Recombinant Mouse Fibroblast growth factor 1 (Fgf1) (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity and Mechanisms

FGF1 (Active) binds FGFR1 and integrins (e.g., ITGAV:ITGB3), triggering signaling cascades (MAPK/ERK, AKT) . Key functional insights include:

  • Mitogenicity: Stimulates 3T3 fibroblast proliferation at ED₅₀ <0.2 ng/mL .

  • Metabolic Effects:

    • A single intracerebroventricular (i.c.v.) injection in diabetic mice (ob/ob, db/db) induces sustained normoglycemia (>48 hours) via enhanced peripheral glucose clearance .

    • Truncated FGF1 (lacking residues 1–24) retains glucose-lowering activity but reduces mitogenicity .

  • Cardiac Role: Promotes cardiomyocyte regeneration and improves post-infarction cardiac function .

Table 2: Key Experimental Models and Outcomes

ApplicationModel SystemKey Finding(s)Source
Diabetes Researchob/ob miceSingle i.c.v. dose restores blood glucose
Cardiac RepairF1A-CreERT2 transgenic miceFGF1A promoter activity in cardiomyocytes
Cell Proliferation3T3 fibroblastsED₅₀ <0.2 ng/mL with heparin co-treatment
CNS SignalingHypothalamic glial cellsGlia-mediated glucose homeostasis via melanocortin

Therapeutic Potential

  • Diabetes: FGF1 bypasses insulin resistance by increasing peripheral glucose uptake without hypoglycemia risk .

  • Cardiovascular Disease: Enhances cardiomyocyte proliferation and reduces oxidative stress in diabetic cardiomyopathy .

  • Safety Profile: Modified FGF1 variants show reduced mitogenicity while retaining metabolic benefits .

Critical Considerations

  • Dosage: Efficacy in mice: 3 µg i.c.v. ; in vitro: 0.05–0.25 ng/mL with heparin .

  • Mechanistic Complexity: Requires heparin for FGFR1 binding and ternary complex formation with integrins .

