Recombinant Human Fibroblast growth factor 9 (FGF9) (Active)

Shipped with Ice Packs
In Stock

Description

Molecular Structure

Recombinant Human Fibroblast Growth Factor 9 is a full-length human protein that typically spans amino acids 3 to 208, expressed in various systems including Escherichia coli with high purity levels exceeding 95% . The protein belongs to the heparin-binding growth factors family, which shares structural motifs related to their interaction with heparin and heparan sulfate proteoglycans . Interestingly, FGF9 exists in multiple molecular species of 30 kDa, 29 kDa, and 25 kDa, which are cleaved at different positions (Leu-4, Val-13, and Ser-34 respectively) . These variants may represent distinct functional forms, with the smaller variants potentially resulting from proteolytic processing.

Production Systems

The production of recombinant FGF9 employs several expression systems. While bacterial expression in E. coli represents one common approach, insect cell-based expression systems using Spodoptera frugiperda (Sf21) with baculovirus vectors offer advantages for certain applications . These different production platforms can affect post-translational modifications and protein folding, potentially impacting the biological activity of the final product. Commercial preparations typically achieve endotoxin levels below 1 EU/μg, making them suitable for cell culture applications and functional studies .

Developmental Roles

FGF9 plays crucial roles in embryonic development, serving as a regulatory molecule that orchestrates cell proliferation, differentiation, and migration during tissue formation . Its signaling contributes to the proper development of multiple organ systems, with particularly notable effects in neural and glial development. The protein may have specialized functions in glial cell growth and differentiation during developmental processes, suggesting its importance in establishing functional neural circuitry .

Cellular Signaling Mechanisms

The biological activities of FGF9 are mediated through interaction with fibroblast growth factor receptors (FGFRs), triggering downstream signaling cascades. FGF9 induces the activation of multiple pathways, including:

  1. ERK (Extracellular signal-Regulated Kinase) pathway

  2. JNK (c-Jun N-terminal Kinase) pathway

  3. p38 MAPK (Mitogen-Activated Protein Kinase) pathway

  4. PLCγ (Phospholipase C gamma) pathway

These pathways are rapidly activated following FGF9 exposure, with phosphorylation events detectable within 15-30 minutes of treatment . The p-ERK1/2 activation shows sustained effects for up to 6 hours, while p-JNK activation persists for approximately 1 hour after FGF9 treatment . This temporal regulation of signaling pathways contributes to the diverse cellular responses elicited by FGF9.

Cellular Proliferation

Recombinant FGF9 demonstrates potent mitogenic activity across multiple cell types. In standardized bioactivity assays using Balb/3T3 mouse embryonic fibroblast cells, FGF9 induces proliferation with an ED₅₀ (effective dose for 50% response) of 1-5 ng/mL . This proliferative effect has been extensively documented in dose-response studies, with concentrations ranging from 10-100 ng/mL showing significant and dose-dependent enhancement of cell proliferation over 24-72 hour periods . The proliferative effects appear to be mediated primarily through the activation of MAPK and PI3K signaling cascades.

Neural Tissue Repair

FGF9 participates in repair and regenerative processes, particularly following tissue damage. In neural tissues, FGF9 contributes to gliosis during repair and regeneration of brain tissue after injury . This process involves the proliferation and migration of glial cells to the site of damage, followed by their participation in forming a supportive environment for neuronal recovery. Additionally, FGF9 appears to support the differentiation and survival of neuronal cells, suggesting a neuroprotective role that could be therapeutically valuable .

Hepatic Function and Metabolism

Recent investigations have revealed unexpected roles for FGF9 in hepatic metabolism and function. FGF9 appears to regulate hepatic lipid metabolism through multiple mechanisms, affecting both lipogenesis and fatty acid oxidation pathways . Experimental evidence indicates that FGF9 overexpression in hepatocytes reduces cellular lipid accumulation, while its knockdown increases triglyceride content . These effects are mediated through the regulation of key metabolic genes:

Effect of FGF9Lipid Synthesis GenesFatty Acid Transport GenesLipid Oxidation Genes
OverexpressionDecreases ChREBP, Fasn, PPARγDecreases CD36Increases Cpt1a, CYP4A10, CYP4A14
KnockdownIncreases ChREBP, Fasn, PPARγIncreases Fabp1, Fabp4, CD36Decreases Cpt1a, CYP4A10, CYP4A14

These findings suggest that FGF9 may function as a metabolic regulator in hepatocytes, potentially affecting systemic metabolism .

