Mouse FGF1 exhibits pleiotropic effects across physiological systems:
Diabetes Remission: A single intracerebroventricular (i.c.v.) injection of FGF1 induces sustained hypoglycemia in diabetic rodents (db/db, ob/ob mice) by activating arcuate nucleus (ARH) proopiomelanocortin (POMC) neurons and dorsal vagal complex (DVC) pathways .
Insulin Sensitization: FGF1 promotes adipose tissue remodeling and enhances insulin sensitivity, mimicking thiazolidinedione effects .
Cardiomyocyte Regeneration: FGF1 stimulates cardiomyocyte proliferation via FGFR1/Fn14 and PKC signaling, improving cardiac function post-myocardial infarction .
Lineage Tracing: The F1A-CreER T2 transgenic mouse model enables tracing of Fgf1A-expressing cardiomyocytes, highlighting its cardioprotective role .
Neuronal Activation: FGF1 increases c-Fos expression in the area postrema and nucleus tractus solitarius (NTS), modulating energy homeostasis .
Angiogenesis and Wound Healing: Compensates for FGF2 in double-knockout mice, supporting vascularization and tissue repair .
Cell Proliferation Assays: ED50 values for 3T3 fibroblast proliferation range from 0.1–1 ng/ml in heparin-containing media .
In Vivo Studies: Used to investigate diabetes remission, cardiac regeneration, and neuroprotection .
Knockout Mice: Fgf1⁻/⁻ mice show no overt phenotype under normal conditions but exhibit impaired wound healing and hematopoiesis when combined with Fgf2⁻/⁻ .
Transgenic Lines: F1A-CreER T2 mice enable tamoxifen-inducible tracing of Fgf1A-expressing cells, validated by LacZ/RFP co-localization with cardiac troponin T .
Antibody Specificity: Sheep anti-mouse FGF1 antibodies show ~50% cross-reactivity with human FGF1 and <1% with other mouse FGFs .
Species Conservation: High sequence conservation allows translational studies in metabolic and cardiovascular research .
Central Nervous System: FGF1 activates POMC neurons via FGFR1, independent of leptin signaling, to reduce hyperglycemia .
Cardiac Repair: Fgf1A promoter-driven CreER T2 models reveal cardiomyocyte-specific regeneration pathways .
Redundant Roles: FGF1 and FGF2 exhibit partial functional overlap, but FGF1 alone is insufficient to compensate for FGF2 in hematopoiesis .
Acidic fibroblast growth factor (FGF-1) belongs to the FGF family, known for their roles in cell growth, survival, and various biological processes. FGFs contribute to embryonic development, tissue repair, and angiogenesis (blood vessel formation). FGF-1 specifically influences endothelial cell activity, crucial for blood vessel formation. It acts as a mitogen, promoting cell division in various cell types derived from mesoderm and neuroectoderm, highlighting its role in organ development. Multiple forms of FGF-1 exist due to alternative splicing, leading to variations in its protein structure. These growth factors stimulate cell growth in various tissues and are potent mitogens for different cell types in laboratory settings. The distribution and concentration of these growth factors vary across tissues.
Recombinant Mouse Fibroblast Growth Factor-acidic (FGF-1), expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 141 amino acids. This protein has a molecular weight of 15.9 kDa.
FGF-acidic is purified using specialized chromatographic techniques.
Lyophilized from a 0.2 µm filtered concentrated solution in phosphate-buffered saline (PBS) at pH 7.5.
Purity is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.
The ED50, determined by the dose-dependent proliferation of mouse BALB/c 3T3 cells, is less than 0.2 ng/ml. This corresponds to a Specific Activity of 5 x 106 IU/mg.
MFNLPLGNYK KPKLLYCSNG GHFLRILPDG TVDGTRDRSD QHIQLQLSAE SAGEVYIKGT ETGQYLAMDT EGLLYGSQTP NEECLFLERL EENHYNTYTS KKHAEKNWFV GLKKNGSCKR GPRTHYGQKA ILFLPLPVSS D.
