Fibroblast Growth Factor-acidic Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 154 amino acids and having a molecular mass of 17.3kDa. The FGF1 is purified by proprietary chromatographic techniques.
HBGF-1, ECGF-beta, FIBP, FGFIBP, FIBP-1, ECGF, ECGFA, GLIO703, FGF1, FGF-a.
Escherichia Coli.
AEGEITTFTA LTEKFNLPPG NYKKPKLLYC SNGGHFLRIL PDGTVDGTRD RSDQHIQLQL SAESVGEVYI KSTETGQYLA MDTDGLLYGS QTPNEECLFL ERLEENHYNT YISKKHAEKN WFVGLKKNGS CKRGPRTHYG QKAILFLPLP VSSD.
The 154 amino acid form of FGF1 represents a long-form variant that comprises the core structured region plus an N-terminal extension . This differs from shorter variants that may lack this extension. Structurally, FGF1 adopts a β-trefoil core configuration, which is evident when examined through techniques such as fluorescence spectroscopy and circular dichroism . The intrinsic fluorescence spectra of FGF1 typically shows an emission maximum centered at 307 nm, indicating that the emission of the conserved tryptophan is quenched . This structural information is critical for understanding FGF1 function as the N-terminal extension can affect protein stability, receptor binding, and biological activity in experimental settings.
Human FGF1 demonstrates remarkable evolutionary conservation across mammalian species, highlighting its fundamental biological importance. According to comparative sequence analyses, human FGF1 shows very high sequence homology with: bovine (99%), porcine (99%), rat (97%), and mouse (94%) . This high degree of conservation suggests that FGF1's biological functions are essential and have been maintained throughout mammalian evolution. Researchers should consider this high homology when designing cross-species experiments or when using animal models to study FGF1 function, as results may translate well between these species due to the conserved protein structure and function.
For optimal research results, recombinant human FGF1 (154 a.a.) requires specific storage and handling protocols. The protein should be stored at -80°C for long-term stability, with aliquoting recommended to prevent freeze-thaw cycles that can compromise protein integrity. When preparing working solutions, researchers should use sterile, low-binding microcentrifuge tubes and perform dilutions in appropriate buffers containing 0.1-0.5% bovine serum albumin (BSA) as a carrier protein to prevent adsorption to plastic surfaces. The protein should be kept on ice during experimental setup and avoid repeated freeze-thaw cycles. Activity testing prior to critical experiments is advisable, as FGF1 activity can decrease over time even under optimal storage conditions. For experimental design, researchers should consider that FGF1 is known to interact with heparin or heparin sulfate, which can influence its biological activity in both in vitro and in vivo models .
When utilizing FGF1 (154 a.a.) for stem cell maintenance, several critical parameters must be optimized. Recombinant human FGF1 (154 a.a.) is particularly valuable for supporting human embryonic stem cell maintenance and proliferation of induced pluripotent and mesenchymal stem cells . For optimal results, supplement cell culture medium with 5-10 ng/mL of FGF1 in the presence of 1-5 μg/mL heparin, which stabilizes FGF1 and enhances its receptor binding. Medium should be refreshed every 24-48 hours as FGF1 has limited stability in culture conditions. For embryonic stem cells, combine FGF1 with other factors such as TGF-β pathway inhibitors for enhanced pluripotency maintenance. When transitioning from FGF2 to FGF1 protocols, gradual adaptation may be necessary as cellular responses can differ despite their similar functions. Monitor cultures for morphological changes, expression of pluripotency markers, and differentiation capacity to verify the efficacy of FGF1 supplementation. Researchers should note that while both FGF1 and FGF2 have been linked to regulating food intake centrally, only FGF1 displays significant glucose-lowering effects after peripheral injection .
