Recombinant Human Fibroblast growth factor 1 protein (FGF1) (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity

FGF1 stimulates proliferation of mesodermal, ectodermal, and neuroectodermal cells via fibroblast growth factor receptors (FGFRs).

Activity Assays

Cell LineED₅₀ (Proliferation)Heparin RequirementSpecific ActivitySource
BALB/c 3T3<2 ng/mlYes>5.0 × 10⁵ units/mg
NR6R-3T30.015–0.15 ng/mlYes (10 µg/ml)>2.0 × 10⁶ IU/mg
Primary endothelialAngiogenesis inducedYesN/A
  • Mechanism: Binds FGFR1 and integrins (ITGAV:ITGB3) in heparin-dependent ternary complexes, activating MAPK/ERK and AKT pathways .

  • Nuclear Role: Regulates p53 activity and apoptosis inhibition intracellularly .

Applications in Research

  • Cardiogenesis: Modulates protein kinase C signaling during heart development .

  • Angiogenesis: Promotes blood vessel formation in cancer and wound healing .

  • Cancer: Overexpressed in early-stage tumors; enhances metastasis in breast cancer models .

  • Neuroprotection: Supports neuronal survival under oxidative stress .

Stability-Mitogenicity Relationship

  • The K118E mutant (reduced heparin affinity) regained mitogenic activity when stabilized via mutations (e.g., Q40P/S47I), demonstrating heparin’s role in protecting FGF1 from degradation rather than receptor binding .

  • Thermodynamic stabilization compensates for weakened heparin interactions, restoring DNA synthesis and cell proliferation .

Clinical Implications

  • Cancer: FGF1 promotes tumor vascularization and survival, making it a therapeutic target .

  • Neurodegeneration: Astrocyte-derived FGF1 induces ApoE/HDL synthesis to combat oxidative stress .

Comparative Vendor Data

VendorMolecular WeightActivity (ED₅₀)Notes
Cell Guidance16 kDa<2 ng/ml (BALB/c 3T3)Discounts for bulk orders
Bio-Techne17.3 kDa<0.5 ng/ml (BALB/c)>2.0 × 10⁶ IU/mg specific activity
R&D Systems19 kDa0.015–0.15 ng/mlSilver-stained SDS-PAGE validation

