FGF17 Human, His is produced via recombinant DNA technology in E. coli, followed by chromatographic purification.
Parameter | Specification |
---|---|
Expression Yield | High-yield soluble and inclusion body forms (up to 1 mg/mL post-purification) |
Bioactivity | EC₅₀ = 1.495–1.639 µg/mL (NIH-3T3 cell proliferation) |
Endotoxin Levels | <1 EU/µg (standard for research-grade proteins) |
FGF17 signals through FGF receptors (FGFRs), particularly FGFR3 and FGFR4, to activate mitogen-activated protein kinase (MAPK) pathways .
Memory Restoration: FGF17 infusion reverses age-related cognitive decline by inducing oligodendrogenesis in mice .
Parkinson’s Disease: FGF17-patterned dopaminergic progenitors rescued motor deficits in rat models .
Prostate Cancer: Overexpressed in high-grade tumors (Gleason score 7–10), correlating with poor prognosis .
Therapeutic Target: FGF17 blockade inhibits tumor growth in vitro (IC₅₀ < 10 ng/mL for prostate cancer cells) .
Mesenchymal Stem Cells (MSCs): Enhances proliferation of hypoxic Wharton’s jelly-derived MSCs via ERK1/2 signaling .
Multiple Sclerosis (MS): Promotes remyelination via oligodendrocyte progenitor activation .
Neurodegeneration: Synergizes with FGF8 to enhance synaptogenesis in preclinical models .
While FGF17 Human, His shows promise, challenges include:
Stability: Sensitivity to pH and temperature during reconstitution .
Specificity: Overlap with FGF8/18 signaling complicates mechanistic studies .
Current research focuses on optimizing expression systems (e.g., tag-free variants) and exploring combinatorial therapies with FGF17 agonists .
Human FGF17 is a member of the fibroblast growth factor family, playing multiple roles in biological functions including angiogenesis, mitogenesis, cell differentiation, and wound repair. FGF17 signals through multiple FGF receptors including FGFR1c, FGFR2c, FGFR3c, and FGFR4 .
Research has demonstrated that FGF17 activates the FGF receptor 3/PI3K/AKT signaling pathway, which is particularly important in protecting against ischemia/reperfusion-induced blood-brain barrier disruption and endothelial cell apoptosis . Treatment with recombinant human FGF17 (rhFGF17) leads to nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation and upregulation of heme oxygenase-1 (HO-1) expression, suggesting involvement in antioxidant response pathways .
FGF17 belongs to the FGF8 subfamily and shares high sequence homology with FGF8 and FGF18. Human and mouse FGF17 share 93% identity, indicating high evolutionary conservation . The protein consists of 195 amino acid residues with a molecular weight of approximately 22.7 kDa .
Unlike some other FGF family members with multiple isoforms, human FGF17 has just two isoforms: FGF17a and FGF17b, with FGF17b considered the canonical sequence . This contrasts with mouse FGF17, which has three isoforms .
A distinguishing functional characteristic of FGF17 is its potency as a mitogen in certain contexts. Studies have shown that low-dose recombinant FGF17 (1 ng/ml) is a more potent mitogen than rFGF1 and rFGF8 in prostate cancer cell lines (LNCaP, DU145, and PC3M) .
For optimal FGF17 activity in cell-based assays, researchers should consider the following parameters based on published studies:
When reconstituting lyophilized recombinant FGF17, it's recommended to:
For carrier-containing protein: Reconstitute at 25 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin .
For carrier-free protein: Reconstitute at 100 μg/mL in sterile PBS .
Validating FGF17 activity requires multiple complementary approaches:
Proliferation assays: Measure cell proliferation using:
Signaling pathway activation: Western blotting for:
Functional assays for specific contexts:
When expressing recombinant human FGF17, researchers may encounter several technical challenges:
Protein solubility issues: The yield of soluble rhFGF17 in E. coli expression systems may be limited by inclusion body formation .
Maintaining biological activity: Ensuring proper folding of the protein, especially after purification from inclusion bodies, is critical for maintaining biological activity .
Purity considerations: His-tagged FGF17 purification requires optimization to achieve sufficient purity while maintaining activity.
Storage stability: FGF17 requires careful handling to avoid freeze-thaw cycles. It's recommended to use a manual defrost freezer and avoid repeated freeze-thaw cycles .
Heparin dependency: Like other FGFs, FGF17 activity is modulated by heparin, which should be included in activity assays at concentrations between 1-10 μg/mL depending on the experimental system .
