FGF 1 Human, Sf9

Fibroblast Growth Factor-Acidic Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Biological Functions

FGF-1 acts as a pleiotropic growth factor with the following activities:

  • Mitogenesis: Stimulates proliferation of endothelial, mesoderm-, and neuroectoderm-derived cells .

  • Angiogenesis: Promotes blood vessel formation via endothelial cell migration and tube structure organization .

  • Neuroprotection: Enhances neuronal survival and regeneration, particularly in brain development and injury repair .

  • Wound Healing: Accelerates tissue repair by activating keratinocytes and fibroblasts .

Mechanistically, FGF-1 binds fibroblast growth factor receptor 1 (FGFR1) and integrin αvβ3, triggering downstream pathways like MAPK/ERK and PI3K/AKT . Heparan sulfate is required for receptor dimerization and signal transduction .

Production and Quality Control

FGF-1 Human, Sf9 is synthesized using baculovirus-infected Sf9 cells, followed by chromatographic purification . Quality metrics include:

ParameterSpecification
Endotoxin Levels<0.1 ng/µg
Biological ActivityED50 <10 ng/ml (BAF3 cell proliferation assay)
StabilityStable at -20°C to -80°C with carrier proteins (e.g., 0.1% HSA/BSA)

Research Applications

FGF-1 Human, Sf9 is widely used in:

  • Neuroscience Studies: Promotes axonal regeneration in spinal cord injury models .

  • Cancer Research: Investigated for its role in tumor angiogenesis and epithelial-mesenchymal transition .

  • Developmental Biology: Explored in organogenesis and limb development .

  • Therapeutic Development: Preclinical trials for ischemic heart disease and diabetic ulcers .

Notably, FGF-1’s ability to cross the blood-brain barrier makes it a candidate for neurodegenerative disease therapies .

Clinical and Pathological Relevance

Dysregulation of FGF-1 is linked to:

  • Neurodegeneration: Reduced levels correlate with neuronal loss in Alzheimer’s disease .

  • Polycystic Kidney Disease: Drives hyperproliferation of renal fibroblasts .

  • Metabolic Disorders: Associated with overnutrition and insulin resistance .

Comparative Insights

FGF-1 differs from other FGFs in secretion mechanisms and receptor-binding dynamics:

FeatureFGF-1 (Acidic)FGF-2 (Basic)
Isoelectric PointAcidic (~5.6)Basic (~9.6)
Secretion PathwayNon-classical Heparin-dependent
Tissue DistributionUbiquitous, intracellularExtracellular matrix

Product Specs

Introduction
Acidic fibroblast growth factor (aFGF or FGF-1) is part of the fibroblast growth factor (FGF) family. FGFs play roles in cell growth, survival, and various biological processes like embryonic development, tissue repair, and tumor progression. FGF-1 specifically influences endothelial cell behavior (migration and proliferation) and acts as an angiogenic factor. It promotes the growth of various cell types derived from mesoderm and neuroectoderm, suggesting its involvement in organ development. Different isoforms exist due to alternative splicing. Heparin-binding growth factors, including FGF-1, are known for their angiogenic properties and ability to stimulate the growth of various cell types in laboratory settings. The distribution and concentration of these growth factors can vary among tissues.
Description
Recombinant Human Fibroblast Growth Factor-1 (FGF-1) is produced in Sf9 insect cells. It is a single glycosylated polypeptide chain with 140 amino acids and a molecular weight of 15.8 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, clear and colorless solution.
Formulation
The protein solution is provided at a concentration of 1.8 mg/ml in a buffer consisting of 20mM Tris HCl (pH 7.9), 100mM KCl, 0.2mM DTT, and 20% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is greater than 95% as determined by reverse-phase high-performance liquid chromatography (RP-HPLC) and SDS-polyacrylamide gel electrophoresis (SDS-PAGE).
Biological Activity
The biological activity of FGF-1 is measured by its ability to stimulate the proliferation of BAF3 cells expressing FGF receptors. This is determined by measuring the incorporation of radioactive thymidine (3H-thymidine) into the DNA of proliferating cells. The ED50, which represents the concentration of FGF-1 required to achieve half-maximal proliferation, is less than 10 ng/ml. This corresponds to a specific activity of 100,000 IU/mg.
Synonyms
HBGF-1, ECGF-beta, FIBP, FGFIBP, FIBP-1, ECGF, ECGFA, GLIO703, FGF1, FGF-a.
Source
Baculovirus.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids as determined and was found to be Met-Phe-Asn-Leu-Pro.