To assess FGF1 activity in metabolic regulation experiments, researchers should implement a multi-parameter approach that captures both acute and chronic effects. For glucose homeostasis assessment, measure blood glucose levels, glucose tolerance (via GTT), and insulin sensitivity (via ITT) at multiple timepoints following FGF1 administration. It's important to note that peripheral and central administration of FGF1 produce different temporal effects—peripheral injection triggers acute glucose lowering within hours while intracerebroventricular (ICV) injection lowers glucose after about a week but sustains this effect beyond 16 weeks . When designing experiments, monitor feeding behavior as FGF1 can suppress food intake, though this effect is typically transitory compared to its persistent glucose-lowering impact . For molecular mechanism studies, analyze FGF receptor activation (particularly FGFR1) and downstream signaling pathways in target tissues. In adipose tissue specifically, assess glucose uptake, as this has been identified as a primary target of FGF1 action . For central administration studies, examine expression of early-response markers like c-Fos and heat shock protein β1 in glucose-sensing tanycytes and periventricular hypothalamic astrocytes, which appear to be primary cellular targets of FGF1 in the brain .
For comprehensive analysis of FGF1-receptor interactions across tissue contexts, researchers should employ complementary approaches that address both binding properties and functional outcomes. Begin with biochemical binding assays using surface plasmon resonance (SPR) or fluorescence anisotropy to determine binding affinities between purified FGF1 and FGFR variants. These methods can quantify how heparin/heparan sulfate modulates binding kinetics, which is crucial as FGF1 is a paracrine FGF that works locally through interaction with heparin or heparin sulfate . For cellular studies, implement receptor visualization techniques including fluorescently-labeled FGF1 combined with confocal microscopy to track receptor binding, internalization, and trafficking. Receptor activation can be assessed through phosphorylation-specific antibodies against FGFRs and downstream signaling components. Tissue-specific receptor distribution should be mapped using immunohistochemistry with FGFR-specific antibodies, contextualizing findings with FGF1 expression patterns. For functional analysis, compare FGF1 responses in cell lines with defined FGFR expression profiles, using CRISPR-Cas9 receptor knockout or knockdown approaches to confirm specificity. When studying metabolic effects, note that adipose tissue has been identified as the primary target site of 'endocrinized' recombinant FGF1, mediated by the FGF1-FGFR1 signaling cascade . For central nervous system studies, focus on ependymal cells lining the ventricular space, which constitute the main source of FGF1 production .
Central and peripheral administration of FGF1 produce distinct glycemic responses that differ in timing, duration, and underlying mechanisms. With peripheral injection, FGF1 triggers acute glucose lowering within hours, and multiple doses promote insulin sensitization after approximately 3 weeks of treatment . In contrast, intracerebroventricular (ICV) injection of FGF1 initiates glucose-lowering effects after about one week but remarkably sustains this effect beyond 16 weeks without evident insulin sensitization . Both routes of administration achieve glucose lowering without causing dangerous hypoglycemia, which is a significant advantage over many current diabetes therapies . The mechanistic differences appear substantial—peripheral FGF1 acts primarily through adipose tissue via the FGF1-FGFR1 signaling cascade, while central FGF1 may function through glucose-sensing neuronal circuits and hypothalamic cells . Researchers investigating FGF1 as a diabetes therapeutic should consider these route-dependent differences when designing preclinical studies. Additionally, while both FGF1 and FGF2 have been linked to central regulation of food intake, only FGF1 demonstrates significant glucose-lowering effects after peripheral injection, highlighting the specificity of FGF1's metabolic functions .
The remarkable sustained glucose-lowering effect of FGF1 in diabetic models involves complex central and peripheral mechanisms that researchers are still elucidating. In the central nervous system, FGF1 appears to influence glucose-sensing neuronal circuits, particularly in the hypothalamus . Upon glucose stimulation, ependymal cells lining the ventricular space secrete FGF1, which induces expression of early-response markers like c-Fos and heat shock protein β1 in glucose-sensing tanycytes lining the third ventricle and in periventricular hypothalamic astrocytes . Surprisingly, FGF1 does not induce changes in c-Fos expression in hypothalamic neurons directly, suggesting tanycytes and astrocytes are the primary cellular targets . In peripheral tissues, adipose tissue serves as the primary target of FGF1 through the FGF1-FGFR1 signaling pathway . This signaling enhances glucose uptake and improves insulin sensitivity without causing hypoglycemia . The sustained effect after a single ICV injection (beyond 16 weeks) suggests FGF1 may induce long-term reprogramming of glucose regulatory networks or persistent changes in hypothalamic neural circuitry . Researchers exploring these mechanisms should focus on both immediate signaling events and long-term transcriptional, epigenetic, or structural changes in glucose-regulating networks across central and peripheral tissues.
