Fibroblast growth factor 9 (FGF9) is a recombinant protein derived from rats, produced in E. coli and widely used in research to study its roles in cellular development, tissue repair, and disease mechanisms. As a member of the FGF family, FGF9 Rat exhibits mitogenic and survival-promoting activities, with implications in neurobiology, reproductive development, and neurodegenerative disorders .
Formulation: Lyophilized from a solution containing 10 mM sodium phosphate (pH 7.5) and 75 mM ammonium sulfate .
Solubility: Reconstituted at ≥100 µg/mL in sterile water, with further dilution into aqueous solutions .
Storage: Stable at −18°C; repeated freeze-thaw cycles should be avoided. Long-term storage requires a carrier protein (e.g., 0.1% BSA) .
Biological Activity: ED₅₀ <0.5 ng/mL in BAF3 cell proliferation assays, corresponding to 2,000,000 Units/mg .
Neurological Development: Expressed in neurons, astrocytes, and oligodendrocytes, FGF9 supports glial cell proliferation and neuronal survival .
Reproductive Development: Essential for male gonadal differentiation and survival of primordial germ cells (PGCs), regulating Dnmt3L and Nanos2 expression in a dose-dependent manner .
Steroidogenesis: Activates Akt and ERK1/2 pathways in Leydig cells to stimulate testosterone production .
Neurodegeneration: Overexpression in the rat cortex induces grey matter lesions, axonal loss, and demyelination, mimicking progressive multiple sclerosis (MS) .
Affective Disorders: Elevated in major depressive disorder (MDD), FGF9 inversely correlates with FGF2 expression and promotes anxiety/depression-like behaviors in rodent models .
Low FGF9 concentrations (0.2 ng/mL) induce male differentiation via p38 MAPK signaling, while high concentrations (25 ng/mL) stimulate proliferation through ERK1/2 activation .
FGF9 Concentration (ng/mL) | Gene Expression (Relative to Control) | Signaling Pathway |
---|---|---|
0.2 | ↑ Dnmt3L (2.5×), ↓ Stra8 (0.5×) | p38 MAPK |
25 | ↓ Dnmt3L (0.5×), ↓ Stra8 (0.5×) | ERK1/2 |
Multiple Sclerosis: AAV-mediated overexpression in the rat motor cortex causes chronic grey matter lesions with axonal injury and microglial activation .
Poststroke Depression (PSD): FGF9 mRNA/protein levels in the hippocampus are elevated in PSD models, correlating with reduced FGFR1/FGFR3 expression .
MDD: Human postmortem studies show FGF9 upregulation in the hippocampus, with chronic social defeat stress inducing similar patterns in rodents .
FGF9/FGFR Pathway: Inhibition of FGF9 reduces anxiety-like behavior in rats, suggesting potential for treating affective disorders .
Remyelination Inhibition: FGF9 suppresses remyelination in MS models, highlighting its role in disease progression .
Used to model neurodegeneration, FGF9 Rat is injected into the rat cortex to study axonal loss and demyelination .
Investigates FGF9’s role in sex determination and germ cell survival, particularly in PGC cultures .
Employed in MDD and anxiety models to elucidate FGF9’s inverse relationship with FGF2 and its effects on synaptic function .
FGF-9 immunoreactivity is prominently expressed in motor neurons of the spinal cord, Purkinje cells, and neurons in the hippocampus and cerebral cortex. Beyond neuronal expression, FGF-9 is also present in glial fibrillary acidic protein (GFAP)-positive astrocytes, particularly in the white matter of spinal cord and brainstem of adult rats. Additionally, FGF-9 immunoreactivity is found in CNPase-positive oligodendrocytes arranged between fasciculi of nerve fibers in cerebellar white matter and corpus callosum of both adult and young rats .
The expression pattern shows some age-related variations, with oligodendrocyte FGF-9 immunoreactivity appearing more pronounced in young rats compared to adult rats. The variation in oligodendrocyte FGF-9 immunoreactivity is also more notable in adult rats than in young rats .
During normal anorectal development in rat embryos, FGF-9 expression shows spatiotemporal changes between embryonic day 13 (E13) and E16. FGF-9-positive cells are predominantly found in the mesenchyme of the cloaca on E13 and E14. By E15, these cells are concentrated at the fusion site of the urorectal septum and cloacal membrane, rectal epithelium, and anal membrane. After the anal membrane ruptures on E16, FGF-9-positive cells significantly decrease .