Q&A

What is human FGF-1 and what are its primary biological functions?

Human FGF-1, also known as acidic fibroblast growth factor (aFGF), endothelial cell growth factor (ECGF), or heparin-binding growth factor 1 (HBGF-1), is a 15.9 kDa secreted protein that functions as a potent mitogen and differentiation factor . FGF-1 stimulates growth and differentiation of endothelial and epithelial cells and plays a crucial role in the development of organoids . Additionally, it maintains oligodendrocytes and astroglia as well as bone marrow-derived mesenchymal and hematopoietic stem cells .

FGF-1 mediates its biological effects by binding to and activating cell-surface fibroblast growth factor receptors (FGFRs), which are transmembrane tyrosine kinases. This binding initiates several signaling cascades, including MAPK1/ERK2, MAPK3/ERK1, and the AKT1 pathways . Unlike other FGF family members, FGF-1 can bind and activate all FGF receptor isoforms, making it a universal FGF ligand.

Why are Sf9 insect cells used for recombinant human FGF-1 production?

Sf9 cells derived from Spodoptera frugiperda (fall armyworm) provide several advantages for recombinant human FGF-1 production compared to other expression systems:

  • High protein yields: The baculovirus expression system in Sf9 cells typically produces significantly higher quantities of recombinant protein compared to mammalian cell systems.

  • Proper protein folding: Sf9 cells contain the necessary chaperones and enzymes to ensure correct folding of complex human proteins.

  • Post-translational modifications: While not identical to mammalian systems, Sf9 cells can perform many post-translational modifications necessary for protein function.

  • Scaling potential: The system can be easily scaled from small research quantities to larger production volumes.

  • Simplified purification: Proteins expressed in Sf9 cells generally require less complex purification strategies than those from bacterial systems.

For FGF-1 specifically, Sf9 expression produces protein with high bioactivity and structural integrity, making it suitable for most research applications where absolute mammalian-identical glycosylation is not critical.

How does Sf9-produced human FGF-1 compare structurally to native human FGF-1?

Recombinant human FGF-1 produced in Sf9 cells closely resembles native human FGF-1 in primary structure and core functional properties. The protein has a molecular weight of approximately 15.9 kDa and migrates as a major band at this position on SDS-PAGE under both reduced and non-reduced conditions .

Key structural comparisons include:

  • Amino acid sequence: Identical to native human FGF-1, with proper N-terminal and C-terminal processing.

  • Disulfide bonds: Correctly formed, maintaining proper tertiary structure.

  • Glycosylation: Differs from native human FGF-1, as Sf9 cells produce simpler, high-mannose type glycans rather than complex mammalian glycosylation patterns.

  • Bioactivity: Despite glycosylation differences, Sf9-produced FGF-1 demonstrates comparable bioactivity to native FGF-1, with EC50 values of approximately 0.81 ng/ml (51 pM) in standard assays .

  • Heparin binding: Retains strong affinity for heparin, an essential characteristic for proper FGFR activation.

These properties make Sf9-produced human FGF-1 suitable for most research applications, particularly where protein yield and cost-effectiveness are prioritized over exact replication of mammalian glycosylation patterns.

What are optimal protocols for measuring FGF-1 bioactivity in research settings?

FGF-1 bioactivity can be reliably assessed through multiple complementary approaches:

  • Luciferase reporter assay: The gold standard for quantitative assessment involves HEK293T cells expressing an FGF-responsive element driving luciferase expression. Cells are treated with serial dilutions of FGF-1 for 3 hours, followed by measurement of firefly luciferase activity normalized to control Renilla luciferase . This typically yields an EC50 of approximately 0.81 ng/ml (51 pM) for high-quality FGF-1 preparations.

  • Proliferation assays: Several cell types respond to FGF-1 with increased proliferation, including:

    • BaF3 cells engineered to express FGFRs

    • NIH-3T3 fibroblasts

    • Human umbilical vein endothelial cells (HUVECs)

    Proliferation can be measured using colorimetric assays (MTT/XTT), BrdU incorporation, or direct cell counting over 24-72 hour periods.

  • Phosphorylation analysis: Western blotting for phosphorylated ERK1/2, AKT, or PLCγ provides a rapid readout of FGF-1 signaling activity, typically performed 5-30 minutes after stimulation.