The development of FGF1-FGF21 chimeric proteins represents an innovative approach to enhance stability and antidiabetic efficacy by combining FGF21's metabolic benefits with FGF1's thermal stability. Structure-based design principles should guide this optimization process, as exemplified by the successful strategy of replacing FGF21's inherently unstable core with a thermally stable paracrine FGF1 (sFGF1) . Biophysical characterization using fluorescence spectroscopy and circular dichroism confirms that such chimeras can maintain the critical β-trefoil core structure . Differential calorimetry data demonstrates that these chimeric proteins exhibit significantly greater stability compared to wild-type FGF21 . To further optimize these constructs, researchers should systematically vary the junction points between FGF1 and FGF21 domains, guided by three-dimensional structural data. Testing should include thermal stability assays, receptor binding affinity measurements for both FGFR1c and β-Klotho (essential for FGF21 activity), and in vitro functional assays measuring glucose uptake in adipocytes. The most promising candidates should undergo in vivo evaluation in diabetic mouse models, assessing both acute and chronic effects on glucose homeostasis, insulin sensitivity, and potential side effects. When designing these chimeric proteins, researchers should consider that while FGF21 doesn't require heparin/heparan sulfate (HS) for function, FGF1 does interact with HS, which could influence the biodistribution and activity of the chimeric protein .
The relationship between FGF1 and the mTOR-S6K1 signaling pathway represents a critical mechanistic link in cancer progression, particularly in colorectal cancer (CRC). Recent evidence demonstrates a significant positive correlation between FGF1 and S6K1 (a downstream target of mTOR) expression in CRC tissues, as revealed by both TCGA dataset analysis and immunohistochemical studies . In normal colorectal tissues, no correlation exists between FGF1 and phosphorylated S6K1 (p-S6K1) expression, but their expression becomes tightly associated in tumor tissues . This relationship between FGF1 and the AKT-mTOR-S6K1 axis appears to strengthen as tumors develop—subgroup analysis showed no correlation in early CRC (TNM stage I-II), but a close association in advanced CRC (TNM stage III-IV) .
The functional significance of this relationship has been demonstrated through FGF1 knockdown experiments, which significantly inhibit CRC tumor growth in vivo, as evidenced by reduced tumor size and weight in xenograft models . The increasing correlation between FGF1 and mTOR-S6K1 signaling during tumor progression suggests FGF1 may serve as a biomarker for tumorigenesis and could represent a potential therapeutic target . Researchers investigating this pathway should employ multiple methodological approaches, including correlation analyses between FGF1 and phosphorylated components of the mTOR pathway in patient samples, genetic manipulation of FGF1 in cell lines with subsequent assessment of mTOR signaling, and pharmacological inhibition studies testing the effects of mTOR inhibitors on FGF1-driven phenotypes.
To effectively study the differential effects of FGF1 in normal versus cancer tissues, researchers should implement a multi-faceted approach that combines sophisticated in vitro and in vivo models with clinical sample analysis. For in vitro studies, matched normal and cancer cell lines derived from the same patient or tissue type should be established to minimize genetic background variability. Three-dimensional organoid cultures from both normal and tumor tissues provide more physiologically relevant systems than traditional 2D cultures and better recapitulate tissue architecture and cellular heterogeneity. Gene editing techniques can be employed to create isogenic cell lines with targeted FGF1 pathway alterations to study specific mechanisms, while inducible expression systems allow for temporal control of FGF1 signaling to study acute versus chronic effects.
In vivo models should include conditional FGF1 knockout or overexpression in specific tissue compartments using Cre-lox systems, which enables the study of FGF1 function in normal tissues before and after malignant transformation. Patient-derived xenograft (PDX) models using both normal and tumor tissue from the same patient provide valuable insights into differential responses. Research has shown that FGF1 expression and its association with the AKT-mTOR signaling pathway become increasingly tight as tumors develop , highlighting the importance of studying this relationship across cancer progression stages.