This pattern suggests that anorectal embryogenesis may depend on the induction of FGF-9 signaling, with expression levels rising during critical developmental windows and declining after key developmental events are completed .
Based on available research, FGF-9 appears to have several important functions in rat physiology:
Neural development and maintenance: FGF-9 is expressed in various neuronal populations and may contribute to their development and maintenance .
Glial cell function: Expression in astrocytes and oligodendrocytes suggests roles in glial cell biology and potentially in myelination processes .
Steroidogenesis regulation: FGF-9 affects steroid production in granulosa cells by influencing key steroidogenic enzymes .
Embryonic development: FGF-9 plays critical roles in anorectal development, with altered expression associated with anorectal malformations .
Neural tissue survival: In in vitro culture models, FGF-9 has been shown to stimulate the survival of neural tissue cells .
FGF-9 exhibits inhibitory effects on steroidogenesis in rat granulosa cells. Experimental evidence shows that FGF-9 suppresses steroid production in both small-to-medium sized granulosa cells (SMGC) and large granulosa cells (LGGC). This inhibition operates through several mechanisms:
Downregulation of steroidogenic enzyme expression: FGF-9 (10 ng/ml) significantly decreases CYP11A1 mRNA abundance by 68% in SMGC and 45% in LGGC, directly affecting the conversion of cholesterol to pregnenolone, a rate-limiting step in steroidogenesis .
Suppression of FSHR expression: FGF-9 decreases FSHR mRNA abundance by 24% in SMGC and 32% in LGGC, potentially reducing the cells' responsiveness to FSH stimulation .
Interference with cAMP-mediated signaling: FGF-9 appears to inhibit steroidogenesis stimulated by FSH and IGF-I, potentially by interfering with the cAMP/PKA signaling pathway that typically upregulates steroidogenic enzyme expression .
These findings suggest that FGF-9 may serve as a physiological regulator of ovarian steroidogenesis, potentially important for follicular development and ovulation in rats.
In rat models of pathological conditions, FGF-9 shows significant expression changes that may reflect underlying disease mechanisms. For example, in middle cerebral artery occlusion (MCAO) rats, both protein and mRNA expression of FGF-9 are significantly upregulated in the dentate gyrus (DG) of the hippocampus compared to control rats .
In contrast, in rat embryos with anorectal malformations (ARM) induced by ethylenethiourea (ETU), FGF-9 expression is significantly decreased compared to normal embryos from E13 to E15. This downregulation may be related to the development of anorectal malformations, suggesting that FGF-9 plays a crucial role in normal anorectal development .
These contrasting patterns—upregulation in stroke/depression and downregulation in developmental abnormalities—highlight the context-specific roles of FGF-9 in different pathological conditions.
While the search results don't provide direct information on cross-talk between FGF-9 and other signaling pathways in rat neural development, we can infer potential interactions based on the available data.
FGF-9 receptors (FGFR1 and FGFR3) show unique expression patterns in rat hippocampus, with different responses to pathological conditions. For instance, in post-stroke depression rats, FGFR1 mRNA decreased by 31% while FGFR3 mRNA decreased by 65%, suggesting differential regulation of these receptors .
This differential regulation may indicate cross-talk with other signaling pathways that selectively modulate specific FGF receptors. Additionally, the fact that fluoxetine treatment affects FGF-9 expression suggests potential interactions with serotonergic signaling pathways .
In developmental contexts, the temporal and spatial expression patterns of FGF-9 during anorectal development suggest coordination with other developmental signaling pathways. The localization of FGF-9-positive cells at the fusion site of the urorectal septum and cloacal membrane implies potential interactions with morphogenic signaling pathways involved in tissue fusion and epithelial-mesenchymal interactions .
Based on the research studies examined, several complementary techniques have proven effective for detecting FGF-9 expression in rat tissues:
Immunohistochemistry: This technique has been successfully used to localize FGF-9 protein in various cell types, including neurons, astrocytes, and oligodendrocytes. Double-label immunohistochemistry with cell-specific markers (e.g., GFAP for astrocytes, CNPase for oligodendrocytes) is particularly valuable for identifying cell types expressing FGF-9 .