  • Migration assays: Endothelial cells or fibroblasts demonstrate chemotactic responses to FGF-1 gradients, measurable via transwell migration assays or wound healing assays.

  • Branching morphogenesis: In three-dimensional culture systems like Matrigel, FGF-1 can induce budding and branching of epithelial structures, providing a functional readout of developmental activity .

When designing bioactivity assays, it's essential to include heparin (1-5 μg/ml) to stabilize FGF-1 and enhance receptor binding, and to incorporate appropriate positive and negative controls to validate assay performance.

How can researchers effectively troubleshoot inconsistent results in FGF-1 experiments?

When confronting variability in FGF-1 experimental outcomes, a systematic troubleshooting approach should address:

  • Protein quality issues:

    • Verify purity via SDS-PAGE (should show a single major band at 15.9 kDa)

    • Confirm bioactivity using validated reporter assays (EC50 ≈ 0.81 ng/ml)

    • Check storage conditions (avoid repeated freeze-thaw cycles)

    • Use carrier protein (0.1% BSA) to prevent surface adsorption

  • Receptor expression variability:

    • Verify FGFR expression levels in your cell model

    • Consider natural variation in FGFR isoform expression between cell passages

    • Remember that FGF-1 activates multiple FGFR subtypes, potentially complicating interpretation

  • Heparin and heparan sulfate proteoglycan effects:

    • Standardize heparin concentrations (1-5 μg/ml is typically optimal)

    • Consider that endogenous HSPG expression varies between cell types

    • Include heparin-free controls to assess dependence

  • Experimental design factors:

    • Use serum starvation to synchronize cells before FGF-1 treatment

    • Control cell density and passage number

    • Perform time-course and dose-response studies rather than single-point measurements

    • Include both positive controls (e.g., FGF-2) and negative controls (heat-inactivated FGF-1)

  • Technical considerations:

    • Increase biological replicates (n≥3)

    • Document all reagent sources and lot numbers

    • Ensure statistical methods are appropriate for data distribution

When troubleshooting, changing only one variable at a time will help identify the source of inconsistency while maintaining experimental control.

How do receptor specificity profiles differ between FGF-1 and other FGF family members?

Human FGF-1 possesses unique receptor binding characteristics compared to other FGF family members:

  • Universal receptor activation: Unlike most FGFs that show restricted receptor specificity, FGF-1 can bind and activate all seven principal FGF receptors (FGFR1b, FGFR1c, FGFR2b, FGFR2c, FGFR3b, FGFR3c, and FGFR4) . This contrasts with FGF-7, for example, which binds almost exclusively to FGFR2b.

  • Relative binding affinities: Despite its broad specificity, FGF-1 exhibits varying affinities for different receptors, generally showing strongest binding to FGFR1c and FGFR2c isoforms.

  • Isoform preferences: FGF-1 typically shows stronger binding to "c" isoforms (expressed predominantly in mesenchymal tissues) than "b" isoforms (expressed in epithelial tissues) of the same receptor.

  • Co-receptor requirements: Like all FGFs, FGF-1 requires heparan sulfate proteoglycans (HSPGs) as co-receptors, but the specific HSPG structural requirements may differ between FGF family members.

  • Functional redundancy: In developmental contexts, FGF-1 can often compensate for the absence of more receptor-restricted FGFs, but the opposite is not always true.

This broad receptor binding profile makes FGF-1 versatile for experimental applications but also creates challenges in interpreting receptor-specific effects. When receptor specificity is critical, researchers should consider using more selective FGF ligands or receptor-specific blocking antibodies alongside FGF-1.

How does FGF-1 induce epithelial branching, and how can this be modeled experimentally?

FGF-1 plays significant roles in epithelial branching morphogenesis, which can be experimentally modeled through several approaches:

  • Mechanism of FGF-induced branching:
    FGF-1 induces branching through coordinated effects on cell proliferation, migration, and differentiation. When applied to embryonic lung tissue (endoderm plus mesenchyme) in culture, FGF-1 elicits a specific sequence of morphological changes: initial cyst-like expansion of the endoderm within 24 hours, followed by extensive budding after 48-60 hours . This response involves increased endodermal cell proliferation rates.

  • 3D culture models:

    • Matrigel culture system: When isolated lung endoderm is embedded in Matrigel and treated with FGF-1, it undergoes immediate budding, demonstrating direct morphogenic effects without mesenchymal interactions .