When analyzing clinical samples, matched normal-tumor tissue pairs should be used for comparative analyses of FGF1 expression and downstream signaling. Immunohistochemical staining for FGF1 and related pathway components should be performed, as prior studies have demonstrated no correlation between FGF1 and phosphorylated S6K1 in normal colorectal tissues, but a positive correlation in tumor tissues . Laser capture microdissection can isolate specific cell populations from heterogeneous samples for more precise molecular analyses. Additionally, researchers should consider gender differences in their experimental design, as some studies have observed differential prognostic effects of FGF1 between male and female patients .
Designing experiments to elucidate the neuronal circuits mediating FGF1's central effects on metabolism requires careful consideration of several technical and biological factors. Researchers should employ stereotactic techniques for targeted delivery of FGF1 to specific hypothalamic nuclei to determine which regions are most responsive to FGF1's metabolic effects. Dose-response studies are critical, as is temporal profiling of responses from minutes to weeks post-administration, given that central FGF1 injection produces glucose-lowering effects after approximately one week but sustains this effect beyond 16 weeks . Cell-type specific analyses should focus particularly on tanycytes and astrocytes, as these appear to be primary cellular targets of secreted FGF1 in the brain, rather than neurons directly . Evidence indicates that ependymal cells lining the ventricular space constitute the main source of FGF1 production , and upon glucose stimulation, these cells secrete FGF1 which induces expression of early-response markers selectively in glucose-sensing tanycytes lining the ventral part of the third ventricle and in periventricular hypothalamic astrocytes .
For functional validation, chemogenetic or optogenetic manipulation of identified neuronal populations can test their necessity and sufficiency in mediating FGF1's effects. Conditional knockout approaches targeting FGF receptors in specific cell populations can identify the direct cellular targets of FGF1. Advanced imaging techniques such as fiber photometry or miniature microscopy can monitor real-time activity of specific neuronal populations during FGF1 administration and subsequent metabolic changes. Throughout these studies, researchers should implement appropriate controls for surgical procedures, vehicle effects, and potential stress responses that might independently affect metabolism.
Technical validation should include parallel analysis using multiple detection methods (e.g., qPCR, immunohistochemistry, western blotting) on the same samples to verify consistency across platforms. Rigorous antibody validation for specificity is crucial, as is the standardization of scoring systems for immunohistochemistry. Researchers should carefully define cutoff values for categorizing "high" versus "low" expression based on objective criteria rather than arbitrary thresholds. Biological variables such as tumor heterogeneity, patient demographics, comorbidities, and treatment history should be systematically recorded and analyzed. Cell-type specific analyses may be necessary, as bulk tissue measurements can mask important differences in expression between specific cell populations. Advanced single-cell approaches can resolve cellular heterogeneity that might explain seemingly contradictory results from bulk analyses.
When publishing results, researchers should provide detailed methodological reporting including antibody catalog numbers, dilutions, incubation conditions, primer sequences, and raw data availability. Collaborative validation studies involving multiple laboratories using standardized protocols can help establish reproducibility and resolve contradictions in the field.
Differentiating between direct and indirect effects of FGF1 on glucose metabolism in complex in vivo systems requires sophisticated experimental approaches that isolate specific pathways and temporal sequences. Researchers should implement tissue-specific genetic manipulation using Cre-lox systems targeting FGF receptors in specific tissues (adipose, liver, hypothalamus) to determine which are necessary for FGF1's metabolic effects. When evaluating metabolic outcomes, it's important to consider that peripheral and central FGF1 administration produce distinctly different temporal responses—peripheral injection causes acute glucose lowering within hours while intracerebroventricular injection lowers glucose after about a week but sustains this effect beyond 16 weeks .
Temporal intervention studies can help establish causality by selectively blocking specific pathways at different time points after FGF1 administration. For instance, blocking neural circuits after peripheral FGF1 administration can determine if central mechanisms are required for sustained metabolic improvements. Ex vivo tissue analysis following in vivo FGF1 treatment allows for detailed examination of molecular changes in isolated tissues under controlled conditions. Researchers can use labeled FGF1 (fluorescent or isotope-tagged) to track its biodistribution and identify primary target tissues, with adipose tissue having been identified as a primary target of 'endocrinized' recombinant FGF1, through the FGF1-FGFR1 signaling cascade .