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer ingredients, storage temperature, and the stability of the protein itself. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Acidic fibroblast growth factor; aFGF; Beta endothelial cell growth factor; Beta-endothelial cell growth factor; ECGF; ECGF beta; ECGF-beta; ECGFA; ECGFB; Endothelial Cell Growth Factor alpha; Endothelial Cell Growth Factor beta; FGF 1; FGF alpha; Fgf1; FGF1_HUMAN; FGFA; Fibroblast Growth Factor 1 Acidic; Fibroblast growth factor 1; GLIO703; HBGF 1; HBGF-1; HBGF1; Heparin binding growth factor 1; Heparin binding growth factor 1 precursor; Heparin-binding growth factor 1
Datasheet & Coa
Please contact us to get it.
Expression Region
16-155aa
Mol. Weight
16.0 kDa
Protein Length
Full Length of Mature Protein
Purity
>95% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Fibroblast Growth Factor 1 (FGF1) plays a crucial role in regulating various cellular processes, including survival, division, angiogenesis, differentiation, and migration. It acts as a potent mitogen in vitro and serves as a ligand for both FGFR1 and integrins. In the presence of heparin, FGF1 binds to FGFR1, inducing dimerization and activation of the receptor through autophosphorylation on tyrosine residues. These phosphorylated residues act as docking sites for interacting proteins, initiating the activation of multiple signaling cascades. FGF1 also interacts with the integrin ITGAV:ITGB3. Binding to integrin, subsequent ternary complex formation with integrin and FGFR1, and the recruitment of PTPN11 to the complex are essential for FGF1 signaling. This interaction leads to the phosphorylation and activation of FGFR1, FRS2, MAPK3/ERK1, MAPK1/ERK2, and AKT1. FGF1 has the ability to induce angiogenesis.
Gene References Into Functions
  1. Gas5 regulates proliferation and apoptosis in the growth plate by controlling FGF1 expression via miR-21 regulation. PMID: 29490650
  2. Modification of the heparin-binding region of FGF1 significantly enhances its cardioprotective efficacy, even in the presence of heparin, identifying a novel FGF ligand for therapeutic use in ischemic heart disease. PMID: 29016740
  3. FGF1 protects neuroblastoma cells from p53-dependent apoptosis through an intracrine pathway regulated by FGF1 phosphorylation. PMID: 29048426
  4. Specific heparin derivatives molecular recognition patterns by FGF1 have been reported. PMID: 27858202
  5. Multiple stepwise linear regression analysis found serum levels of FGF1 to be dependent on anti-diabetic drugs, hemoglobin A1C, body mass index, and sex. Serum levels of FGF1 are associated with a decreased risk of obesity in humans. PMID: 28303556
  6. Our study suggests that the genetic variants of FGF1 rs34011, more so than FGF2 rs2922979, may play a role in PE pathogenesis in Tunisian women. PMID: 27324104
  7. FGF1 and 2 strongly prevent the osteogenic commitment and differentiation of hBMSCs. PMID: 27305863
  8. Transfected FGF1 promotes angiogenic proliferation. PMID: 28281780
  9. These observations suggest a crucial role for cancer-associated fibroblasts and fibroblast growth factor-1/fibroblast growth factor receptor 4 signaling in the progression of ovarian cancer. The expression level of Snail1 and MMP3 was reduced, while the expression level of E-cadherin increased. PMID: 28718374
  10. Enzyme-linked immunosorbent assay was used to detect the levels of chemokine (C-X-C motif) ligand 12, chemokine (C-X-C motif) ligand 7, hepatocyte growth factor, and fibroblast growth factor 1 in the supernatants of the laryngeal squamous cell carcinoma and control cells. PMID: 28475003
  11. Fibroblast growth factor 1 (FGF1) is synergistically induced by heat shock and wounding. PMID: 27638903
  12. The results indicate that suramin blocks the interaction between hFGF1 and FGFR2 D2. PMID: 27387234
  13. Activation of AurA kinase through FGF1/FGFR signaling axis sustains the stem cell characteristics of glioblastoma cells. PMID: 27138904
  14. Study showed that both aFGF and bFGF were highly expressed in cervical cancer tissues. In tumors of higher clinical stages, the expression of these factors was further enhanced, suggesting that they play a role in facilitating cervical cancer cell proliferation. PMID: 27966750
  15. FGF1 may play a role in the pathogenesis of T2 diabetes mellitus. PMID: 26806193
  16. Anti-importin alpha1 antibody treatment suppressed the importin alpha1-FGF1 complex formation and ERK1/2 activation, resulting in decreased cell growth. This study provides novel evidence that functional importin alpha1 is located at the cell surface, where it accelerates the proliferation of cancer cells. PMID: 26887791
  17. The mutational introduction of a novel Cys residue (Ala66Cys) that forms a stabilizing disulfide bond (i.e., cystine) with one of the extant Cys residues (Cys83) effectively eliminates one Cys while increasing overall stability. PMID: 27019961
  18. The role of intracellular FGF1 is to protect the cell against stress conditions by providing an additional signal for cell survival, independently of receptor-activated signaling cascades. PMID: 26840910
  19. Data suggest folding of FGF1 is critical for its nonclassical secretion via permeability of the lipid bilayer; mutation of proline135 in the C-terminus of FGF1 leads to partial unfolding/decrease in FGF1's ability to permeabilize phosphatidylserine bilayers. PMID: 26836284
  20. The analysis identified a signaling axis between FGF signaling and the transcription factor Sox1, which is preferentially expressed in stem- and mesenchymal-like breast cancers. PMID: 26365194