Tumor Biology

FGF9 has emerged as a significant factor in tumor microenvironments, particularly in hepatocellular carcinoma (HCC). Notably, FGF9 is primarily expressed by hepatic stellate cells and stromal myofibroblasts rather than the cancer cells themselves . This pattern suggests a paracrine signaling mechanism in the tumor microenvironment, where stromal-derived FGF9 influences cancer cell behavior. High expression levels of FGF9 in HCC tissues correlate with poor patient survival, indicating its potential value as a prognostic marker .

Cancer Cell Behavior and Drug Resistance

Recombinant FGF9 treatment significantly enhances several aspects of cancer cell behavior that contribute to tumor progression. These include:

  1. Increased proliferation

  2. Enhanced clonogenicity (colony-forming ability)

  3. Accelerated cell migration

Metabolic Disorders

FGF9 appears to play a protective role in metabolic disorders, particularly non-alcoholic fatty liver disease (NAFLD). In mouse models of diet-induced obesity, FGF9 expression increases in the liver, potentially as a compensatory mechanism . Experimental overexpression of FGF9 in the livers of diet-induced obese mice produces multiple beneficial effects:

  1. Decreased liver weight-to-body weight ratio

  2. Improved glucose tolerance

  3. Enhanced insulin sensitivity

  4. Reduced hepatic triglyceride content

  5. Lower serum ALT and AST levels (markers of liver injury)

These findings suggest that FGF9 may represent a potential therapeutic target for metabolic disorders, particularly those affecting hepatic lipid metabolism .

Standardization and Quality Control

Commercial preparations of Recombinant Human FGF9 undergo rigorous quality control testing to ensure consistency and reliability. These typically include:

  1. Purity assessment by SDS-PAGE

  2. Endotoxin testing (typically <1 EU/μg)

  3. N-terminal sequence analysis

  4. Bioactivity testing in standardized cell proliferation assays

These quality control measures are essential for research applications, particularly when investigating dose-dependent cellular responses or developing therapeutic applications .

Research Applications

Recombinant Human FGF9 serves as a valuable research tool across multiple disciplines. Its applications include:

  1. Studies of embryonic development and tissue patterning

  2. Investigation of cellular signaling mechanisms

  3. Cancer biology research, particularly in understanding tumor-stroma interactions

  4. Metabolic research focused on hepatic lipid metabolism

  5. Neurobiological studies of glial function and neural repair

The availability of highly purified, well-characterized recombinant protein has accelerated research in these fields by providing consistent experimental reagents .

Anti-Cancer Strategies

The involvement of FGF9 in cancer progression, particularly in HCC, suggests potential therapeutic approaches targeting this signaling pathway. The finding that FGFR1/2/3 inhibitors can block the protumorigenic effects of FGF9 presents a promising avenue for drug development . Since high FGF9 expression correlates with poor prognosis in HCC patients, it may serve as both a prognostic marker and a potential therapeutic target. Combination therapies involving FGFR inhibitors alongside traditional chemotherapeutics may help overcome drug resistance mechanisms in certain cancers.

Metabolic Disease Applications

The beneficial effects of FGF9 on hepatic lipid metabolism present exciting possibilities for treating metabolic disorders, particularly NAFLD and potentially type 2 diabetes . The ability of FGF9 to improve glucose tolerance, enhance insulin sensitivity, and reduce hepatic steatosis in animal models suggests significant therapeutic potential. Future research may explore recombinant FGF9 delivery systems or small molecule activators of the FGF9 pathway as potential treatments for these increasingly prevalent metabolic conditions.

Neural Regeneration

The involvement of FGF9 in glial cell function, neuronal survival, and tissue repair suggests potential applications in neurological injury and neurodegenerative diseases . While still in early stages, research into FGF9's neuroprotective and neuroregenerative properties may yield new approaches for conditions with limited treatment options, such as traumatic brain injury, stroke, or certain neurodegenerative disorders.