Metabolic flux analysis using isotope tracers can track glucose metabolism pathways in different tissues following FGF1 administration. Pharmacological approaches with tissue-selective drug delivery systems can target specific organs to determine their role in FGF1's metabolic effects. Multi-omics integration (transcriptomics, proteomics, metabolomics) across tissues and time points can help construct pathway models that distinguish direct targets from secondary responses. When designing these experiments, researchers should consider that tanycytes and astrocytes appear to be primary cellular targets of secreted FGF1 in the brain, rather than neurons directly , which might help explain some of the indirect effects observed in metabolic regulation.
Before advancing FGF1 as a diabetes therapeutic candidate, comprehensive preclinical validation must address safety, efficacy, optimal delivery, and patient stratification considerations. Long-term safety studies in multiple species are essential, with particular attention to potential oncogenic risks given FGF1's role in cell proliferation and angiogenesis . Researchers should conduct extensive dose-finding studies to establish minimal effective doses for both peripheral and central administration, noting that these routes produce distinctly different temporal responses—peripheral injection causes acute glucose lowering within hours while central administration lowers glucose after about a week but sustains effects beyond 16 weeks . The remarkable finding that FGF1 achieves glucose lowering without causing hypoglycemia represents a significant safety advantage that should be thoroughly characterized across different conditions.
Efficacy validation should include diverse diabetic models representing different disease etiologies (insulin resistance, β-cell dysfunction), stages, and comorbidities. Comparison studies with existing diabetes therapeutics are necessary to establish potential advantages. For delivery optimization, researchers need to develop formulations that enhance stability and tissue-specific targeting, potentially exploring the enhanced stability observed in FGF1-FGF21 chimeric proteins . Investigation of alternative delivery routes beyond injection (such as oral, inhaled, or transdermal) would enhance clinical applicability. Potential patient stratification biomarkers should be identified, as FGF1 responsiveness may vary based on individual metabolic profiles or genetic factors. Manufacturing and scale-up feasibility must be addressed early, developing robust production processes that maintain FGF1's structural integrity and biological activity. Throughout preclinical development, researchers should also investigate potential differences in efficacy between genders, as some studies have observed differential effects of FGF1 in male versus female subjects .
The unique properties of FGF1 present several innovative therapeutic strategies for addressing metabolic syndrome beyond traditional approaches. FGF1's dual capacity to normalize glucose levels without causing hypoglycemia offers a significant advantage over current diabetes medications that risk dangerous blood sugar drops. This safety profile could enable more aggressive glucose management in patients with variable insulin sensitivity. The remarkable sustained effect of central FGF1 administration (beyond 16 weeks after a single intracerebroventricular injection) suggests potential for long-acting therapies that overcome adherence challenges associated with daily medications. Furthermore, FGF1's effects on both central appetite regulation and peripheral glucose metabolism could simultaneously address multiple components of metabolic syndrome, including obesity and hyperglycemia.
Researchers can explore several innovative therapeutic strategies based on these properties. Structure-based design of FGF1 variants with enhanced stability and receptor selectivity could optimize metabolic benefits while minimizing unwanted effects. The successful development of FGF1-FGF21 chimeric proteins with increased stability demonstrates the feasibility of this approach. Targeted delivery systems could direct FGF1 to specific tissues like adipose (identified as a primary target site) while limiting exposure elsewhere. Combination therapies pairing FGF1 with complementary agents targeting other metabolic syndrome components could produce synergistic benefits. Personalized medicine approaches might identify patient subgroups most likely to benefit from FGF1-based therapies based on specific metabolic profiles or genetic markers.
Advanced formulation technologies could potentially enable non-invasive delivery methods that approximate central administration effects without requiring direct brain injection, making the remarkable sustained glucose-lowering effects of central FGF1 more clinically accessible. Throughout development, researchers should remain attentive to potential gender differences in response, as some studies have noted differential effects of FGF1 between males and females .
