent mobility shifts

    • Functional significance:

      • Regulation of nuclear transport

      • Modulation of receptor binding affinity

      • Altered protein stability and turnover

  • Glycosylation:

    • Analysis approaches:

      • Glycosidase treatment followed by Western blotting

      • Lectin binding assays to characterize glycan structures

      • Mass spectrometry for glycan profiling

    • Functional implications:

      • Secretion efficiency

      • Extracellular matrix interactions

      • Receptor binding specificity

  • Proteolytic processing:

    • Detection methods:

      • N-terminal sequencing of purified protein

      • Antibodies recognizing different domains

      • In vitro protease sensitivity assays

    • Biological significance:

      • Generation of bioactive fragments

      • Altered tissue distribution

      • Modified receptor binding properties

  • Other modifications:

    • Methylation, acetylation, and SUMOylation have been reported

    • These modifications primarily affect nuclear functions

    • Detection requires specific antibodies or mass spectrometry approaches

Understanding these modifications provides crucial insights into the regulation of FGF2's diverse biological activities and may reveal new therapeutic opportunities.

Product Science Overview

Introduction

Fibroblast Growth Factor-2 (FGF-2), also known as basic fibroblast growth factor (bFGF), is a member of the fibroblast growth factor family. This family consists of proteins involved in various biological processes, including cell growth, development, and tissue repair. FGF-2 is particularly notable for its role in angiogenesis, wound healing, and embryonic development.

Structure and Function

FGF-2 is a heparin-binding protein that exists in multiple isoforms due to alternative translation initiation sites. These isoforms vary in their molecular weight and cellular localization. The low molecular weight (18 kDa) isoform is predominantly found in the cytoplasm and extracellular matrix, while the high molecular weight isoforms (22-34 kDa) are primarily nuclear.

FGF-2 exerts its biological effects by binding to specific cell surface receptors known as fibroblast growth factor receptors (FGFRs). This binding triggers a cascade of downstream signaling pathways, including the MAPK/ERK, PI3K/Akt, and PLCγ pathways, which ultimately lead to cellular proliferation, differentiation, and survival.

Role in Development and Disease

FGF-2 plays a crucial role in various physiological and pathological processes:

  • Angiogenesis: FGF-2 is a potent angiogenic factor that promotes the formation of new blood vessels from pre-existing ones. This process is essential for wound healing and tissue regeneration.
  • Wound Healing: FGF-2 accelerates wound healing by stimulating the proliferation and migration of fibroblasts and endothelial cells.
  • Embryonic Development: FGF-2 is involved in the development of various tissues and organs during embryogenesis, including the nervous system, skeletal system, and cardiovascular system.
  • Cancer: Aberrant expression of FGF-2 has been implicated in tumorigenesis. FGF-2 promotes cancer cell proliferation, invasion, and metastasis by enhancing angiogenesis and modulating the tumor microenvironment .
Mouse Anti-Human FGF-2 Antibody

Mouse anti-human FGF-2 antibodies are monoclonal antibodies produced by immunizing mice with human FGF-2. These antibodies are highly specific and are used in various research and diagnostic applications, including:

  • Western Blotting: Detecting FGF-2 protein levels in cell and tissue lysates.
  • Immunohistochemistry: Localizing FGF-2 in tissue sections.
  • ELISA: Quantifying FGF-2 levels in biological samples.
  • Flow Cytometry: Analyzing FGF-2 expression on the cell surface.

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.