  • Species Specificity: Rat FGF1 also effective in Zucker Diabetic Fatty rats .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered solution containing 20 mM Tris-HCl, 500 mM NaCl, pH 6.6.
Form
Available in both liquid and lyophilized powder forms.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the chosen purchase method and location. For specific delivery details, please consult your local distributor.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance, as additional fees will apply.
Shelf Life
The shelf life is influenced by various factors including storage conditions, buffer composition, temperature, and the inherent stability of the protein itself.
Generally, the shelf life of liquid form is 6 months when stored at -20°C/-80°C. For lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Fgf1; Fgf-1; Fgfa; Fibroblast growth factor 1; FGF-1; Acidic fibroblast growth factor; aFGF; Heparin-binding growth factor 1; HBGF-1
Datasheet & Coa
Please contact us to get it.
Expression Region
16-155aa
Mol. Weight
15.7 kDa
Protein Length
Full Length of Mature Protein
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor 1 (FGF1) plays a critical role in regulating various cellular processes, including cell survival, division, angiogenesis (blood vessel formation), differentiation, and migration. In vitro, it acts as a potent mitogen (stimulates cell division). FGF1 serves as a ligand for both FGFR1 (fibroblast growth factor receptor 1) and integrins. In the presence of heparin, FGF1 binds to FGFR1, triggering dimerization and activation of the receptor via sequential autophosphorylation of tyrosine residues. These phosphorylated tyrosines act as docking sites for interacting proteins, leading to the activation of several signaling cascades. FGF1 also binds to integrin ITGAV:ITGB3. This binding, subsequent ternary complex formation with integrin and FGFR1, and the recruitment of PTPN11 to the complex are essential for FGF1 signaling. This complex ultimately induces phosphorylation and activation of FGFR1, FRS2, MAPK3/ERK1, MAPK1/ERK2, and AKT1. FGF1 can also induce angiogenesis.
Gene References Into Functions
  1. Research indicates that aFGF (acidic fibroblast growth factor) accelerates the progression of atherosclerosis and suggests its potential as a therapeutic target for preventing atherosclerosis development. PMID: 29845277
  2. TUG1 knockdown (reduction in expression) mitigates atherosclerosis by modulating FGF1 via miR-133a in ApoE knockout mice. PMID: 29268138
  3. Fibroblast growth factors (FGF1 and FGF2), unlike vascular endothelial growth factor (VEGF), rescued Psen1-/- cells from serum starvation-induced apoptosis. In the absence of serum, FGF2 immunoreactivity was diffusely distributed in cytoplasmic and nuclear vesicles of both wild-type and Psen1-/- cells, with no significant difference in FGF2 levels in nuclear and cytosolic fractions. PMID: 27443835
  4. Suboptimal FGFR activation by a weak FGF1-FGFR dimer is sufficient to trigger a metabolic response, while full FGFR activation through stable and sustained dimerization is required for a mitogenic response. PMID: 28813681
  5. FGF1 protects neuroblastoma cells from p53-dependent apoptosis through an intracrine pathway regulated by FGF1 phosphorylation. PMID: 29048426
  6. The HDAC3-N-CoR corepressor complex leaves the Fgf1b promoter, and a complex involving translocated CRTC1, phosphorylated CREB, and histone acetyltransferase CBP induces transient transcription. PMID: 28076781
  7. lncRNA-Map2k4 is the target gene of miR-199a, and its down-regulation promotes miR-199a expression in neurons. miR-199a targeted regulation of FGF1 expression in neurons. PMID: 28655615
  8. Studies clearly demonstrate the distinct specificity of FGF12-FGFR1c2 and FGF22-FGFR1c2 for well-defined HS structures and suggest the feasibility of chemoenzymatically synthesizing precise HS polysaccharides that can selectively mediate growth factor signaling. PMID: 28031461
  9. Findings support a pro-adipogenic role for betaKlotho in skeletal muscle fibro/adipogenesis and necessitate further investigation into the involvement of the FGF-FGFR-betaKlotho axis in the fibro/adipogenic infiltration associated with functional deterioration of skeletal muscle during aging and muscular dystrophy. PMID: 26881702
  10. Fibroblast growth factor 1 (FGF1) is synergistically induced by heat shock and wounding. PMID: 27638903
  11. Data indicate that Fgf1-mediated signaling represents a significant signaling cascade related to adipogenesis and visceral adiposity; expression of Fgf1 (fibroblast growth factor 1) and Fgfr1 (fibroblast growth factor receptor 1) is up-regulated in adipose tissue of obese mice (both those with a high-fat diet and those with genetic deletion of leptin). PMID: 26847131
  12. These results suggest that FGFs promote hair growth by inducing the anagen phase in resting hair follicles and could potentially be a hair growth-promoting agent. PMID: 25685806
  13. The study presents a transcript profiling of remyelinated multiple sclerosis lesions and identifies FGF1 as a promoter of remyelination. PMID: 25589163
  14. Nucleolin-FGF1 interaction is critical for the intranuclear phosphorylation of FGF1 by PKCdelta and, consequently, the regulation of nuclear export of FGF1. PMID: 24595027
  15. Parenteral delivery of a single dose of recombinant FGF1 (rFGF1) results in potent, insulin-dependent lowering of glucose levels in diabetic mice, which is dose-dependent but does not lead to hypoglycemia. PMID: 25043058