  21. FGF-1 synthesis and secretion by synovial fibroblasts were significantly increased in osteoarthritis. PMID: 26400350
  22. Expression of human FGF1 solely in beta-cells in fgf1(-/-) animals prevented overnutrition-induced compensatory beta-cell differentiation. PMID: 26420862
  23. Results show that stress functionally associates FGF1 with AHNAK2 and both proteins with the cytoskeleton and their co-localization in the vicinity of the cell membrane. AHNAK2 seems to be an important element of the FGF1 export pathway. PMID: 25560297
  24. These findings suggest that the presence of FGF1 may serve as a prognostic indicator and a potential therapeutic target for non-small cell lung cancer patients, especially for lung squamous cell carcinoma. PMID: 26391572
  25. FGF1 expression is increased in the lungs of patients with idiopathic pulmonary fibrosis. PMID: 26138239
  26. The FGF1-FGFR1 axis promotes tongue squamous cell carcinoma metastasis through the epithelial-mesenchymal transition pathway. PMID: 26362179
  27. p120RasGAP shields Akt from deactivating phosphatases in FGF1 signaling but loses this ability once cleaved by caspase-3. PMID: 26109071
  28. The study presents a transcript profiling of remyelinated multiple sclerosis lesions and identified FGF1 as a promoter of remyelination. PMID: 25589163
  29. These results suggested that the human FGF1B promoter was active in ependymal cells, neurons, and a portion of dopaminergic neurons. PMID: 25104610
  30. This review focuses on the role of HDGF in tumorigenesis and metastasis, providing insight for application in clinical cancer therapy and its clinical implications as a prognostic marker in cancer progression. PMID: 25236340
  31. The sulfate in position 6 of d-glucosamine was essential for the mitogenic activity but not for the interaction with FGF-1. PMID: 25015527
  32. While results are awaited from these clinical investigations in squamous NSCLC and other disease settings, additional research is needed to elucidate the role of FGF/FGFR signaling in the biology of NSCLC of different histologies. PMID: 24711160
  33. FGF1 gene polymorphism is associated with a lower risk of developing cleft palate or cleft lip in Iranian patients. PMID: 24613087
  34. The precursor of the hormone Irisin (FNCD5) was abundantly expressed in all three fat depots, along with fibroblast growth factors (FGF) FGF1, FGF7, and FGF10, whereas FGF19 and FGF21 were undetectable. PMID: 23949615
  35. Nucleolin-FGF1 interaction is critical for the intranuclear phosphorylation of FGF1 by PKCdelta and thereby the regulation of nuclear export of FGF1. PMID: 24595027
  36. The genotype distribution of rs34011 within fibroblast growth factor 1 was significantly different between the Alzheimer's disease and control groups. PMID: 24464990
  37. Discovery of unexpected, neomorphic insulin-sensitizing action for exogenous non-mitogenic human FGF1 with therapeutic potential for the treatment of insulin resistance and type 2 diabetes. PMID: 25043058
  38. By introducing two stabilizing mutations in the C-terminal part of the protein, variants highly resistant proteolytically with prolonged half-life and enhanced mitogenic activity were obtained. PMID: 24304385
  39. Data suggest that the enhanced cell growth was likely due to the electrical stimulation up-regulated secretion of FGF-1 and FGF-2. PMID: 23990967
  40. Both FGF1 and ERBB2 significantly influenced overall survival in breast cancer patients, especially among women with low levels of Native American ancestry. PMID: 23912956
  41. We demonstrate that mesenchymal stromal cells increase FGF-1 secretion on co-culture with human primary chondrocytes, which, in turn, is responsible for increased human primary chondrocytes proliferation in pellet co-cultures. PMID: 23557133
  42. FGF1 polymorphism is associated with breast cancer. PMID: 23143756
  43. R50E suppresses angiogenesis induced by FGF1 or FGF2, and thereby indirectly suppresses tumorigenesis, in addition to its possible direct effect on tumor cell proliferation in vivo. PMID: 23469107
  44. The functional consequences of HSulf-2 activity on fibroblast growth factor (FGF)-induced mitogenesis were studied and found that the enzyme could differentially regulate FGF1 and FGF2 activities. PMID: 23457216
  45. The expression of FGF-1 in the in vitro fertilization (IVF) implantation failure group is less than in the fertile group, suggesting that growth factors such as FGF-1 are important maternal factors influencing implantation. PMID: 23426545
  46. Forms of FGF-1 that enhance thermostability or eliminate buried reactive thiols demonstrate a shorter distribution half-life, a longer elimination half-life, and a longer mean residence time (MRT) in comparison to wild-type FGF-1. PMID: 23133616
  47. The folding transition state of FGF-1 is shown to be highly polarized, with the majority of turns adopting either native-like or denatured-like structure in the folding transition state. PMID: 23047594
  48. LRRC59 facilitates transport of cytosolic FGF1 through nuclear pores by interaction with Kpns and movement of LRRC59 along the ER and NE membranes. PMID: 22321063
  49. [review] This review discusses recent and ongoing research into the role of fibroblast growth factor and transforming growth factor-beta in the etiopathogenesis of craniosynostosis. PMID: 21806346
  50. Plasticity in interactions of fibroblast growth factor 1 (FGF1) N terminus with FGF receptors underlies the promiscuity of FGF1. PMID: 22057274