    • Collagen gel culture: In collagen matrices, FGF-1 induces initial cystic expansion but not subsequent budding, highlighting the matrix-dependency of the branching response .

  • Comparison with other FGFs:

    • FGF-1: Elicits immediate endodermal budding in Matrigel culture

    • FGF-7: Induces expansion of endoderm but never progresses to bud formation

    • FGF-10: Initially causes expansion followed by multiple bud formation within 24 hours

  • Experimental considerations:

    • Optimal FGF-1 concentration typically ranges from 50-200 ng/ml for branching assays

    • Heparin supplementation (1-5 μg/ml) enhances FGF-1 activity

    • Matrix composition significantly influences morphogenic outcomes

    • Time-lapse imaging captures the dynamic nature of the branching process

These experimental models provide valuable insights into the molecular mechanisms of epithelial branching morphogenesis and allow for testing of factors that modulate this fundamental developmental process.

How can researchers distinguish between direct FGF-1 effects and secondary signaling cascade effects?

Differentiating between primary FGF-1 responses and secondary effects requires methodical experimental approaches:

  • Temporal analysis:

    • Immediate responses (5-30 minutes): Receptor phosphorylation, ERK/MAPK activation, and Ca²⁺ flux represent direct FGF-1 effects

    • Intermediate responses (1-4 hours): Transcriptional changes of immediate early genes

    • Delayed responses (>4 hours): Secondary signaling cascades, including effects mediated by induced autocrine/paracrine factors

  • Inhibitor strategies:

    • FGFR-specific tyrosine kinase inhibitors (e.g., PD173074) block all FGF-1 effects

    • Pathway-specific inhibitors help pinpoint primary signaling cascades:

      • U0126 (MEK/ERK pathway)

      • LY294002 (PI3K/AKT pathway)

      • U73122 (PLCγ pathway)

    • Cycloheximide (protein synthesis inhibitor) blocks secondary effects requiring new protein production

  • Receptor analysis:

    • Use of cell lines expressing defined FGFR subtypes

    • Dominant-negative FGFR constructs to block specific receptor activation

    • FGFR knockdown/knockout approaches using siRNA or CRISPR-Cas9

  • Secretome analysis:

    • Analyze conditioned media from FGF-1-treated cells for secondary secreted factors

    • Use neutralizing antibodies against suspected secondary mediators

    • Perform transcriptional profiling to identify induced cytokines and growth factors

  • Cell type-specific responses:

    • Compare FGF-1 effects in heterogeneous cultures versus isolated cell populations

    • Use cell type-specific markers to distinguish direct versus indirect effects in complex tissues

These approaches, often used in combination, help construct a comprehensive picture of the direct signaling events triggered by FGF-1 versus the downstream consequences mediated by secondary factors.

What is the role of FGF-1 in mesenchymal-epithelial interactions during development?

FGF-1 serves as a critical mediator of mesenchymal-epithelial interactions during embryonic development and tissue homeostasis:

  • Spatial expression patterns:
    During lung morphogenesis, FGF-1 is dynamically expressed in the mesenchyme adjacent to distal epithelial buds . This localized expression suggests a role in directional outgrowth and induction of epithelial buds.

  • Regulatory mechanisms:
    The spatial and temporal expression of FGF-1 is tightly controlled by factors produced by the endoderm. For example, Sonic Hedgehog (Shh) appears to downregulate FGF-1 expression, as transgenic lungs overexpressing Shh in the endoderm show reduced FGF-1 transcription .

  • Directional morphogenesis:
    The localized source of FGF-1 in the mesenchyme creates concentration gradients that guide epithelial budding in specific directions. This spatial restriction of signaling is essential for proper branching architecture.

  • Cellular effects at the interface:

    • On epithelial cells: FGF-1 promotes proliferation, survival, and modulates differentiation

    • On mesenchymal cells: Creates autocrine reinforcement loops and regulates extracellular matrix production

    • At the basement membrane: Influences deposition and remodeling of matrix components

  • Coordination with other signaling pathways:
    FGF-1 signaling interfaces with other key developmental pathways, including:

    • Wnt signaling for cell fate decisions

    • BMP pathway for growth control

    • Notch signaling for boundary formation

    • Hedgehog pathway for spatial patterning

Understanding these mesenchymal-epithelial interactions mediated by FGF-1 provides insights into developmental processes and informs approaches to tissue engineering and regenerative medicine.