  16. Although FGF1 transgenic mice exhibited a normal phenotype with unperturbed kidney structure, they showed severely inhibited kidney repair after unilateral ischemia/reperfusion. This was manifested by a significant decrease in postischemic kidney size and weight. PMID: 22606265
  17. Retinoic acid (RA), via RALDH2, has distinct functions in the developing spinal cord to (i) maintain high levels of FGF and Notch signaling and (ii) drive stem cell differentiation, thus restricting both the numbers and the pluripotent character of neural stem cells. PMID: 22396766
  18. The discovery of a phenotype for the FGF1 knockout mouse establishes the PPARgamma-FGF1 axis as crucial for maintaining metabolic homeostasis and insulin sensitization. PMID: 22522926
  19. The results demonstrate that the FGF1/FGFR1 complex constitutes a signaling module that, independently of the receptor tyrosine kinase, can convey a signal that initiates a strictly timed and periodic release of endocytosed FGF1 into the cytosol/nucleus. PMID: 21223966
  20. RFX1 may negatively regulate the self-renewal of GBM-SCs (glioblastoma stem cells) by modulating FGF-1B and FGF1 expression levels through binding to the 18-bp cis-elements of the F1B promoter. PMID: 20189986
  21. The neuroprotective effects of FGF1 involve inactivation of GSK3beta. PMID: 12095987
  22. FGF-1, FGF-2, and FGF receptor-1 levels in the cochlear nucleus following acoustic overstimulation. PMID: 12121735
  23. Depletion of FGF1 may be a key regulatory component in the initial phase of branching morphogenesis of the lung bud epithelium in vitro. PMID: 12128203
  24. Agonist role for FGF1 and FGF2 specifically in insult-induced liver matrix deposition and hepatic fibrogenesis, highlighting their potential as targets for preventing hepatic fibrosis. PMID: 14507672
  25. Non-classical release is mediated by synaptotagmin 1. PMID: 14559220
  26. High steady-state levels of ONOO(-) may induce cysteine oxidation, tyrosine nitration, and irreversible inactivation of FGF-1, potentially serving as an inhibitory feedback mechanism restoring cellular homeostasis during inflammation resolution and repair. PMID: 14592461
  27. Gab1 is essential for FGF1 stimulation of both PI 3-kinase and the antiapoptotic protein kinase Akt, while FGF1-induced MAPK stimulation is not affected by Gab1 deficiency. PMID: 15199124
  28. Data show that mouse and human fibroblast growth factor 1 (FGF-1) internal ribosome entry sites exhibit similar activity profiles and share a conserved structural domain at both the nucleotide sequence and RNA structure levels. PMID: 15314170
  29. Phosphorylation of Fgf1 occurs in the nucleus by pkcdelta and is subsequently exported to the cytosol. PMID: 15574884
  30. Hsp90 is required for the translocation of FGF-1 and FGF-2 across the endosomal membrane. PMID: 16495214
  31. FGF7- and FGF1-induced mitogenesis and downstream signaling are controlled by distinct heparin octasaccharide motifs. PMID: 16728399
  32. FGF1 and p40 synaptotagmin 1 release correlates with membrane destabilizing ability. PMID: 16930531
  33. These results identify a novel aspect of the crosstalk between FGF and thrombin signaling pathways, both of which play crucial roles in tissue repair and angiogenesis. PMID: 17027650
  34. Shisa antagonizes Wnt and Fgf signalings. PMID: 17481602
  35. In apoE-deficient mouse astrocytes, FGF-1 stimulated cholesterol biosynthesis without enhancing its release, indicating a signaling pathway independent of apoE biosynthesis upregulation. PMID: 17548887
  36. Formation of the FGF-1-exportin-1-Ran-GTP complex in vitro, as well as nuclear export of FGF-1 in vivo, was dependent on phosphorylation of FGF-1 and was abolished by leptomycin B. PMID: 17616529
  37. Sphingosine kinase 1 is a component of the copper-dependent FGF1 release pathway. PMID: 17643421
  38. A conserved, specific, and stage-dependent requirement for Erk1/2 signaling downstream of FGF-induced neural specification in higher vertebrates. PMID: 17660197
  39. FGF and PDGF have roles in cell proliferation and migration in osteoblastic cells. PMID: 17852407
  40. Analysis of expression of a bicistronic vector driven by the FGF-1 IRES (internal ribosome entry site) in mouse muscle. PMID: 17963525
  41. 6-O-sulfate in HS (heparan sulfate) may regulate the signalings of some HB-GFs (heparin-binding growth factors), including FGF-1, FGF-2, and FGF-4, by inducing different interactions between ligands and their receptors. PMID: 18281280
  42. Fibroblast growth factor signaling and Wnt signaling interact to control the proximal-distal pattern of forming airways in the mammalian lung. PMID: 18694942
  43. These data demonstrate the existence of novel cross-talk between thrombin, FGF, and Notch signaling pathways, which play important roles in vascular formation and remodeling. PMID: 18784255
  44. An interplay between retinoic acid, Fgf, and Shh signaling is likely to be an important mechanism underlying the tight regulation of caudal embryonic development. PMID: 19168680
  45. A detailed double-label immunohistochemical investigation of the localization patterns of FGF1 and its receptors FGFR1 and FGFR2 in adult and early postnatal mouse retinas is reported. PMID: 19408015
  46. Results revealed a mechanism of molecular coupling of mRNA transcription and translation, involving a unique process of IRES activation by a FGF1 promoter element. PMID: 19561198
  47. Heparin's main role in FGF-induced signaling is to protect this naturally unstable protein against heat and/or proteolytic degradation. Heparin is not essential for direct FGF1-FGFR interaction and receptor activation. PMID: 19574212