FGF1, also known as acidic FGF or HBGF-1, is a 17 kDa non-glycosylated member of the FGF family that regulates numerous biological and physiological processes in mice. It functions as a signaling molecule involved in cell proliferation, differentiation, and tissue repair. In mouse models, FGF1 has been extensively studied for its roles in cardiac function, adipose tissue homeostasis, and metabolic regulation. The protein is expressed in multiple tissues with particularly notable expression in the heart and kidneys . FGF1 induces proliferation in mouse fibroblast cell lines such as NR6R-3T3 in a dose-dependent manner, which can be neutralized using specific antibodies, providing a useful bioassay for functional studies .
In transgenic mouse models, FGF1 has demonstrated significant capacity to stimulate cardiomyocyte cell cycle reentry through the FGFR1/Fn14 pathway and regulate cardiomyocyte differentiation via the FGF1/FGFR/PKC signaling axis. Furthermore, it shows therapeutic potential in cardiac injury models by inducing cardiomyocyte regeneration and improving function after myocardial infarction .
In mice, the Fgf1 gene comprises at least three tissue-specific promoters that generate three alternatively spliced transcripts: Fgf1A, Fgf1B, and Fgf1G. These variants differ in their 5' untranslated regions but encode the same protein, as they all splice to the first protein-coding exon . The transcript variants show distinct tissue-specific expression patterns:
Fgf1A: Predominantly expressed in the heart and kidney under normal conditions; also expressed in adipose tissue in obesity models
Fgf1B: Shows different tissue distribution compared to Fgf1A
Fgf1G: Expressed in specific tissues distinct from Fgf1A and Fgf1B
The mouse Fgf1A transcript includes a 5' UTR of approximately 127.7 kb, followed by exon 1A, which is critical for tissue-specific expression . This complex gene structure allows for precise regulation of FGF1 expression in different physiological contexts.
Several complementary methods can be employed to detect and quantify FGF1 in mouse tissues:
Western Blot Analysis: Using specific antibodies such as Sheep Anti-Mouse FGF acidic/FGF1 Antigen Affinity-purified Polyclonal Antibody. Western blot can detect FGF1 in tissue lysates (e.g., heart and kidney tissues) as a specific band of approximately 18 kDa under reducing conditions .
Quantitative PCR (qPCR): For measuring Fgf1 transcript levels in different tissues. This approach can distinguish between different Fgf1 transcript variants using specific primers .
Immunohistochemistry/Immunofluorescence: For in situ detection of FGF1 protein in tissue sections, often combined with markers for specific cell types (e.g., cardiac troponin T for cardiomyocytes) .
Reporter Systems: Transgenic mice expressing reporters (LacZ, RFP) under the control of Fgf1 promoters enable visualization of Fgf1 expression patterns .
Cell Proliferation Assays: Functional assays using NR6R-3T3 mouse fibroblasts can measure FGF1 bioactivity. The neutralization dose (ND50) is typically 0.1-0.6 μg/mL in the presence of 1 ng/mL recombinant mouse FGF1 .
For optimal stability and activity of mouse FGF1 reagents, the following storage conditions are recommended:
Long-term storage: Use a manual defrost freezer at -20 to -70 °C for up to 12 months from date of receipt for unopened reagents
After reconstitution under sterile conditions:
It is crucial to avoid repeated freeze-thaw cycles as these can significantly reduce protein activity. Aliquoting reconstituted protein into single-use volumes is recommended for research requiring multiple experiments over time .
The F1A-CreERT2 transgenic mouse line provides a powerful tool for time-dependent and lineage tracing of Fgf1A-expressing cells in vivo. This model was generated using a BAC clone containing the Fgf1A 5'UTR (127.7 kb) and exon 1A sequence, with an RFP-IRES-CreERT2-polyA cassette inserted into exon 1A through homologous recombination .
Experimental methodology for lineage tracing:
Cross F1A-CreERT2 mice with reporter strains: The F1A-CreERT2 mice should be crossed with reporter mice such as ROSA26 LacZ reporter mice to generate F1 mice carrying both transgenes .
Tamoxifen administration: Inject tamoxifen to activate the CreERT2 recombinase at specific time points. This induces recombination specifically in cells where the Fgf1A promoter is active at the time of injection .