How do heparan sulfate proteoglycans modulate FGF-1 signaling, and how should this be accounted for experimentally?

Heparan sulfate proteoglycans (HSPGs) are critical regulators of FGF-1 signaling through multiple mechanisms that must be considered in experimental designs:

  • Molecular mechanisms of HSPG function:

    • Stabilize the FGF-1:FGFR complex by forming a ternary structure

    • Facilitate FGFR dimerization required for signaling

    • Protect FGF-1 from proteolytic degradation and thermal denaturation

    • Create local reservoirs of FGF-1 in the extracellular matrix

    • Modulate receptor specificity by selectively enhancing binding to certain FGFR isoforms

  • Experimental considerations:

    • Heparin supplementation: Include 1-5 μg/ml of heparin in FGF-1 preparations to enhance stability and activity

    • Standardization: Use the same heparin source and concentration across experiments

    • Control experiments: Include heparin-only controls to account for potential direct effects of heparin

    • Dose-response analysis: Test multiple FGF-1 concentrations with and without heparin

    • Cell culture variables: Consider that HSPG expression varies by cell type and culture conditions

  • Advanced experimental approaches:

    • Heparinase treatment to selectively remove cell surface HSPGs

    • Comparison of native FGF-1 with engineered heparin-independent variants

    • Use of synthetic heparan sulfate oligosaccharides with defined structures

    • Sodium chlorate treatment to inhibit cellular sulfation of heparan sulfate

  • Interpretation considerations:

    • FGF-1 experiments without appropriate heparin/HSPG consideration may yield misleading results

    • Differences in HSPG profiles between cell types can explain variable responses to the same FGF-1 concentration

    • In vivo and in vitro results may differ due to distinct HSPG environments

By systematically accounting for HSPG effects, researchers can achieve more consistent and physiologically relevant results in FGF-1 signaling studies.

What are the distinct and overlapping functions of FGF-1 compared to other FGF family members?

FGF-1 shares functional similarities with other FGF family members while possessing unique properties that distinguish it within this growth factor family:

  • Receptor activation profile:

    • FGF-1: Unique as a universal FGF ligand, activates all seven principal FGF receptors

    • FGF-2: Primarily activates FGFR1c and FGFR2c with moderate activity on other receptors

    • FGF-7: Highly specific for FGFR2b

    • FGF-10: Primarily activates FGFR2b with some activity on FGFR1b

  • Developmental functions:

    • FGF-1: Broad roles in tissue repair and homeostasis

    • FGF-2: Critical for early embryogenesis and neural development

    • FGF-10: Essential for lung, limb, and branching organ development

    • FGF-7: Epithelial wound repair and developmental epithelial-mesenchymal interactions

  • Experimental activity in branching morphogenesis:

    • FGF-1: Elicits immediate endodermal budding in isolated epithelium

    • FGF-7: Induces epithelial expansion without progression to budding

    • FGF-10: Causes initial expansion followed by multiple bud formation

  • Evolutionary conservation:
    FGF-1 shows remarkable conservation across species: 96% similarity between human and mouse/rat, 97% with porcine, and 92% with bovine versions . This high conservation suggests fundamental biological importance.

  • Experimental complementation studies:
    Due to its universal receptor binding, FGF-1 can often compensate functionally for other FGFs in experimental settings, while more receptor-restricted FGFs cannot substitute for each other.

Understanding these distinct and overlapping functions allows researchers to select the appropriate FGF for specific experimental questions and to interpret results in the context of the broader FGF signaling network.

How can researchers distinguish between FGF-1-mediated effects through different FGFR subtypes?

Differentiating between the contributions of individual FGF receptor subtypes to FGF-1 responses requires specialized experimental approaches:

  • Receptor-selective inhibitors and blocking antibodies:

    • Use receptor-specific tyrosine kinase inhibitors with differential IC50 values (e.g., PD173074 has higher affinity for FGFR1 than FGFR4)

    • Apply receptor-specific neutralizing antibodies to block individual FGFRs

    • Employ soluble receptor ectodomains as competitive inhibitors

  • Genetic manipulation approaches:

    • Knockout/knockdown: siRNA, shRNA, or CRISPR-Cas9 targeting specific FGFR subtypes

    • Dominant-negative receptors: Express truncated receptors lacking kinase domains

    • Receptor replacement: Rescue experiments in receptor-null backgrounds

    • Conditional gene deletion: Tissue-specific receptor knockout models

  • Cell models with defined receptor expression:

    • BaF3 cells (normally FGFR-negative) engineered to express single FGFR subtypes

    • Cell lines derived from receptor knockout animals

    • Compare responses in cells with known differential receptor expression profiles

  • Comparative ligand approach:

    • Test FGF-1 alongside receptor-selective FGFs (e.g., FGF-7 for FGFR2b)

    • Create dose-response curves for different endpoints across multiple ligands

    • Use chimeric FGF ligands with engineered receptor selectivity

  • Receptor isoform-specific readouts:

    • Monitor phosphorylation of receptor-specific adapter proteins

    • Analyze activation of distinct downstream pathways

    • Measure receptor-specific transcriptional responses

  • Advanced imaging techniques:

    • FRET-based assays to measure specific receptor activation

    • Live-cell imaging with receptor-specific fluorescent reporters

    • Proximity ligation assays to detect specific FGF-1/FGFR interactions

These approaches can be combined in complementary experimental designs to comprehensively map the contributions of individual FGF receptors to the biological effects of FGF-1.

How can recombinant FGF-1 be modified to enhance stability and specificity for research applications?

Advanced engineering approaches have created FGF-1 variants with enhanced properties for specialized research applications:

  • Stability enhancements:

    • Thermostabilizing mutations: Single point mutations like K12V or triple mutations (K12V/H138E/K144I) can increase thermal stability while maintaining activity

    • Disulfide engineering: Introduction of strategic disulfide bonds (e.g., L44C/F132C) can dramatically improve half-life

    • N-terminal modifications: Addition of short amino acid sequences or PEGylation to protect from N-terminal degradation

    • Core structure optimization: Mutations that enhance hydrophobic core packing improve resistance to denaturation

  • Receptor specificity modifications:

    • Isoform-selective mutations: F108A mutation increases FGFR3c selectivity

    • Affinity-enhancing changes: R35E modification enhances FGFR1c binding

    • Chimeric constructs: Domain swapping with other FGFs to engineer novel specificity profiles

    • Receptor-targeted variants: Addition of receptor-specific binding domains from other proteins

  • Functional enhancements:

    • Heparin-independence: Engineered variants that function without heparin cofactors

    • Extended serum half-life: Fusion to albumin-binding domains or Fc fragments

    • Controlled release forms: Fusion to matrix-binding domains for localized retention

    • Cell-specific targeting: Addition of cell-targeting peptides or antibody fragments

  • Experimental tool development:

    • Fluorescently labeled FGF-1: Site-specific conjugation of fluorophores for tracking

    • Photo-activatable variants: Caged FGF-1 that can be activated by light

    • Split complementation systems: FGF-1 fragments that regain activity upon reunification

    • Biosensor integration: FGF-1 fused to reporters that signal upon receptor binding

These engineered variants expand the experimental toolkit available to researchers studying FGF signaling and provide potentially superior reagents for specific applications in developmental biology, tissue engineering, and disease modeling.

What cell-free experimental systems can be used to study FGF-1 signaling mechanisms?

Cell-free systems offer powerful approaches to dissect FGF-1 signaling mechanisms with precise control over experimental variables:

  • Biochemical interaction analysis:

    • Surface Plasmon Resonance (SPR): Measures real-time binding kinetics between FGF-1 and purified FGFR ectodomains, with or without heparin

    • Isothermal Titration Calorimetry (ITC): Determines thermodynamic parameters of FGF-1/FGFR/heparin interactions

    • Analytical Ultracentrifugation: Characterizes the stoichiometry of FGF-1/FGFR/heparin complexes

    • Fluorescence Polarization: Monitors binding of labeled FGF-1 to receptors under various conditions

  • Structural approaches:

    • X-ray crystallography: Reveals atomic-level details of FGF-1/FGFR/heparin complexes

    • Cryo-electron microscopy: Visualizes receptor complexes in near-native states

    • NMR spectroscopy: Examines dynamic aspects of FGF-1 interactions

    • Hydrogen-deuterium exchange mass spectrometry: Maps binding interfaces and conformational changes

  • Reconstituted signaling systems:

    • Liposome-reconstituted receptors: FGFRs incorporated into artificial membranes

    • Cell-free kinase assays: Purified FGFRs and downstream substrates in solution

    • In vitro transcription/translation: Expression of signaling components from DNA templates

    • Xenopus oocyte extracts: Biological matrix containing signaling machinery

  • Advanced biophysical techniques:

    • Single-molecule FRET: Observes individual molecular interactions and conformational changes

    • Optical tweezers: Measures forces in FGF-1/FGFR interactions

    • Microfluidic systems: Controls gradients and temporal aspects of signaling

    • Label-free biosensors: Detects binding events without modification of interacting partners

These cell-free approaches complement cellular experiments by isolating specific components of the signaling pathway and enabling precise manipulation of molecular interactions that would be difficult to achieve in intact cells.