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered solution containing 20 mM phosphate buffer, 220 mM sucrose, 0.02% Tween 80, adjusted to pH 6.0.
Form
Lyophilized powder
Lead Time
Typically, we can ship products within 5-10 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery information.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
Prior to opening, we recommend briefly centrifuging the vial to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our default final glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer components, temperature, and the inherent stability of the protein.
Generally, the shelf life of liquid formulations is 6 months at -20°C/-80°C. For lyophilized forms, the shelf life 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
FGF 9; FGF-9; FGF9; FGF9_HUMAN; Fibroblast growth factor 9; GAF (Glia-activafibroblast growth factor 9 (glia-activating factor); GAF; Glia Activating Factor; Glia-activating factor; HBFG 9; HBFG9; HBGF-9; Heparin-binding growth factor 9; MGC119914; MGC119915; SYNS3
Datasheet & Coa
Please contact us to get it.
Expression Region
1-208aa
Mol. Weight
23.44 kDa
Protein Length
Full Length
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor 9 (FGF9) plays a critical role in regulating embryonic development, cell proliferation, differentiation, and migration. It is implicated in various biological processes, including glial cell growth and differentiation during development, gliosis during brain tissue repair and regeneration after damage, differentiation and survival of neuronal cells, and growth stimulation of glial tumors.
Gene References Into Functions
  1. Our research suggests that miR-4317 can inhibit Non-small cell lung cancer (NSCLC) cell growth and metastasis by targeting FGF9 and CCND2. These findings highlight the potential of miR-4317 as a non-invasive biomarker and therapeutic target for NSCLC. PMID: 30227870
  2. We have demonstrated for the first time that mutations in FGF9 cause craniosynostosis in humans, confirming that FGF9 mutations cause multiple synostoses. PMID: 28730625
  3. Forced expression of miR-187 inhibited the subcutaneous growth of cervical cancer cells in nude mice. Furthermore, FGF9 was identified as a downstream target of miR-187 in cervical cancer cells. PMID: 28849071
  4. Results indicate that homodimerization controls receptor binding specificity of FGF9 by maintaining the concentration of active FGF9 monomers at a level sufficient for normal FGFR "c" isoform binding/signaling, but insufficient for illegitimate FGFR "b" isoform binding. Mutations in FGF9 N or C-terminus shift the ligand equilibrium towards active monomers, leading to off-target binding and activation of FGFb. PMID: 28757146
  5. Upregulation of FGF9 or downregulation of miR-372-3p significantly reduced lung squamous cell carcinoma (LSCC) cell growth, mitosis, and invasion. MiR-372-3p promoted LSCC cell proliferation and invasion by inhibiting FGF9. PMID: 28440022
  6. Our data suggest that fibroblast growth factor 9 (FGF9) may provide anti-apoptotic function and serve as a novel independent marker for evaluating gastric cancer (GC) prognosis. PMID: 27166269
  7. CCND1 mRNA expression is increased by FGF9 in bovine theca cells and granulosa cells. PMID: 27816766
  8. The present data suggest that non-natural FGFR2 ligands, such as FGF10 and FGF19, are crucial factors in the pathophysiology of Aspert syndrome. PMID: 27339175
  9. In FGF9-overexpressing colorectal cancer cell lines, FGF9 overexpression induced strong resistance to anti-EGFR therapies through the enforced FGFR signal. This resistance was reversed by the application of an FGFR inhibitor. PMID: 26916220
  10. FGF9 and FGF18 enhanced the migratory capacities of human lung fibroblasts, and FGF9 actively modulated matrix metalloproteinase activity in idiopathic pulmonary fibrosis. PMID: 26773067
  11. Our findings indicate that the S99N mutation in Fgf9 causes multiple synostoses syndrome (SYNS) through the disruption of joint interzone formation. These results further highlight the critical role of Fgf9 during embryonic joint development. PMID: 28169396
  12. These studies identify FGF9 as a target of DICER1 in lung epithelium that functions as an initiating factor for pleuropulmonary blastoma. PMID: 25978641
  13. Data indicate that expressing either human FGF9 in the kidney subcapsular space of female BALB/c mice resulted in rapidly expanding local tumors. PMID: 26183774
  14. FGF9 was strongly expressed in CAFs compared to NGFs, consistent with microarray data suggesting that FGF9 is a novel growth factor overexpressed in Cancer-associated fibroblasts. PMID: 25925261
  15. The relative levels of FGF9 compared to other members of the FGF family may be key to understanding vulnerability or resilience in affective disorders. PMID: 26351673
  16. Our data demonstrate that Kl treatment impairs Nodal mRNA expression and Fgf9-mediated Nanos2 induction, reinforcing the antagonistic effect of these two growth factors on the meiotic fate of male germ cells. PMID: 25766327
  17. Our findings indicate that FGF9 can initiate a complex astrocytic response predicted to compromise remyelination, while simultaneously stimulating microglial/macrophage recruitment in multiple sclerosis lesions. PMID: 25907862
  18. Expression is associated with poor prognosis in lung cancer. PMID: 24239165
  19. MAP3K1 mutations shift the balance in the sex-determining pathways by downregulating SOX9 and FGF9. PMID: 24135036
  20. The data demonstrates that FGF9 IRES functions as a cellular switch to activate FGF9 protein synthesis during hypoxia, a likely mechanism underlying FGF9 overexpression in cancer cells. PMID: 24334956
  21. FGF9 was proven to be a direct target of miR-26a. PMID: 24015269
  22. FGF9 can contribute to epithelial-to-mesenchymal transition and invasion by inducing VEGF-A expression in prostate cancer cells. PMID: 24511001
  23. In addition to its role in sex determination, FGF9 is expressed in postnatal Leydig cells and is involved in cell-to-cell interaction of testicular function. Aberrant expression of testicular FGF9 is associated with SCOS. PMID: 24011613
  24. The results demonstrate that FGF9 protein increased in regions of active cellular hyperplasia, metaplasia, and fibrotic expansion of idiopathic pulmonary fibrosis lungs. PMID: 23797050
  25. The importance of Fgf9 in hair follicle regeneration suggests its potential therapeutic use in humans. PMID: 23727932
  26. Neither DMRT1 nor FGF9 abnormalities are commonly involved in dysgenetic male gonad development in patients with non-syndromic 46,XY disorder of sex development. PMID: 22939835
  27. The FGF9(S99N) monomer preferentially binds with the FGFR3c receptor to form an inactive complex, leading to impaired FGF signaling. This impaired FGF signaling is considered a potential cause of synostoses syndrome, highlighting the crucial role of FGF9 in normal joint development. PMID: 22920789
  28. These results suggest that FGF9 can stimulate proliferation and invasion in prostate cancer cells, making FGF9 a potential predictive factor for recurrence after radical prostatectomy. PMID: 22006051
  29. Fibroblast growth factor 9 was also overexpressed in all serous ovarian tumors, with greater than 1000-fold increase in gene expression in 4 tumors. PMID: 21666490
  30. Microvessels formed in the presence of FGF9 exhibited enhanced capacity to receive flow and were vasoreactive. PMID: 21499246
  31. FGF9 serves as an autocrine estromedin endometrial stromal growth factor, playing roles in the cyclic proliferation of uterine endometrial stroma. PMID: 12072406
  32. The proliferative capability of endometriotic stromal cells during menstruation, when ovarian 17 beta-estradiol is at its lowest point, may be mediated, at least in part, by autocrine estrogen-stimulated expression of FGF-9 and its receptors. PMID: 14602803
  33. Recombinant human FGF-9 signaling enhances the intrinsic osteogenic potential by selectively expanding committed chick embryo osteogenic cell populations and inversely regulating bone morphogenetic protein 2 (BMP-2) and noggin gene expression. PMID: 15780951
  34. Mesothelial and epithelial transgenic FGF9 directs lung development by regulating mesenchymal growth, and the pattern and expression levels of mesenchymal growth factors that signal back to the epithelium. PMID: 16540513
  35. Our findings may also provide a molecular framework for considering roles for PGE2 in FGF-9-related embryonic development and/or human diseases. PMID: 16982695
  36. Polymorphic microsatellite in the 3'-UTR of FGF9 in patients with Gonadal dysgenesis. PMID: 17154280
  37. FGF9 mutant tumors exhibited normal membranous beta-catenin expression and the absence of mutation in the beta-catenin gene. PMID: 18165946
  38. Inhibition of fibroblast growth factor 19 reduces tumor growth by modulating beta-catenin signaling. PMID: 18593907
  39. Androgen receptor-negative human prostate cancer cells induce osteogenesis in mice through FGF9-mediated mechanisms. PMID: 18618013
  40. The study ruled out microdeletions on the critical region as a common cause of Moebius syndrome and excluded FGF9 gene. PMID: 19460469
  41. Data demonstrate that homodimerization autoregulates FGF9 and FGF20's receptor binding and concentration gradients in the extracellular matrix. PMID: 19564416
  42. Data demonstrate a previously uncharacterized mutation in FGF9 as one of the causes of Multiple synostoses syndrome, implicating an important role of FGF9 in normal joint development. PMID: 19589401