Analysis of reporter expression:
Co-localization studies: Combine reporter detection with immunostaining for cell-type specific markers (e.g., cardiac troponin T for cardiomyocytes) to identify the specific cell populations expressing Fgf1A .
The F1A-CreERT2 mouse model has revealed that in adult mice, Fgf1A promoter activity is predominantly found in cardiomyocytes, with reporter signals detected in various heart regions including right atrium (RA), right ventricle (RV), atrioventricular node (AV), interventricular septum (IVS), left atrium (LA), and left ventricle (LV) .
FGF1 plays significant roles in cardiomyocyte regeneration and cardiac repair through several mechanisms. Research has demonstrated that FGF1 can stimulate neonatal cardiomyocyte cell cycle reentry through the FGFR1/Fn14 pathway, regulate cardiomyocyte differentiation from embryonic stem cells via the FGF1/FGFR/PKC signaling axis, and induce cardiomyocyte regeneration after myocardial infarction .
Experimental models for studying FGF1 in cardiac regeneration:
In vitro models:
In vivo models:
Combination approaches:
The F1A-CreERT2 mouse line provides a valuable tool for these studies by allowing researchers to track the fate of Fgf1-expressing cells during cardiac development, injury, and regeneration processes .
FGF1 plays crucial roles in metabolic regulation, particularly in glucose homeostasis and adipose tissue function. In mouse models of diabetes and obesity, several key findings have emerged:
Adipose tissue homeostasis: FGF1 regulates adipose tissue homeostasis, with Fgf1 knockout mice showing an aggressive diabetic phenotype when fed a high-fat diet, indicating its importance in metabolic adaptation .
Glucose regulation: Subcutaneous injection of FGF1 protein significantly improves insulin sensitivity and reduces serum glucose levels in multiple mouse models of diabetes:
Rapid and sustained effects: Significant reduction in serum glucose levels can be achieved within 24 hours of FGF1 administration and can last more than 48 hours. Importantly, even at higher doses, FGF1 does not induce hypoglycemia in diabetic mice, suggesting a regulated mechanism of action .
β-cell independent effects: FGF1 can reduce serum glucose levels even in streptozotocin-induced diabetic mice, which have lost β-cells, indicating mechanisms beyond insulin secretion .
Anti-inflammatory action: Long-term FGF1 administration ameliorates systemic inflammation in ob/ob mice, functioning through the FGFR1 pathway and acting similar to thiazolidinediones as an insulin sensitizer .
Experimental approaches to study FGF1 in metabolism:
High-fat diet feeding with FGF1 administration
Glucose tolerance tests and insulin tolerance tests
Analysis of inflammatory markers
Adipose tissue-specific gene expression analysis
Comparison of wild-type and Fgf1 knockout mice under metabolic challenge
Creating and properly characterizing FGF1 transgenic mouse models requires careful attention to several technical considerations:
Promoter selection and design:
Recombination system design:
Founder selection and validation:
Multiple founder lines should be generated and compared
The F1A-CreERT2 model identified three potential founders, but only one (#13A) showed sufficient expression levels for functional studies
Quantitative PCR for transgene expression in target tissues is essential for selecting appropriate founders
Reporter system integration:
Validation of expression patterns:
Functional validation:
Cross with reporter strains (e.g., ROSA26) to confirm recombinase activity
Test induction protocols (e.g., tamoxifen dosing) for optimal temporal control
Verify phenotype absence in unmodified conditions to ensure the model doesn't have inherent defects
Quantitative assessment of FGF1 activity in mouse models can be approached through several complementary methods:
Cell proliferation assays:
The NR6R-3T3 mouse fibroblast cell line shows dose-dependent proliferation in response to FGF1
Proliferation can be quantified and used as a bioassay for FGF1 activity
The response can be neutralized by anti-FGF1 antibodies, with a typical neutralization dose (ND50) of 0.1-0.6 μg/mL in the presence of 1 ng/mL recombinant mouse FGF1
Western blot quantification:
Transcriptional analysis:
qPCR for Fgf1 mRNA variants and FGF1-responsive genes
RNA-seq for comprehensive transcriptional profiling
Analysis should distinguish between different Fgf1 transcript variants (1A, 1B, 1G)
In vivo functional assays:
For cardiac studies: echocardiography, pressure-volume measurements
For metabolic studies: glucose tolerance tests, insulin sensitivity tests
For regeneration studies: BrdU or EdU incorporation to measure proliferation
Reporter systems:
ELISA and immunoassays:
For measuring circulating FGF1 levels in serum or plasma
For quantifying FGF1 in tissue homogenates or culture supernatants
These methods can be combined to provide comprehensive assessment of FGF1 activity at molecular, cellular, and physiological levels in experimental mouse models.