In situ hybridization: This method effectively detects FGF-9 mRNA in tissue sections, allowing for the visualization of cells actively transcribing the FGF-9 gene. This technique has revealed that FGF-9 mRNA expression in glial cells is lower than in neurons, and not all glial cells express FGF-9 .
Western blotting: Western blot analysis using specific antibodies can identify FGF-9 protein as an immunopositive band with a molecular weight around 21 kDa. This technique is useful for quantitative analysis of protein expression levels across different experimental conditions .
Quantitative real-time PCR (qRT-PCR): This method provides quantitative measurements of FGF-9 mRNA expression and has been used to show that changes in FGF-9 mRNA levels often correspond with protein expression changes .
For optimal results, researchers should consider combining multiple detection methods to provide complementary information about both mRNA and protein expression patterns.
Several experimental models have proven valuable for studying different aspects of FGF-9 function in rats:
In vitro cell culture models: Isolated rat granulosa cells (both SMGC and LGGC) have been effectively used to study FGF-9's effects on steroidogenesis. These models allow for controlled experimental conditions and facilitate the investigation of dose-dependent effects and interactions with other factors .
In vitro embryo culture: This model has been used to study FGF-9's influence on rat embryonic development. Researchers have found that FGF-9 can affect the growth of rat fetuses cultivated in vitro, with effects depending on the timing of application and concentration .
Middle cerebral artery occlusion (MCAO) model: This stroke model in rats has revealed important insights about FGF-9 expression changes in response to cerebral ischemia and its potential role in post-stroke depression .
Ethylenethiourea (ETU)-induced anorectal malformation model: Administration of ETU to pregnant rats on embryonic day 10 (E10) induces anorectal malformations in the offspring, providing a valuable model for studying FGF-9's role in normal and abnormal anorectal development .
The choice of model should be guided by the specific research question being addressed, with consideration of the particular aspect of FGF-9 biology under investigation.
While the search results don't provide comprehensive information on all methods for manipulating FGF-9 signaling, several approaches can be inferred:
Exogenous FGF-9 administration: Direct application of recombinant FGF-9 protein at various concentrations (3-30 ng/ml) has been used to study its effects on steroidogenesis in granulosa cells and on embryonic development in culture models . This approach allows for dose-response studies and investigation of timing-dependent effects.
Pharmacological modulation: Fluoxetine treatment has been shown to partially reverse FGF-9 upregulation in post-stroke depression rats, suggesting that certain pharmacological agents can indirectly modulate FGF-9 expression .
In vitro models with controlled conditions: Researchers have used in vitro culture systems where they can manipulate the timing, duration, and concentration of FGF-9 exposure, as well as the presence of other factors (e.g., FSH, IGF-I) .
Developmental timing studies: By studying FGF-9 expression at different embryonic stages and in models of developmental abnormalities, researchers can gain insights into the temporal requirements for FGF-9 signaling .
For more targeted manipulations, researchers might consider additional approaches not explicitly mentioned in the search results, such as:
Viral vector-mediated overexpression or knockdown of FGF-9
CRISPR-Cas9 gene editing to modify FGF-9 or its receptors
Conditional knockout models using Cre-loxP systems to achieve tissue-specific or temporally controlled manipulation of FGF-9 signaling
Research on FGF-9 in rat models has provided valuable insights into its potential roles in neurological disorders:
Post-stroke depression: The significant upregulation of FGF-9 in the hippocampus of rats with post-stroke depression, and the partial reversal of this upregulation by fluoxetine treatment, suggests that FGF-9 may be involved in the pathophysiology of this condition. This finding could potentially lead to new therapeutic approaches targeting FGF-9 signaling .
Neural tissue survival: FGF-9's ability to stimulate the survival of neural tissue cells in vitro suggests potential neuroprotective properties that could be relevant for neurodegenerative disorders .
Developmental neurobiology: The expression of FGF-9 in various neuronal populations and glial cells in the rat CNS indicates its importance in neural development and potentially in neurological disorders with developmental origins .
The rat model offers several advantages for studying FGF-9 in neurological contexts, including the ability to perform detailed behavioral assessments, accessibility of brain tissue for molecular and histological analyses, and the availability of established models for various neurological conditions.