Show More

Hide All

Database Links

HGNC: 3687

OMIM: 600921

KEGG: hsa:2254

STRING: 9606.ENSP00000371790

UniGene: Hs.111

Involvement In Disease
Multiple synostoses syndrome 3 (SYNS3)
Protein Families
Heparin-binding growth factors family
Subcellular Location
Secreted.
Tissue Specificity
Glial cells.

Q&A

How is recombinant human FGF9 typically produced and what quality control parameters should researchers monitor?

Recombinant human FGF9 is predominantly produced using bacterial expression systems, most commonly in Escherichia coli, although some suppliers use insect cell expression systems such as Spodoptera frugiperda (Sf21) . For research applications, quality control should focus on:

Quality ParameterStandard SpecificationTesting Method
Purity>95%SDS-PAGE analysis
Endotoxin level<1.0 EU/μgLAL method
BioactivityED50 typically 1-5 ng/mLCell proliferation assay using Balb/3T3 cells
Protein concentrationVerified by reconstitutionSpectrophotometric methods
Molecular weight23.44 kDa (full-length)Mass spectrometry/SDS-PAGE

When evaluating recombinant FGF9 preparations, researchers should confirm these parameters in the certificate of analysis and perform additional functional validation through bioactivity assays relevant to their experimental system .