How can researchers effectively apply systems biology approaches to understand FGF-1 signaling networks?

Systems biology offers powerful frameworks to comprehensively map and model the complex signaling networks initiated by FGF-1:

  • Multi-omics data integration:

    • Phosphoproteomics: Captures the immediate signaling events following FGF-1 stimulation

    • Transcriptomics: Reveals the gene expression changes over time

    • Metabolomics: Identifies metabolic shifts resulting from FGF-1 signaling

    • Interactomics: Maps protein-protein interactions in the FGF-1 signaling network

    • Integration approaches: Computational methods to combine multiple data types into unified models

  • Network reconstruction and analysis:

    • Pathway enrichment analysis: Identifies canonical pathways activated by FGF-1

    • Network inference algorithms: Discovers novel connections within signaling networks

    • Perturbation analysis: Systematic inhibition of pathway components to map dependencies

    • Cross-talk identification: Reveals interactions between FGF-1 and other signaling systems

  • Dynamic modeling approaches:

    • Ordinary differential equations: Mathematical models of signaling dynamics

    • Logic-based models: Qualitative representation of signaling relationships

    • Agent-based models: Simulation of individual molecules in signaling cascades

    • Parameter estimation: Fitting models to experimental time-course data

  • Experimental validation strategies:

    • CRISPR screening: Systematic gene knockout to validate predicted network components

    • Optogenetic approaches: Precise temporal control of pathway activation

    • Live-cell biosensors: Real-time monitoring of multiple signaling nodes

    • Microfluidic single-cell analysis: Captures cell-to-cell variability in responses

  • Computational tools and resources:

    • Pathway databases: Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome

    • Protein interaction databases: STRING, BioGRID

    • Software platforms: CellDesigner, COPASI, VCell for modeling

    • Data visualization tools: Cytoscape for network visualization

By applying these systems biology approaches, researchers can move beyond reductionist views of FGF-1 signaling to understand emergent properties of the system, identify feedback and feed-forward loops, and predict cellular responses under diverse conditions.

Product Science Overview

Source and Production

The human recombinant FGF-a is produced in Sf9 insect cells using a baculovirus expression system. This method results in a single, glycosylated polypeptide chain containing 140 amino acids and having a molecular mass of approximately 15,803 Daltons . The protein is purified using proprietary chromatographic techniques to ensure high purity and biological activity .

Physical and Chemical Properties

The recombinant FGF-a is typically provided as a sterile, filtered liquid formulation. The formulation contains:

  • 20 mM Tris HCl (pH 7.9)
  • 100 mM KCl
  • 0.2 mM DTT
  • 20% glycerol

The protein solution is stable when stored at 4°C for short-term use (2-4 weeks) and at -20°C for long-term storage. It is recommended to add a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) to prevent multiple freeze-thaw cycles .

Biological Activity

FGF-a functions as a modifier of endothelial cell migration and proliferation, acting as an angiogenic factor. It is a potent mitogen for various mesoderm- and neuroectoderm-derived cells in vitro, suggesting its involvement in organogenesis . The biological activity of FGF-a is measured by its ability to stimulate the proliferation of BAF3 cells expressing FGF receptors, with an effective dose (ED50) of less than 10 ng/ml, corresponding to a specific activity of 100,000 IU/mg .

Applications

Due to its significant role in cell growth and development, FGF-a is widely used in laboratory research. It is particularly valuable in studies related to:

  • Angiogenesis: The formation of new blood vessels from pre-existing ones.
  • Wound Healing: Promoting tissue repair and regeneration.
  • Cancer Research: Investigating tumor growth and invasion mechanisms.
  • Developmental Biology: Understanding the processes of embryonic development and organogenesis.
Safety and Handling

FGF-a is intended for laboratory research use only and should not be used as a drug, agricultural or pesticidal product, food additive, or household chemical. Proper safety measures should be taken when handling the protein to avoid contamination and degradation .

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.