Show More

Hide All

Database Links

HGNC: 3665

OMIM: 131220

KEGG: hsa:2246

STRING: 9606.ENSP00000338548

UniGene: Hs.483635

Protein Families
Heparin-binding growth factors family
Subcellular Location
Secreted. Cytoplasm. Cytoplasm, cell cortex. Cytoplasm, cytosol. Nucleus.
Tissue Specificity
Predominantly expressed in kidney and brain. Detected at much lower levels in heart and skeletal muscle.

Q&A

What is FGF1 and what are its basic biological functions?

FGF1, also known as acidic Fibroblast Growth Factor, is a 16-17 kDa nonglycosylated member of the FGF family of mitogenic peptides. It functions as a potent inducer of DNA synthesis, cell proliferation, and demonstrates chemotactic activities . FGF1 is produced by multiple cell types and stimulates the proliferation of cells of mesodermal origin and many cells of neuroectodermal, ectodermal, and endodermal origin .

Beyond its classical mitogenic role, FGF1 regulates cardiogenesis through protein kinase C signaling, functions as an insulin sensitizer, and mediates adipose tissue remodeling . It plays significant roles in development, regeneration, and angiogenesis . Recent research has also uncovered its importance in glucose homeostasis .

What are the alternative names for FGF1 in the scientific literature?

FGF1 appears in scientific literature under numerous alternative designations:

  • Fibroblast Growth Factor 1

  • Acidic Fibroblast Growth Factor

  • HBGF-1 (Heparin-binding growth factor 1)

  • β-endothelial growth factor

  • ECGF (Endothelial cell growth factor)

  • ECGF-β (Endothelial cell growth factor-β)

  • GLIO703

  • FGFA

  • ECGFA

  • AFGF

  • Endothelial cell growth factor-α

  • ECGFB

  • FGF-α

  • aFGF

This diversity of names reflects the protein's discovery in different contexts and its multiple biological roles across various tissues and cell types.

What is the typical structure and molecular characteristics of recombinant human FGF1?

Recombinant human FGF1 typically has the following characteristics:

PropertySpecification
Length141 amino acids (full length) or 154 amino acids (aa 2-155)
Molecular Weight16-19 kDa (depending on expression system and tags)
StructureMonomeric
SourceTypically produced in E. coli expression systems
Accession NumberP05230
Purity≥95% by SDS-PAGE under reducing and non-reducing conditions
Endotoxin Level≤1.00 EU/μg as measured by kinetic LAL

The amino acid sequence of full-length human FGF1 is: MFNLPPGNYK KPKLLYCSNG GHFLRILPDG TVDGTRDRSD QHIQLQLSAE SVGEVYIKST ETGQYLAMDT DGLLYGSQTP NEECLFLERL EENHYNTYIS KKHAEKNWFV GLKKNGSCKR GPRTHYGQKA ILFLPLPVSS D .

What typical bioassays are used to confirm FGF1 activity?

FGF1 activity is commonly assessed through cell proliferation assays using specific responsive cell lines. The most frequently used methods include:

  • Proliferation of BALB/c 3T3 cells: Human FGF1 stimulates proliferation with an ED50 typically less than 2 ng/ml, corresponding to an expected specific activity of 5.0 × 10^5 units/mg .