What signaling pathways does FGF9 activate and how can researchers effectively assess its functional activity?

FGF9 binds and activates the 'c' splice isoforms of FGF receptors 1-3 (FGFR1-3), with particular affinity for FGFR3 (IIIb) . Upon receptor binding, FGF9 activates several downstream signaling cascades:

  • ERK (Extracellular signal-regulated kinase) pathway

  • JNK (c-Jun N-terminal kinase) pathway

  • STAT (Signal transducer and activator of transcription) pathway

  • PI3K (Phosphoinositide 3-kinase) pathway

  • MAP kinase pathway

To assess FGF9 functional activity, researchers can employ several methodological approaches:

  • Proliferation assays: The standard bioactivity assay measures cell proliferation in Balb/3T3 mouse embryonic fibroblast cells, with an expected ED50 of 1-5 ng/mL .

  • Luciferase reporter assays: Serum response element (SRE) luciferase reporter assays in transfected HEK293T cells provide quantitative assessment of FGF9 activity (typical EC50 ≈ 246.2 pM or 5.7 ng/mL) .

  • Phosphorylation analysis: Western blotting for phosphorylated ERK, JNK, and other pathway components following FGF9 stimulation .

  • Clonogenicity assays: Measuring colony number and size in responsive cell lines (e.g., Hep3B, HepG2) .

  • Migration assays: Transwell Boyden chamber assays to assess directed migration of cells in response to FGF9 .

How should researchers properly reconstitute and store recombinant FGF9 to maintain optimal activity?

Proper handling of recombinant FGF9 is critical for maintaining its biological activity:

Reconstitution Protocol:

  • Centrifuge the vial briefly before opening to bring the contents to the bottom.

  • Reconstitute lyophilized FGF9 in deionized sterile water or sterile PBS to a concentration of 0.1-1.0 mg/mL.

  • For long-term storage stability, add 5-50% glycerol (final concentration) .

  • Allow the protein to sit for 10-15 minutes at room temperature to ensure complete solubilization.

Storage Recommendations:

  • Store reconstituted protein at -20°C or -80°C in small aliquots to avoid repeated freeze-thaw cycles.

  • For lyophilized protein, the recommended shelf life is 12 months at -20°C/-80°C.

  • For reconstituted protein in liquid form, the shelf life is approximately 6 months at -20°C/-80°C .

Researchers should note that different commercial preparations may have specific reconstitution buffers optimized for stability, such as "20 mM PB, 150 mM NaCl, 5% Trehalos, pH 7.4" or "20 mM PB, 220 mM Sucrose, 0.02% Tween 80, pH 6.0" . Always refer to the product-specific Certificate of Analysis for optimal handling conditions.

How does FGF9 contribute to hepatocellular carcinoma (HCC) progression and what experimental models best recapitulate its effects?

FGF9 plays a significant role in hepatocellular carcinoma (HCC) progression through several mechanisms:

  • Cellular source in HCC: FGF9 is primarily expressed by activated hepatic stellate cells (HSC) and cancer-associated myofibroblasts in the tumor microenvironment, not by HCC cells themselves .

  • Clinical correlation: High expression levels of FGF9 significantly correlate with poor patient survival, and FGF9 expression positively correlates with alpha-smooth muscle actin (alpha-sma) expression in HCC tissues .

  • Pro-tumorigenic effects: FGF9 enhances:

    • Cell proliferation through ERK and JNK pathway activation

    • Clonogenicity (reflecting stem cell properties and cell survival)

    • Migratory capacity of HCC cells

    • Therapeutic resistance, particularly to sorafenib

  • Receptor specificity: FGF9's effects on HCC cells are mediated primarily through FGFR1/2/3, as the selective FGFR4 inhibitor BLU9931 had no significant effect, while the FGFR1/2/3 inhibitor BGJ398 abrogated FGF9's pro-tumorigenic effects .