For effective temporal control of Cre-mediated recombination in F1A-CreERT2 mice, proper tamoxifen administration is crucial. While the provided search results don't specify exact protocols for this particular mouse line, general considerations based on standard practices for CreERT2 systems include:
Delivery methods:
Intraperitoneal (IP) injection: Most common method for adult mice
Oral gavage: Alternative to IP injections
Tamoxifen-containing food: For more gradual, chronic induction
Dosage considerations:
For adult mice, 1-2 mg tamoxifen per 25g body weight is typically used
Multiple injections (3-5 consecutive days) are often needed for optimal recombination
Dose finding studies should be performed to determine optimal conditions for each specific CreERT2 line
Timing for lineage tracing:
Allow 24-72 hours after the last tamoxifen dose before analyzing acute effects
For long-term lineage tracing, reporter expression becomes more robust 5-7 days after induction
Preparation of tamoxifen:
Dissolve in corn oil or sunflower oil at concentrations of 10-20 mg/mL
For complete dissolution, sonication and heating (37°C) may be required
Prepare fresh solutions or store at -20°C protected from light for limited periods
Controls:
Oil-only injections in F1A-CreERT2 x reporter mice
Tamoxifen injections in reporter-only mice
These controls are essential to rule out leaky Cre activity or tamoxifen-related effects
In the F1A-CreERT2 mice, tamoxifen administration resulted in CreERT2-mediated recombination specifically in cardiomyocytes, with no recombination observed in other cell types, confirming the cardiomyocyte-specific activation of the Fgf1A promoter .
Proper tissue preparation is critical for reliable detection of FGF1 protein and reporter signals in transgenic mouse models. Based on the provided research, the following protocols have proven effective:
For direct fluorescence detection (e.g., RFP):
Anesthetize mice with isoflurane
Perform intracardiac perfusion with ice-cold PBS without calcium and magnesium
Fix harvested tissues in pre-chilled 95% ethanol for 20-24h at 4°C
Dehydrate in 4 changes of pre-chilled 100% ethanol for 1h at 4°C
Embed in optimal cutting temperature (OCT) compound
Store at -70°C
Cryosection to 6-μm thickness
Rinse with water and observe directly under a fluorescent microscope without additional staining
For LacZ staining:
For immunohistochemical detection:
Use appropriate fixation based on antibody requirements (typically 4% paraformaldehyde)
For co-localization studies with cell type markers (e.g., cardiac troponin T), optimize fixation to preserve epitopes for all antibodies
When detecting both RFP signals and immunofluorescence staining, careful optimization of fixation is crucial to maintain RFP fluorescence while allowing antibody binding
For Western blot analysis:
These protocols can be adapted based on specific experimental requirements and should be optimized for each application and tissue type.