FGF17 shows significant upregulation in prostate cancer with strong correlation to disease aggressiveness. Research has revealed:
Expression correlation with disease severity:
A significant linear correlation exists between increasing Gleason sum scores and FGF17 expression using both immunohistochemistry (p < 0.0001, rho = 0.99) and RT-PCR (p = 0.008, rho = 0.99)
High-grade prostate cancer (Gleason sum score 7-10) shows approximately fourfold upregulation of FGF17 mRNA expression compared to benign prostatic hyperplasia (p < 0.0001)
Patients with tumors displaying high FGF17 expression have worse survival outcomes (p = 0.044) and higher risk of metastatic progression (p < 0.0001)
Methodological approaches for studying FGF17 in cancer:
Semi-quantitative RT-PCR to assess mRNA expression levels in tumor vs. normal tissue
Immunohistochemistry for protein-level analysis with correlation to clinicopathological parameters
In vitro proliferation assays comparing FGF17 potency to other FGFs
Analysis of cross-talk between FGF17 and FGF8, as FGF8 induces expression of FGF17 in prostate cancer cell lines
Experimental models:
Recent research has uncovered FGF17's neuroprotective role in cerebral ischemia, providing several methodological approaches to study its therapeutic potential:
Observed pathophysiological changes:
Therapeutic effects of rhFGF17 administration:
In vitro experimental approaches:
Molecular mechanism studies:
When facing contradictory results regarding FGF17 function, consider these methodological approaches to reconciliation:
Context-dependent effects analysis:
Systematically compare cell types, as FGF17 effects may vary due to differential receptor expression
Evaluate developmental timing influences, as FGF17 functions are stage-specific
Assess disease state context (normal vs. pathological conditions)
Concentration-dependent response profiling:
Experimental design harmonization:
Isoform specification:
Clearly identify which FGF17 isoform (FGF17a or FGF17b) is being studied
Consider potential differential functions between isoforms
Interaction analysis with related FGFs:
FGF17 and FGF8 exhibit significant cross-talk with important experimental design implications:
Known interactions:
Optimal experimental designs for cross-talk studies:
Sequential stimulation experiments: Treat cells with FGF8 first, then measure FGF17 expression kinetics
Knockdown studies: Use siRNA against FGF17 to determine which FGF8 effects are FGF17-dependent
Co-immunoprecipitation: Assess potential protein-protein interactions
Receptor competition assays: Determine if pre-treatment with one FGF affects binding or signaling of the other
Critical controls and considerations:
Include dose-response analyses for both factors individually before combination studies
Validate antibody specificity due to high sequence homology
Consider temporal dynamics, as some effects may be immediate while others require new protein synthesis
Measure both mRNA and protein levels to distinguish transcriptional from post-transcriptional effects
Differentiating FGF17's unique contributions presents several analytical challenges requiring sophisticated methods:
Spatiotemporal expression mapping challenges:
Functional redundancy assessment:
Conditional and inducible knockout models are needed to bypass developmental lethality
Domain-swap experiments between FGF17 and related FGFs can identify unique functional domains
Receptor-specific blocking antibodies can help determine which receptor interactions are unique to FGF17
Technical considerations for specific developmental contexts:
For midbrain/hindbrain junction studies, ex vivo explant cultures with localized FGF17 application
For vascular development, endothelial-specific conditional expression systems
For skeletal development, mesenchymal stem cell differentiation assays with FGF17 supplementation
Data integration approaches:
Multi-omics analysis combining transcriptomics, proteomics, and phosphoproteomics
Systems biology modeling of FGF signaling networks
Machine learning algorithms to identify FGF17-specific gene expression signatures
Despite significant progress, several knowledge gaps remain in FGF17 biology that require innovative approaches:
Structural biology of FGF17-receptor complexes:
Need for crystal structures of FGF17 bound to different FGFRs
Comparison with FGF8-receptor complexes to identify specificity determinants
Application of cryo-EM for visualizing signaling complexes in native-like environments
Isoform-specific functions:
Differentiation between FGF17a and FGF17b functions remains poorly characterized
Development of isoform-specific antibodies and expression constructs
CRISPR-based isoform-specific genome editing approaches
Tissue-specific roles beyond currently known functions:
FGF17's role in tissues where expression has been documented but function is unclear
Development of tissue-specific inducible expression systems
Spatial transcriptomics to map FGF17 effects with high anatomical resolution
Long-term physiological impacts of FGF17 dysregulation:
Longitudinal studies in conditional knockout or overexpression models
Integration of metabolomics with transcriptomics for comprehensive phenotyping
Development of non-invasive imaging methods to track FGF17-dependent processes in vivo
Current research suggests several promising therapeutic applications for FGF17 modulation:
Neuroprotection in ischemic stroke:
Cancer therapy approaches:
Methodological considerations for therapeutic development:
Pharmacokinetic and pharmacodynamic profiling of rhFGF17
Biomarker development to identify patients likely to respond to FGF17-targeted therapies
Consideration of FGF8-FGF17 relationships when targeting either factor
Development of controlled-release formulations to optimize biological effects
The human recombinant FGF17 protein, tagged with a polyhistidine (His) tag, is typically expressed in E. coli cells . The His tag facilitates purification and detection of the protein. The recombinant protein contains amino acids 23 to 216 of the native FGF17 sequence, with a calculated molecular weight of approximately 24.5 kDa . When analyzed under non-reducing conditions using SDS-PAGE, the protein migrates as a 28-30 kDa band .
The recombinant FGF17 protein is highly purified, with a purity greater than 95% as determined by SDS-PAGE and greater than 90% as determined by size-exclusion chromatography (SEC-HPLC) . The endotoxin level is less than 0.1 EU per microgram, ensuring minimal contamination . Additionally, the protein is tested for host cell protein and DNA contamination, with levels below 0.5 ng/µg and 0.02 ng/µg, respectively .
FGF17 is known to stimulate the proliferation of NIH-3T3 cells, with an EC50 value ranging from 1.495 to 1.639 µg/mL . This bioactivity is crucial for its role in various cellular processes and developmental stages. FGF17, along with FGF8, is a key factor in the patterning of the mid-hindbrain region during cerebellar development .
Recombinant FGF17 protein is widely used in research to study its role in cellular processes and development. It is also used in various assays to investigate its interactions with other proteins and receptors. The His tag allows for easy purification and detection, making it a valuable tool in biochemical and cell biology research .
For long-term storage, the lyophilized FGF17 protein should be kept at -20°C or lower to maintain its stability . After reconstitution, the protein can be stored at -70°C for up to three months under sterile conditions . It is important to avoid repeated freeze-thaw cycles to preserve the protein’s integrity .