  • NR6R-3T3 mouse fibroblast cell proliferation: Recombinant Human FGF1 stimulates cell proliferation with an ED50 of 0.015-0.15 ng/mL in the presence of 10 μg/mL of heparin .

  • SDS-PAGE analysis: Protein quality is assessed through SDS-PAGE under reducing and non-reducing conditions, with silver staining typically showing a single band at the expected molecular weight (16-19 kDa) .

These standardized bioassays provide reliable measures of both protein quality and functional activity, essential for research applications.

How can researchers optimize FGF1 signaling in experimental systems?

Optimizing FGF1 signaling in experimental systems requires careful consideration of several parameters:

  • Heparin co-administration: FGF1 activity is significantly enhanced by heparin or heparan sulfate. For optimal activity, researchers should include 10 μg/mL of heparin in their experimental systems . The heparin-binding properties of FGF1 are critical for stabilizing the FGF1-FGFR complex .

  • Receptor expression profiling: Before designing experiments, characterize the FGFR expression profile of your cell system. FGF1 can bind and activate all FGFR isoforms, but the expression levels will determine sensitivity .

  • Thermal stability considerations: FGF1 complexed with its receptor and heparin demonstrates increased thermal stability. In experimental setups requiring elevated temperatures, ensure the presence of stabilizing factors. The FGF1 WT-FGFR1c-HS complex shows significantly higher thermal stability (higher Tm) compared to complexes without heparin .

  • Careful storage and handling: Reconstituted FGF1 should be stored at appropriate temperatures (-20°C to -80°C) and minimize freeze-thaw cycles to maintain activity. Consider adding carrier proteins for dilute solutions to prevent adsorption to surfaces .

  • Signal pathway monitoring: For comprehensive analysis, monitor multiple downstream pathways as FGF1 activates diverse signaling cascades including MAPK/ERK, PI3K/AKT, and PLCγ pathways .

How can researchers engineer FGF1 variants with altered functional properties?

Engineering FGF1 variants with modified properties has become an important approach for dissecting its various functions. Key strategies include:

  • Heparin-binding site modifications: Mutating key residues in the heparin-binding site (HBS) can alter the stability of FGF1-FGFR dimers and subsequently modify signaling strength. For example, the FGF1^ΔHBS triple mutant (K127D, K128Q, K133V) shows compromised heparin-binding affinity and reduced ability to promote HS-assisted FGFR dimerization .

  • Structure-guided design: Using crystallographic data of FGF1-FGFR-heparin complexes to identify critical interaction residues. This approach allows for precise modifications that affect specific aspects of FGF1 function .

  • Biophysical validation: Employ techniques such as surface plasmon resonance (SPR) to confirm altered binding properties, size-exclusion chromatography with multi-angle light scattering (SEC-MALS) to assess complex formation, and thermal shift assays to evaluate stability changes .

  • Functional validation: Compare wild-type and variant FGF1 proteins in cell-based assays measuring both mitogenic responses (e.g., BrdU incorporation) and metabolic responses (e.g., insulin sensitization, glucose uptake) .

The utility of this approach is demonstrated by the FGF1^ΔHBS variant, which showed a ~13°C lower unfolding temperature than wild-type FGF1 when in complex with FGFR1c and heparin, confirming reduced stability of the signaling complex .

What methodologies are used to study FGF1-FGFR signaling networks?

Studying FGF1-FGFR signaling networks requires multifaceted approaches:

  • Curated pathway databases: Researchers have developed resources like NetPath that document specific pathway reactions triggered by the FGF-1/FGFR system across various cell types and tissues .

  • Systematic literature mining: Comprehensive literature searches using databases like NCBI PubMed to retrieve and document molecular events triggered by FGF1/FGFR signaling, including:

    • Physical associations between proteins

    • Posttranslational modifications (PTMs)

    • Changes in subcellular localization

    • Activation or inhibition of specific proteins

    • Regulation of gene expression

  • Phosphoproteomics: Mass spectrometry-based approaches to identify proteins phosphorylated following FGF1 stimulation, revealing activation of signaling cascades .