Recommended experimental models:

Model TypeMethodologyAdvantagesKey Measurements
HCC cell lines (Hep3B, HepG2, PLC)Stimulation with recombinant FGF9Simple, controlled environmentProliferation, clonogenicity, migration
Conditioned mediaCM from HSC with siRNA-mediated FGF9 suppressionModels paracrine signalingGrowth effects on HCC cells
3D spheroid modelsMixed HSC-HCC spheroidsRecapitulates cell-cell interactionsSpheroid size, invasive capacity
Patient-derived xenograftsImplantation of patient tumor fragments with FGF9 manipulationMost physiologically relevantTumor growth, metastasis, therapeutic response

For comprehensive analysis of FGF9's role in HCC, researchers should consider combining these models with analysis of signaling pathway activation (ERK, JNK) and therapeutic response assays .

What are the methodological considerations for using FGF9 in kidney organoid generation?

FGF9 is frequently used in the generation of kidney organoids due to its developmental roles. When incorporating FGF9 into kidney organoid protocols, researchers should consider:

  • Concentration optimization: Titrate FGF9 concentrations (typically starting in the 50-200 ng/mL range) to determine optimal dosing for your specific protocol and cell lines.

  • Timing of administration: FGF9 signaling is stage-specific during kidney development. Consider pulse treatments rather than continuous exposure to mimic developmental signaling dynamics.

  • Receptor expression verification: Confirm expression of appropriate FGF receptors (especially FGFR1-3) in your stem cell population before FGF9 treatment using RT-PCR or western blotting.

  • Combinatorial signaling: FGF9 often works in concert with other growth factors. Consider combining with:

    • WNT pathway modulators (e.g., CHIR99021)

    • BMP inhibitors (e.g., Noggin)

    • Activin/TGF-β pathway components

  • Functional readouts: Validate organoid maturation using:

    • Immunohistochemistry for nephron segment markers

    • Single-cell RNA sequencing to confirm appropriate cell populations

    • Functional assays (albumin uptake, fluid transport)

  • Stability considerations: Refresh FGF9-containing media every 1-2 days, as the protein may have limited stability at 37°C in complex media .

To enhance reproducibility, it's recommended to validate the bioactivity of each new lot of FGF9 using a standardized proliferation assay before application in organoid protocols.

How does FGF9 differ from other FGF family members in structure and function?

FGF9 belongs to a subfamily within the broader FGF family that includes FGF9, FGF16, and FGF20, which share 65-71% amino acid sequence identity . These key differences distinguish FGF9 from other FGF family members:

CharacteristicFGF9 Subfamily (FGF9, FGF16, FGF20)Other FGF Family Members
Signal sequenceUncleavable, bipartite signal sequenceTypically have conventional cleavable signal sequences
Secretion mechanismEfficiently secreted despite unusual signal sequenceVariable secretion efficiency
Receptor specificityBinds FGFR3 (IIIb) with high affinityVariable receptor preferences
DimerizationConstitutive dimerization that buries receptor interaction sitesMost are monomeric or form different types of dimers
Heparin bindingDimerization increases heparin affinity and inhibits diffusionVariable heparin binding properties
Developmental rolesNeural patterning, skeletal development, sex determinationDiverse developmental roles

A unique feature of FGF9 is its constitutive dimerization, which regulates its activity and diffusion properties. Mutations that interfere with dimerization (as in the mouse Eks mutant) result in monomeric, more diffusible FGF9 that causes joint fusions (synostoses) . In humans, FGF9 mutations affecting receptor binding cause multiple synostoses syndrome (SYNS) .

FGF9 is particularly relevant in glial cell biology, kidney development, and sex determination, while other FGF family members may have more prominent roles in other developmental processes .

What are the common troubleshooting approaches when recombinant FGF9 shows reduced or no activity in experimental systems?

When researchers encounter reduced or absent FGF9 activity, systematic troubleshooting is essential:

  • Protein Quality Issues:

    • Verification test: Analyze by SDS-PAGE under reducing and non-reducing conditions to check for degradation or aggregation.

    • Solution: Obtain a new lot of recombinant FGF9 with verified bioactivity.

  • Improper Reconstitution:

    • Verification test: Measure protein concentration after reconstitution.

    • Solution: Ensure protein is properly solubilized; avoid vortexing; consider adding carrier proteins (0.1% BSA) for very dilute solutions.

  • Receptor Expression:

    • Verification test: Perform RT-PCR or Western blot analysis for FGFR1-3 expression in your cell system.

    • Solution: Use a positive control cell line (e.g., Balb/3T3) to verify FGF9 activity in parallel.

  • Heparan Sulfate Proteoglycan (HSPG) Availability:

    • Verification test: Add exogenous heparin (1-10 μg/mL) to your assay.

    • Solution: If activity increases with heparin, your system may lack sufficient HSPGs for proper FGF9 signaling.

  • Downstream Signaling Pathway Integrity:

    • Verification test: Analyze ERK phosphorylation at 5-15 minutes after FGF9 treatment.