FGF-9 research in rat models has revealed important roles in reproductive physiology, particularly in ovarian function:
Regulation of steroidogenesis: FGF-9 inhibits FSH and IGF-I-stimulated estradiol and progesterone production in rat granulosa cells, suggesting a role in modulating follicular development and potentially ovulation .
Molecular mechanisms: FGF-9 decreases expression of key steroidogenic enzymes (CYP11A1) and FSH receptor (FSHR), indicating specific molecular targets through which it regulates ovarian function .
Cell-type specific effects: FGF-9's effects on both small-to-medium sized granulosa cells (SMGC) and large granulosa cells (LGGC) suggest roles throughout follicular development .
These findings from rat models have implications for understanding:
Follicular development and ovulation in mammals
Ovarian disorders involving dysregulated steroidogenesis
Potential contraceptive approaches targeting FGF-9 signaling
Fertility treatments that might need to account for FGF-9's inhibitory effects on steroidogenesis
Research on FGF-9 in rat developmental models has provided significant insights into its role in congenital malformations, particularly anorectal malformations (ARM):
Temporal expression pattern: FGF-9 shows a specific spatiotemporal expression pattern during normal anorectal development in rat embryos, with expression in the mesenchyme of the cloaca on E13-E14 and at the fusion site of the urorectal septum and cloacal membrane, rectal epithelium, and anal membrane on E15 .
Downregulation in ARM: FGF-9 expression is significantly decreased in ethylenethiourea (ETU)-induced ARM embryos compared to normal embryos from E13 to E15, suggesting that reduced FGF-9 signaling may contribute to the development of these malformations .
Mechanistic insights: The finding that FGF-9 expression is downregulated in ARM embryos suggests that anorectal embryogenesis might depend on the induction of FGF-9 signaling, providing a potential mechanistic link between FGF-9 and normal anorectal development .
These findings contribute to our understanding of the molecular basis of congenital malformations and could potentially lead to improved diagnostic approaches or therapeutic interventions for these conditions. Additionally, this research highlights the importance of proper growth factor signaling during embryonic development and the consequences of disruptions to these signaling pathways.
Several emerging techniques could significantly advance FGF-9 research in rat models:
Single-cell RNA sequencing: This technique could provide unprecedented insights into cell-specific FGF-9 expression patterns and identify novel cell populations responsive to FGF-9 signaling. Given the heterogeneous expression of FGF-9 observed in glial cells , single-cell analyses could reveal important subpopulation-specific effects.
Spatial transcriptomics: This approach could map FGF-9 and FGF receptor expression with spatial resolution, providing a more comprehensive understanding of signaling interactions within tissues without losing contextual information.
Optogenetic and chemogenetic tools: These techniques could enable temporally precise manipulation of FGF-9 signaling in specific cell populations, allowing for more sophisticated functional studies.
Advanced in vivo imaging: Techniques for visualizing FGF-9 signaling in live animals could provide dynamic information about signaling events during development or in response to pathological conditions.
CRISPR-Cas9 gene editing: More sophisticated gene editing approaches could enable precise manipulation of FGF-9 or its receptors in specific cell types or at specific developmental stages.
These advanced techniques could help address remaining questions about FGF-9's roles in development, homeostasis, and disease in rat models.
Integrated multi-omics approaches could provide comprehensive insights into FGF-9 biology:
Combining transcriptomics, proteomics, and metabolomics: This approach could reveal how FGF-9 signaling affects not only gene expression but also protein levels and metabolic pathways, providing a more complete picture of its cellular effects.
Epigenomic analyses: Investigating how FGF-9 signaling affects chromatin structure and DNA modifications could reveal mechanisms of sustained or delayed effects on gene expression.
Interactome mapping: Comprehensive analysis of protein-protein interactions involving FGF-9 and its downstream effectors could identify novel signaling components and pathway interconnections.
Systems biology approaches: Computational integration of multi-omics data could help model the complex networks through which FGF-9 exerts its diverse biological effects.
These integrated approaches could help resolve apparent contradictions in FGF-9 research, such as its context-dependent effects on different cell types or in different pathological conditions.
FGF-9 binds to its receptor with the help of heparin, which induces receptor dimerization, subsequent transphosphorylation, and downstream activation of several signaling pathways, including Erk, Akt, and PLCγ . These pathways are essential for various cellular processes such as proliferation, differentiation, and survival.