Show More

Hide All

Database Links
Protein Families
Heparin-binding growth factors family
Subcellular Location
Secreted. Cytoplasm. Cytoplasm, cell cortex. Cytoplasm, cytosol. Nucleus.

Q&A

What is the molecular structure and functional role of FGF1?

Recombinant mouse FGF1 is typically produced as a protein encompassing amino acids 16-155, with the sequence: MFNLPLGNYKKPKLLYCSNGGFFLRILPDGTVDGTRDRSDQHIQLQLSAESAGEVYIKGTETGQYLAMDTEGLLYGSQTPNEECLFLERLEENHYNTYTSKKHAEKN WFVGLKKNGSCKRGPRTHYGQKAILFLPLPVSSD . FGF1 adopts a β-trefoil core structure, with characteristic tryptophan residues that produce fluorescence emission spectra centered at 307 nm when properly folded .

Functionally, FGF1 plays important roles in regulating cell survival, cell division, angiogenesis, cell differentiation, and cell migration . It acts as a potent mitogen in vitro and functions as a ligand for FGFR1 and integrins . When bound to FGFR1 in the presence of heparin, FGF1 induces receptor dimerization and activation through sequential autophosphorylation on tyrosine residues . This creates docking sites for interacting proteins and activates several signaling cascades including MAPK3/ERK1, MAPK1/ERK2, and AKT1 pathways .

How does FGF1 signaling operate at the cellular level?

FGF1 operates through both extracellular and intracellular mechanisms:

Extracellular signaling:

  • Binds to FGFR1 in the presence of heparin, leading to receptor dimerization

  • Forms a ternary complex with integrin ITGAV:ITGB3 and FGFR1

  • Recruits PTPN11 (SHP-2) to this complex, which is essential for signaling

  • Activates downstream pathways including MAPK and AKT cascades

Intracellular actions:

  • Can cross the cellular membrane and translocate to the cytosol and nucleus

  • Interacts with at least 20 identified intracellular binding partners

  • Many of these binding partners are involved in apoptosis, cell cycle, and proliferation, suggesting a role in cell survival

What experimental systems best demonstrate FGF1 activity?

Researchers should consider these experimental approaches when studying FGF1:

In vitro systems:

  • Cell proliferation assays to measure mitogenic activity

  • Receptor phosphorylation detection via Western blotting (FGFR1, FRS2, MAPK, AKT)

  • Angiogenesis assays such as endothelial tube formation

  • Neuronal electrophysiology to measure membrane potential changes in responsive neurons

In vivo models:

  • Diabetic mouse models (db/db, ob/ob) for studying metabolic effects

  • Cardiomyopathy models for cardiac protection studies

  • Central nervous system administration to study neuronal activation

How should researchers validate recombinant mouse FGF1 activity?

A comprehensive validation approach includes:

Structural validation:

  • SDS-PAGE to confirm molecular weight and purity (>95%)

  • Circular dichroism to verify proper protein folding

  • Fluorescence spectroscopy to confirm characteristic emission patterns (307 nm)

Functional validation:

  • Cell-based assays measuring:

    • Proliferation (mitogenic activity)

    • Receptor phosphorylation (FGFR1, FRS2, MAPK, AKT)

    • Angiogenic response

  • Binding assays (SPR or pull-down) to confirm interactions with FGFR1, integrins, or known binding partners

  • Electrophysiological recordings to confirm neuronal activation patterns

Quality control parameters:

  • Endotoxin levels should be <1 EU/μg for in vivo applications

  • Confirmation of absence of microbial contamination

  • Batch-to-batch consistency verification

What experimental design considerations are critical when studying FGF1's role in metabolism?