    • Solution: If no phosphorylation occurs, test other growth factors that utilize the same pathways to determine if the issue is FGF9-specific.

  • Handling and Storage Problems:

    • Verification test: Test bioactivity of a freshly reconstituted aliquot versus stored protein.

    • Solution: Store in smaller aliquots to minimize freeze-thaw cycles; add stabilizers (e.g., 0.1% BSA) for dilute solutions.

  • Experimental Design Factors:

    • Verification test: Review cell density, serum starvation conditions, and timing of FGF9 addition.

    • Solution: Optimize serum starvation (12-24h), cell density (50-70% confluence), and duration of FGF9 treatment.

Systematic testing of these parameters will help identify the source of reduced activity and guide appropriate corrective actions to restore experimental functionality.

How can FGF9 be utilized in investigating the tumor microenvironment and developing targeted cancer therapies?

FGF9's role in the tumor microenvironment, particularly in HCC, offers several research avenues for developing targeted cancer therapies:

  • Targeting the HSC-Cancer Cell Axis:

    • FGF9 is expressed by activated hepatic stellate cells (HSC) and cancer-associated myofibroblasts but not by HCC cells themselves .

    • This creates an opportunity to disrupt the paracrine signaling between stromal cells and cancer cells.

    • Research methodology: Use co-culture systems of HSCs and HCC cells with selective inhibition of FGF9 production or signaling.

  • FGF9 as a Prognostic Biomarker:

    • High FGF9 expression correlates with poor patient survival in HCC .

    • Research methodology: Analyze FGF9 expression in patient biopsies using immunohistochemistry and correlate with clinical outcomes.

  • Combinatorial Therapy Approaches:

    • FGF9 contributes to sorafenib resistance in HCC .

    • Research methodology: Test combinations of FGFR inhibitors (particularly targeting FGFR1/2/3) with sorafenib in preclinical models.

  • Novel Targeting Strategies:

    • FGF traps or anti-FGF9 antibodies could be developed to specifically target FGF9-mediated signaling .

    • Research methodology: Design and test FGF9-specific traps or antibodies in vitro and in animal models.

  • MicroRNA-Based Regulation:

    • miR-140-5p has been identified as a tumor suppressor in HCC that may act partly through suppressing FGF9 .

    • Research methodology: Investigate miRNA-based therapies targeting FGF9 expression.

Experimental approaches should incorporate these methodologies with appropriate controls and validation in multiple model systems, progressing from in vitro studies to animal models and ultimately clinical samples.

What are the latest techniques for studying FGF9's role in developmental processes and tissue engineering applications?

Advanced methodologies for investigating FGF9's developmental roles and applications in tissue engineering include:

  • CRISPR/Cas9 Gene Editing:

    • Precise modification of FGF9 or its receptors in model organisms and stem cells

    • Methodology: Design guide RNAs targeting specific FGF9 domains to create functional mutants that affect specific aspects of FGF9 activity.

  • Spatiotemporal Control of FGF9 Signaling:

    • Optogenetic approaches to control FGF9 signaling with light-inducible systems

    • Methodology: Engineer light-responsive FGFR systems that activate upon illumination, allowing precise temporal and spatial control.

  • Biomaterial-Based Delivery Systems:

    • Controlled release of FGF9 using advanced biomaterials

    • Methodology: Incorporate FGF9 into hydrogels, microspheres, or nanoparticles designed for sustained or triggered release in specific microenvironments.

  • Organ-on-Chip Technologies:

    • Microfluidic systems mimicking developmental microenvironments

    • Methodology: Create multi-chamber devices with controlled gradients of FGF9 to study migration, differentiation, and morphogenesis in real-time.

  • Single-Cell Analysis:

    • Profiling FGF9 effects on heterogeneous cell populations

    • Methodology: Apply single-cell RNA-seq to identify cell-type-specific responses to FGF9 signaling during development or disease progression.

  • 3D Bioprinting with FGF9 Incorporation:

    • Spatial patterning of FGF9 in engineered tissues

    • Methodology: Develop bioinks containing FGF9 or FGF9-expressing cells for specific spatial distribution in printed constructs.

These advanced techniques allow researchers to better recapitulate the complexity of developmental processes and create more sophisticated tissue engineering applications leveraging FGF9's biological activities in cell proliferation, differentiation, and tissue patterning.

How can researchers optimize experiments involving FGF9's role in neuronal development and neuroprotection?

FGF9 plays important roles in neuronal development, differentiation, survival, and has been implicated in neuroprotection against neurodegenerative diseases . When designing experiments to study these functions, researchers should consider:

  • Neural Cell Type Specificity:

    • FGF9 affects different neural cell populations distinctly (neurons, astrocytes, oligodendrocytes)

    • Methodology: Use cell type-specific markers (MAP2, GFAP, O4) in immunocytochemistry to distinguish effects on different neural populations.