  • Pathway visualization tools: Software platforms that enable visualization and analysis of complex signaling networks, integrating data from multiple experimental sources .

  • ERK-dependent signaling analysis: Monitoring phosphorylation of downstream targets such as DRP1 at Ser616, which mediates processes like mitochondrial fission and mitophagy in response to FGF1 stimulation .

These methodologies provide a comprehensive understanding of the complex signaling cascades initiated by FGF1, essential for both basic research and therapeutic development.

How can FGF1 be optimally used for muscle stem cell (MuSC) research?

For muscle stem cell research, FGF1 plays a crucial role in promoting proliferation while maintaining stemness. Optimal utilization involves:

  • Serum-free culture conditions: Recombinant FGF1 significantly enhances the proliferation of porcine MuSCs under serum-free conditions, enabling animal component-free culture systems essential for both basic research and applications like cultured meat production .

  • Mitochondrial dynamics assessment: FGF1 induces mitochondrial fission and mitophagy by activating ERK-dependent phosphorylation of DRP1 at Ser616, resulting in improved mitochondrial function and proliferation capacity in MuSCs. Researchers should monitor these parameters when studying FGF1 effects on muscle progenitors .

  • Species-specific recombinant proteins: While human FGF1 shows cross-reactivity with cells from multiple species due to high sequence conservation (human FGF1 shares 92%, 96%, 96%, and 96% amino acid sequence identity with bovine, mouse, porcine, and rat FGF1, respectively), species-matched recombinant proteins may provide optimal results for specific applications .

  • Dosage optimization: Determine the optimal concentration of FGF1 for MuSC proliferation without triggering premature differentiation. This typically requires dose-response experiments for each specific cell isolation and culture system .

  • Combination with other factors: For long-term expansion, combining FGF1 with other growth factors and small molecules that maintain stemness can enhance outcomes .

What are the research applications of FGF1 in neural and epithelial cell systems?

FGF1 has diverse applications in neural and epithelial cell research:

For neural systems:

  • Neural progenitor maintenance: FGF1's neurotrophic properties make it ideal for maintaining neural progenitors in culture .

  • Glial cell support: FGF1 supports oligodendrocytes and astroglia, making it valuable for studying neuron-glia interactions .

  • Remyelination studies: In multiple sclerosis research, FGF1 has shown potential to promote remyelination of neurons, making it a valuable tool for both basic and translational neuroscience .

For epithelial systems:

  • Organoid development: FGF1 promotes branching of epithelial cells, making it useful for embryonic lung epithelium cultures and human iPSC-derived ureteric bud organoids .

  • Endothelial cell proliferation: FGF1 is used to promote the differentiation and proliferation of endothelial cells, supporting vascular research .

  • Angiogenesis models: Due to its role in blood vessel formation, FGF1 is employed in models of developmental and therapeutic angiogenesis, including potential applications for myocardial infarction treatment .

These applications highlight the versatility of FGF1 across different cell types and research areas, from basic developmental biology to disease modeling and regenerative medicine.

How can researchers utilize FGF1 for metabolic research and diabetes studies?

FGF1 has emerged as a significant factor in metabolic research, particularly for diabetes studies:

  • Glucose homeostasis models: FGF1 has been discovered to play important roles in glucose regulation. In experimental models, FGF1 injections can lower glucose levels without risk of hypoglycemia through effects on glucose-sensing neuronal circuits .

  • Uncoupling mitogenic and metabolic functions: Engineered FGF1 variants like FGF1^ΔHBS allow researchers to separate the protein's growth-promoting effects from its metabolic regulatory effects. This enables more precise studies of metabolic pathways without confounding effects on cell proliferation .

  • Insulin sensitivity research: FGF1 functions as an insulin sensitizer, making it valuable for studying mechanisms of insulin resistance and for developing potential therapies for Type 2 diabetes .

  • Adipose tissue remodeling: FGF1 mediates adipose tissue remodeling processes, offering a tool for investigating adipose tissue biology and obesity-related research .

  • Central nervous system regulation: Studies suggest FGF1 affects metabolic regulation through central nervous system mechanisms, opening avenues for research into brain-peripheral tissue communication in metabolic disorders .