When investigating FGF1's metabolic effects, researchers should consider:

Model selection:

  • Diabetic models (db/db, ob/ob mice or Zucker diabetic fatty rats) are appropriate for studying glucose regulation

  • Diet-induced obesity models can reveal effects in metabolic syndrome contexts

Administration routes:

  • Intracerebroventricular (i.c.v.) injection into the lateral ventricle can induce long-term reduction in hyperglycemia (up to 18 weeks)

  • Direct injection into specific brain nuclei like the arcuate nucleus (ARH) can be sufficient to reduce hyperglycemia

Neuronal targeting:

  • Focus on proopiomelanocortin (POMC) neurons in the ARH, which are activated by FGF1

  • In contrast, neuropeptide Y (NPY) neurons in the ARH do not respond to FGF1, but NPY neurons in the nucleus tractus solitarius (NTS) do respond

Readouts:

  • Blood glucose measurements over extended time periods

  • Food intake monitoring

  • Expression of metabolic genes

  • Mitochondrial function parameters (fragmentation, ROS generation, respiration rate, β-oxidation)

How can researchers effectively study the heparin-dependency of FGF1 functions?

The interaction between FGF1 and heparin is critical for many of its functions:

Experimental approaches:

  • Compare activities with and without heparin supplementation

  • Use FGF1 variants with modified heparin-binding sites (e.g., FGF1 ΔHBS) to distinguish heparin-dependent and independent functions

  • Study differential effects on proliferation versus metabolic activities (FGF1 ΔHBS retains metabolic effects while showing reduced proliferative potency)

Relevant assays:

  • Receptor binding and activation with/without heparin

  • Cell proliferation and metabolic assays in parallel

  • In vivo studies comparing wild-type FGF1 and FGF1 ΔHBS

Key findings to consider:

  • FGF1 ΔHBS prevents diabetes-induced cardiac injury and remodeling through AMPK/Nur77-dependent mechanisms

  • The favorable metabolic activity combined with reduced proliferative properties makes FGF1 ΔHBS a promising candidate for treating metabolic disorders

How can FGF1 be engineered for enhanced therapeutic properties?

Based on current research, several strategies have proven successful:

Chimeric protein design:

  • Structure-based chimeras combining FGF1 with other FGF family members can enhance stability and function

  • For example, replacing the core of FGF21 with a thermally stable paracrine FGF1 (sFGF1) creates a chimera with increased stability and enhanced antidiabetic activities

Functional domain modification:

  • Creating variants with modified heparin-binding sites (FGF1 ΔHBS) reduces proliferative potential while maintaining beneficial metabolic effects

Validation approaches:

  • Differential scanning calorimetry to confirm increased thermal stability

  • Fluorescence spectroscopy and circular dichroism to verify proper protein folding

  • Functional assays comparing wild-type and engineered variants

  • In vivo testing in appropriate disease models

What approaches should be used to study FGF1's neuronal activation mechanisms?

FGF1 activates specific neuronal populations through mechanisms that require careful experimental design:

Neuronal subtype targeting:

  • Use transgenic reporter mice (NPY-GFP, POMC-EGFP) to identify specific neuronal populations

  • Focus on POMC neurons in the arcuate nucleus, which are depolarized by FGF1 (100 nM)

  • Note that NPY neurons in the arcuate nucleus do not respond to FGF1, but NPY neurons in the NTS do respond

Electrophysiological approaches:

  • Whole-cell patch-clamp recordings to measure membrane potential changes

  • Test the effects of tetrodotoxin (TTX) to determine if effects require voltage-gated sodium channels

  • Compare responses in healthy versus metabolically compromised models (e.g., diet-induced obesity)

Activation mapping:

  • c-Fos immunohistochemistry to map neuronal activation patterns following FGF1 administration

  • Focus on key brain regions including arcuate nucleus, area postrema (AP), and nucleus tractus solitarius (NTS)

How should researchers investigate intracellular FGF1 binding partners?

For studying FGF1's intracellular interactions, use these methodological approaches:

Protein interaction discovery methods:

  • Yeast two-hybrid screening (use FGF1 aa 22-155 as bait)

  • Tandem Affinity Purification (TAP) followed by mass spectrometry analysis

  • Co-precipitation from cell lysates using recombinant FGF1 as bait

Interaction validation:

  • Pull-down assays with recombinant proteins

  • Surface Plasmon Resonance (SPR) measurements to confirm direct interactions

  • Co-immunoprecipitation from cells expressing tagged versions of the proteins

Known binding partners to consider:

  • Previously identified: FGF-BP1, protein kinase CK2, FGF1 intracellular binding protein (FIBP), mortalin (GRP75/hthsp70/PB74), and p34

  • Twenty additional novel intracellular proteins have been identified, many involved in apoptosis, cell cycle regulation, and proliferation

What are the critical factors affecting FGF1 stability and activity in experimental settings?