  • Developmental Timing:

    • FGF9's effects vary with developmental stage

    • Methodology: Create precise developmental timelines for FGF9 addition in neural differentiation protocols with specific markers for each stage.

  • Receptor Expression Profiling:

    • Different neural cell types express varying levels of FGFR1-3

    • Methodology: Quantify receptor expression using qRT-PCR or flow cytometry before FGF9 treatment.

  • Neuroprotection Assays:

    • For Parkinson's, Huntington's, or Alzheimer's disease models

    • Methodology: Pre-treat cultures with FGF9 before adding neurotoxic compounds (MPP+, amyloid-β, glutamate) and measure:

      • Cell viability (MTT, LDH release)

      • Apoptotic markers (cleaved caspase-3, TUNEL)

      • Oxidative stress (ROS production, GSH levels)

  • Dose-Response Considerations:

    • Optimal concentrations for neuroprotection vs. differentiation may differ

    • Methodology: Perform comprehensive dose-response studies (1-200 ng/mL) with multiple readouts.

  • Compound Experimental Models:

    Model SystemApplicationsKey Readouts
    Primary neuron culturesDirect effects on neuronsNeurite outgrowth, synaptogenesis
    Neural organoids3D developmental effectsCytoarchitecture, layer formation
    Ex vivo brain slicesCircuit-level effectsElectrophysiology, connectivity
    In vivo modelsBehavioral outcomesCognitive/motor testing, histology
  • Co-factor Considerations:

    • FGF9 may require heparan sulfate proteoglycans as co-factors

    • Methodology: Include heparin (1-5 μg/mL) in experimental systems to optimize FGF9 activity.

Integrating these methodological considerations will enhance the rigor and reproducibility of neuronal FGF9 research while enabling more translational applications in neurodegenerative disease models.

What are the critical parameters to consider when using FGF9 in conjunction with other growth factors in complex developmental models?

When incorporating FGF9 into complex developmental models alongside other growth factors, several critical parameters must be considered to achieve optimal outcomes:

  • Signaling Pathway Cross-talk:

    • FGF9 activates ERK, JNK, STAT, and PI3K pathways, which may interact with pathways activated by other factors

    • Methodology: Use specific pathway inhibitors (U0126 for MEK/ERK, SP600125 for JNK) to parse out individual contributions of each growth factor.

  • Temporal Sequencing:

    • The order of growth factor exposure can dramatically affect cellular responses

    • Methodology: Design factorial experiments testing different sequences and durations of growth factor treatments (e.g., FGF9 before, after, or simultaneously with other factors).

  • Concentration Ratios:

    • The relative concentrations of multiple growth factors are often more important than absolute concentrations

    • Methodology: Create response surface methodologies testing different concentration ratios rather than single-factor dose-response curves.

  • Extracellular Matrix Context:

    • ECM components can modulate FGF9 binding to receptors and co-receptors

    • Methodology: Test FGF9 activity on cells cultured on different ECM substrates (laminin, fibronectin, Matrigel).

  • Common Growth Factor Combinations with FGF9:

    Developmental ContextGrowth Factor CombinationConcentration RangesNotes
    Kidney organoidsFGF9 + BMP7 + GDNFFGF9: 50-200 ng/mL; BMP7: 10-50 ng/mL; GDNF: 50-100 ng/mLSequential addition often more effective than simultaneous
    Neural developmentFGF9 + SHH + NogginFGF9: 10-50 ng/mL; SHH: 200-500 ng/mL; Noggin: 100-250 ng/mLPosition-dependent patterning requires gradient formation
    Testicular developmentFGF9 + WNT4 inhibitionFGF9: 50-100 ng/mL; WNT inhibitor: context-dependentAntagonistic relationship critical for sex determination
  • Readout Selection:

    • Different combinations produce unique cellular responses requiring specific assays

    • Methodology: Include both short-term (signaling) and long-term (differentiation, morphogenesis) readouts.

  • Supporting Technologies:

    • Microfluidic systems for precise spatial control

    • Methodology: Use gradient generators to create defined concentration gradients of multiple factors simultaneously.

  • Statistical Design Considerations:

    • Multi-factor experiments require appropriate statistical approaches

    • Methodology: Employ factorial design and multivariate analysis rather than simple t-tests or ANOVA.

By systematically addressing these parameters, researchers can more effectively develop physiologically relevant models that recapitulate complex developmental processes involving FGF9 signaling.

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.