These diverse applications in metabolic research highlight FGF1's potential beyond its classical mitogenic role, positioning it as an important molecule for both basic and translational diabetes research.

What are common challenges in recombinant FGF1 production and how can they be addressed?

Production of high-quality recombinant FGF1 presents several challenges:

  • Soluble expression: Achieving high yields of soluble FGF1 in E. coli can be challenging. Recent advances have established efficient methods for soluble expression, with reported yields of up to 48 mg of purified protein per liter of culture for porcine FGF1 . Strategies include:

    • Optimization of induction temperature (typically lower temperatures improve solubility)

    • Selection of appropriate E. coli strains

    • Use of fusion tags that enhance solubility

    • Co-expression with chaperones

  • Protein stability: FGF1 can be relatively unstable in solution. Approaches to improve stability include:

    • Addition of heparin during purification and storage

    • Inclusion of stabilizing excipients

    • Careful buffer optimization

    • Storage at appropriate temperatures (-20°C to -80°C)

  • Endotoxin removal: As FGF1 is typically produced in E. coli, endotoxin contamination must be minimized, especially for sensitive cell culture applications. Commercial preparations typically guarantee endotoxin levels ≤1.00 EU/μg .

  • Activity verification: Ensuring consistent biological activity between batches is critical. Standardized bioassays using responsive cell lines (BALB/c 3T3 or NR6R-3T3) should be employed to verify each preparation's potency .

  • Animal component-free production: For certain applications, animal component-free production is essential. This requires careful selection of all media components and purification reagents throughout the production process .

How should researchers optimize FGF1 storage and handling for maximum activity?

Proper storage and handling of FGF1 is crucial for maintaining its biological activity:

  • Reconstitution recommendations:

    • Use sterile, buffered solutions (typically PBS) for reconstitution

    • For low concentration solutions, consider adding carrier proteins (e.g., BSA) to prevent adsorption to surfaces

    • Filter sterilize through 0.2 μm filters if necessary

  • Storage conditions:

    • Store lyophilized FGF1 at -20°C to -80°C

    • After reconstitution, prepare single-use aliquots to avoid repeated freeze-thaw cycles

    • Short-term storage (1-2 weeks) at 4°C is possible for working solutions with appropriate preservatives

  • Stability considerations:

    • FGF1 is more stable in the presence of heparin

    • Avoid repeated freeze-thaw cycles which can lead to significant loss of activity

    • Monitor pH during storage as shifts can affect protein stability

  • Working solution preparation:

    • Prepare fresh working solutions when possible

    • When using FGF1 in cell culture, include heparin (typically 10 μg/mL) for maximum activity

    • Calculate ED50 based on known values (0.015-0.15 ng/mL in the presence of heparin for NR6R-3T3 cells)

  • Quality control:

    • Periodically verify activity of stored FGF1 using standardized bioassays

    • Monitor protein integrity through techniques like SDS-PAGE if activity appears compromised

These recommendations will help ensure consistent experimental results when working with this sensitive protein.

What controls should be included in FGF1 signaling experiments?

Robust FGF1 signaling experiments require comprehensive controls:

  • Activity controls:

    • Positive control: A standardized preparation of FGF1 with verified activity

    • Negative control: Vehicle-only treatment following identical handling procedures

    • Dose-response curve: Multiple concentrations of FGF1 to demonstrate specificity and determine optimal working concentration

  • Receptor specificity controls:

    • FGFR inhibitor controls: Small molecule inhibitors of FGFR kinase activity (e.g., PD173074)

    • Heparin control: Experiments with and without heparin to demonstrate its effect on signaling

    • Heparinase treatment: To demonstrate the requirement for endogenous cell surface heparan sulfate

  • Pathway validation controls:

    • Inhibitors of downstream pathways (e.g., U0126 for MEK/ERK pathway)

    • Positive controls for each pathway being investigated

    • Time-course analysis to capture both early and late signaling events

  • FGF1 variant controls:

    • When studying specific aspects of FGF1 signaling, engineered variants like FGF1^ΔHBS can serve as valuable controls to distinguish between different functional outcomes

    • Wild-type comparison for any modified FGF1 protein

  • Cell-specific controls:

    • Cell lines with known FGFR expression profiles

    • FGFR knockdown/knockout controls

    • For metabolic studies, insulin sensitivity controls to differentiate FGF1-specific effects from general metabolic changes

What are emerging therapeutic applications of FGF1 research?