Storage and handling considerations:

  • Store at -80°C for long-term storage

  • Avoid repeated freeze-thaw cycles by preparing single-use aliquots

  • Use low-protein binding tubes to prevent loss through adsorption

  • Include stabilizers in storage buffer (specific formulations may vary)

Activity-preserving factors:

  • Heparin supplementation for receptor-dependent activities

  • Temperature control during experiments (particularly important as FGF1 has thermal stability considerations)

  • Proper pH maintenance (typically physiological range)

  • Use of carrier proteins for dilute solutions may prevent loss of activity

Validation methods:

  • Periodically test activity in established assays (cell proliferation, receptor activation)

  • Verify protein integrity by SDS-PAGE

  • Use positive controls from previously validated batches

How can researchers distinguish between direct and indirect effects of FGF1?

Experimental strategies:

  • Time-course studies (direct effects typically occur rapidly, within minutes to hours)

  • Pathway inhibition (use specific inhibitors of known FGF1 signaling pathways)

  • Receptor blocking (FGFR inhibitors or neutralizing antibodies)

  • Use of FGF1 variants with altered receptor binding properties (e.g., FGF1 ΔHBS)

  • Transcription/translation inhibitors to determine if effects require new gene expression

Neuronal studies specific approaches:

  • Use tetrodotoxin (TTX) to block action potentials and determine if effects are direct or require neuronal network activity

  • Compare responses of different neuronal populations (e.g., POMC vs NPY neurons)

  • Use cell-type specific receptor knockouts to confirm direct targeting

What are the key considerations when investigating FGF1's role in diabetic cardiomyopathy?

Based on research with FGF1 ΔHBS , critical methodological considerations include:

Model selection:

  • Use established diabetic models (db/db mice, ob/ob mice, Zucker diabetic fatty rats)

  • Consider using AMPK null mice as controls to verify pathway involvement

Cardiac function assessment:

  • Echocardiography to measure fraction shortening and other functional parameters

  • Hemodynamic measurements to assess cardiac performance

Molecular analyses:

  • RNA-Seq to identify gene expression changes in cardiac tissues

  • Focus on anti-oxidative genes and Nur77 expression

  • Western blotting for AMPK activation and downstream targets

Mitochondrial function assessment:

  • Measure mitochondrial fragmentation

  • Quantify reactive oxygen species (ROS) generation

  • Assess cytochrome c leakage

  • Determine mitochondrial respiration rate and β-oxidation capacity

Clinical relevance:

  • Consider correlations with human data (serum FGF1 levels have been found to positively correlate with fraction shortening in diabetic cardiomyopathy patients)

What emerging applications of FGF1 warrant further investigation?

Metabolic disease therapeutics:

  • Long-lasting glycemic control following central administration

  • Potential applications in type 2 diabetes treatment using FGF1 variants with reduced proliferative potency

  • Further exploration of brain-periphery connections in FGF1-mediated metabolic effects

Cardiac protection:

  • Mechanisms of FGF1 ΔHBS in preventing diabetes-induced cardiac injury and remodeling

  • Application to other cardiomyopathies beyond diabetic etiology

Neuronal regulation:

  • Further characterization of FGF1's effects on specific neuronal populations

  • Investigation of potential applications in appetite regulation and energy balance

Engineered FGF1 variants:

  • Development of additional chimeric proteins with enhanced stability and function

  • Creation of tissue-specific targeting variants to reduce off-target effects

What technologies will advance FGF1 research in the coming years?

Emerging methodologies:

  • Single-cell transcriptomics to identify cell-specific responses to FGF1

  • CRISPR-based approaches for receptor and pathway component manipulation

  • Advanced imaging techniques for tracking FGF1 signaling in real-time

  • In silico protein engineering to design optimized FGF1 variants

Data integration approaches:

  • Multi-omics analysis combining transcriptomics, proteomics, and metabolomics

  • Systems biology modeling of FGF1 signaling networks

  • Machine learning for prediction of FGF1 binding partners and pathway interactions

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 2025 TheBiotek. All Rights Reserved.