When facing challenges with weak or variable FGF1 expression in mouse models, researchers should consider several troubleshooting approaches:
Founder line selection:
As demonstrated with the F1A-CreERT2 mice, significant variation can exist between founder lines
In the reported study, strain #13A showed the highest gene expression levels, with Ct value differences of 8.5-9.3 compared to other strains
Always generate and screen multiple founder lines when creating new transgenic models
Tissue collection and processing optimization:
Use rapid extraction and appropriate preservation methods
For RNA analysis, use RNase inhibitors and quick freezing
For protein analysis, include protease inhibitors in all buffers
Age and sex considerations:
FGF1 expression may vary with age and sex
Standardize experimental groups by age and include both sexes or justify single-sex studies
Consider developmental timing for embryonic or neonatal studies
Environmental and physiological factors:
Feeding status, time of day, stress levels, and housing conditions can affect gene expression
Standardize conditions and collection times
Record any potential confounding variables
Detection method sensitivity:
For low expression, consider more sensitive methods (e.g., droplet digital PCR instead of qPCR)
Use amplification steps in immunohistochemistry protocols
Consider concentration steps for protein detection
Genetic background effects:
Backcross to a consistent genetic background for more reliable results
Be aware that modifier genes can influence FGF1 expression and function
Induction protocol optimization:
For inducible systems like F1A-CreERT2, optimize tamoxifen dosing and timing
Consider dose-response studies to determine optimal conditions
Proper experimental controls are critical for reliable interpretation of results when studying FGF1 function. Essential controls include:
Genetic controls:
Wild-type littermates: Essential baseline control
Heterozygous vs. homozygous comparisons: To assess gene dosage effects
Cre-only and floxed-only controls: For conditional systems to rule out effects of Cre expression or loxP sites
Induction controls for CreERT2 systems:
Tissue specificity controls:
Functional assay controls:
Technical controls:
For Western blots: Loading controls, negative control tissues, recombinant protein standards
For PCR: No-template controls, reverse transcriptase negative controls
For immunostaining: Primary antibody omission, isotype controls, blocking peptide controls
Physiological controls:
Age-matched and sex-matched animals
Controlled environmental conditions (diet, housing, handling)
Consistent time of day for measurements of physiologically variable parameters
These controls help distinguish specific FGF1-related effects from background variation, technical artifacts, and non-specific responses to experimental manipulations.
Based on findings from mouse models, FGF1 shows therapeutic potential in several disease areas that warrant further investigation:
Cardiac regeneration and repair:
FGF1 induces cardiomyocyte regeneration and improves cardiac function after myocardial infarction
The combination of FGF1 with neurogenin1 stimulates cardiomyocyte proliferation and facilitates cardiac remodeling
Future research could optimize delivery methods, timing, and combination therapies for maximal regenerative effect
Diabetes and metabolic disorders:
FGF1 significantly improves insulin sensitivity and reduces serum glucose levels in multiple mouse models of diabetes
Unlike insulin, higher doses of FGF1 do not cause hypoglycemia, suggesting a regulated mechanism
FGF1 administration ameliorates systemic inflammation in obese mice
Future studies could focus on developing FGF1 variants with enhanced stability or receptor specificity for metabolic applications
Diabetic cardiomyopathy:
Adipose tissue homeostasis:
Developmental biology applications:
The F1A-CreERT2 mouse model enables precise temporal control for developmental studies
Future research could use this system to dissect the role of FGF1A-expressing cells during critical developmental windows
This could inform regenerative medicine approaches that recapitulate developmental processes
Single-cell technologies offer powerful new approaches to understand FGF1 biology at unprecedented resolution:
Single-cell RNA sequencing (scRNA-seq):
Can identify specific cell populations expressing Fgf1 and its receptors
Would reveal heterogeneity within seemingly uniform tissues
Could track transcriptional changes in FGF1-responsive cells during development, homeostasis, and disease
Would be particularly valuable for heart studies, where the F1A-CreERT2 model has shown cardiomyocyte-specific expression
Single-cell ATAC-seq:
Spatial transcriptomics:
CRISPR-based lineage tracing:
Single-cell proteomics and metabolomics:
These technologies could transform our understanding of FGF1 biology by revealing cell-type specific responses, identifying new therapeutic targets, and enabling precision approaches to modulating FGF1 signaling in specific contexts.
FGF-acidic is a single-chain protein with a molecular weight of approximately 15.8 kDa . It binds to and activates high-affinity FGF receptors (FGFR1-4) on target cells, initiating a signal transduction cascade involving kinases and transcription factors . This activation leads to several cellular effects, such as:
FGF-acidic is highly regulated and acts as a direct angiogenesis factor . It is known for its mitogenic activity, particularly targeting endothelial cells . However, unlike some other FGFs, FGF-acidic lacks a signal peptide segment, meaning it is not secreted via the endoplasmic reticulum (ER) and Golgi bodies .
Due to its role in angiogenesis, FGF-acidic is considered a potential oncogene. Its overexpression is closely associated with several cancers, including those affecting the eyes, joints, and pancreas . Understanding the regulation and function of FGF-acidic can provide insights into therapeutic targets for these diseases.