FGF1 research is opening several promising therapeutic frontiers:

  • Diabetes treatment: FGF1's ability to lower glucose levels without hypoglycemia risk makes it a promising candidate for treating Type 2 diabetes. The engineering of FGF1 variants with selective metabolic activity while minimizing mitogenic potential represents a significant advance in this direction .

  • Neurodegenerative diseases: FGF1 shows potential in treating conditions like multiple sclerosis through its ability to promote remyelination of neurons. Research is exploring delivery methods and optimal dosing regimens for neurological applications .

  • Cardiovascular therapies: FGF1's role in angiogenesis has potential applications for novel therapies following myocardial infarction, potentially promoting revascularization of damaged heart tissue .

  • Regenerative medicine: Applications in tissue regeneration and wound healing leverage FGF1's mitogenic properties for accelerating repair processes .

  • Cancer therapies: While FGF1 can promote cancer progression in some contexts, understanding its signaling has led to investigations into targeted therapies to inhibit FGF1 signaling in cancer cells .

These diverse therapeutic possibilities highlight the importance of continued fundamental research into FGF1 biology and signaling mechanisms.

How is FGF1 research contributing to sustainable food technology?

Recent advances demonstrate FGF1's emerging role in sustainable food technology:

  • Cultured meat production: Recombinant FGF1 is being employed to develop animal component-free in vitro systems for muscle stem cell (MuSC) expansion. Recent research reported in January 2025 established an efficient method for producing recombinant porcine FGF1 (rpFGF1) with yields of 48 mg of purified protein per liter of bacterial culture .

  • Serum-free culture systems: Treatment with rpFGF1 significantly enhanced proliferation of porcine MuSCs under serum-free conditions, addressing a key challenge in scaling cultured meat production without animal-derived components .

  • Mitochondrial function enhancement: rpFGF1 was shown to induce mitochondrial fission and mitophagy by activating ERK-dependent phosphorylation of DRP1 at Ser616, resulting in improved mitochondrial function and proliferation capacity in porcine MuSCs. This mechanistic understanding enables optimization of culture conditions .

  • Scalability improvements: The development of efficient bacterial expression systems for FGF1 production contributes to the economic feasibility of cultured meat by providing cost-effective growth factors .

This research addresses the challenges associated with traditional livestock farming while working toward meeting future protein demands through technological innovation.

What new methodologies might advance our understanding of FGF1 signaling specificity?

Several innovative approaches are poised to deepen our understanding of FGF1 signaling specificity:

  • Single-cell signaling analysis: Applying single-cell technologies to track FGF1 responses would reveal cell-to-cell variability in signaling outcomes and potentially identify previously unrecognized subpopulations with distinct response patterns.

  • Structural biology advances: Cryo-electron microscopy and advanced crystallography of FGF1-FGFR complexes in different cellular contexts could reveal subtleties in receptor conformation that dictate downstream signaling specificity .

  • Systems biology integration: Comprehensive network mapping of FGF1/FGFR signaling using multi-omics approaches (transcriptomics, proteomics, metabolomics) can provide a holistic view of pathway interactions and cross-talk .

  • CRISPR-based signaling perturbation: Systematic CRISPR screens targeting components of FGF1 signaling networks could identify new regulators and reveal unexpected pathway connections.

  • Engineered cellular systems: Designer cell lines with controlled expression of specific FGFR isoforms and downstream effectors would allow precise dissection of signaling branch points.

  • Advanced imaging approaches: Techniques such as live-cell FRET sensors for monitoring FGF1-induced conformational changes in real-time could provide dynamic insights into signaling initiation and propagation.

These methodological advances will be essential for developing the next generation of FGF1-based therapeutics and research tools.

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.