To thoroughly evaluate FGF1's therapeutic potential beyond diabetes, researchers must implement experimental designs that address both traditional and novel applications while systematically assessing benefits and risks. For wound healing and tissue repair applications, controlled studies should compare FGF1 with standard-of-care treatments across acute and chronic wound models, evaluating not only healing rates but also tissue quality and functionality. Sophisticated in vivo imaging techniques can track real-time tissue regeneration and vascularization. Given FGF1's role in cell proliferation, survival, migration, invasion, differentiation, and angiogenesis , researchers should carefully monitor for potential oncogenic effects, particularly in chronic administration scenarios.
For neurodegenerative conditions, preclinical models of conditions like Alzheimer's and Parkinson's disease should assess FGF1's effects on neuronal survival, synaptic plasticity, and cognitive or motor outcomes. The finding that ependymal cells lining the ventricular space constitute a main source of FGF1 production in the brain suggests potential neuroregenerative applications worth exploring. In cancer contexts, researchers face a paradox—while FGF1 may promote tumor progression in some contexts , targeted approaches could potentially exploit tumor dependence on FGF1 signaling. Experimental designs should include selective inhibition of FGF1 signaling in FGF1-dependent tumors while monitoring for both intended anti-tumor effects and potential complications from disrupting normal tissue FGF1 function.
For cardiovascular applications, ischemia-reperfusion models can assess FGF1's angiogenic and cardioprotective properties, while atherosclerosis models can evaluate effects on plaque formation and stability. The chimeric FGF1-FGF21 approach might be particularly valuable for metabolic conditions with cardiovascular components. Throughout these diverse applications, researchers should implement tissue-specific and inducible expression systems to control FGF1 activity with temporal and spatial precision, minimizing potential systemic effects. Long-term safety monitoring is essential across all potential therapeutic applications, with particular attention to unexpected effects from disrupting FGF1's physiological roles in multiple tissues.
Purification strategy development must balance yield, purity, and retention of biological activity. Multi-step chromatography approaches are typically required, often including heparin affinity chromatography since FGF1 naturally binds heparin . Quality control testing should verify protein identity (mass spectrometry, N-terminal sequencing), purity (SDS-PAGE, HPLC), endotoxin levels (LAL assay), and biological activity (cell-based proliferation assays). Batch-to-batch consistency is essential for research reproducibility and requires stringent standardization of production protocols. For long-term storage, researchers must establish conditions that maintain FGF1 stability and activity, typically involving lyophilization or storage in the presence of stabilizing agents. Addressing these challenges is crucial for ensuring that experimental outcomes reflect true biological effects rather than artifacts of variable protein quality.
Emerging technologies present unprecedented opportunities to elucidate the temporal and spatial dynamics of FGF1 signaling with resolution and precision previously unattainable. Advanced live-cell imaging techniques utilizing fluorescent protein-tagged FGF1 and its receptors enable real-time visualization of protein movements, interactions, and signaling events within living cells. Super-resolution microscopy methods like STORM, PALM, and STED can resolve FGF1-receptor interactions at the nanoscale, potentially revealing signaling microdomains not visible with conventional microscopy. Optogenetic and chemogenetic tools allow for precise temporal control of FGF1 signaling pathway components, enabling researchers to dissect the sequence of events following receptor activation and determine critical timing windows for different downstream effects.
CRISPR-based genomic tagging facilitates endogenous labeling of FGF1 pathway components, providing more physiologically relevant visualization than overexpression systems. Biosensor technologies can monitor second messengers and protein modifications in real time following FGF1 stimulation, revealing the dynamics of pathway activation. Single-cell multi-omics approaches can characterize cell-to-cell variability in FGF1 responses and identify distinct cellular states that influence responsiveness to FGF1. These technologies are particularly valuable for understanding the differential effects of FGF1 in normal versus cancer tissues, where varying expression levels and correlations with signaling pathways like mTOR-S6K1 have been observed .
For in vivo applications, intravital microscopy allows visualization of FGF1 signaling in living organisms, while tissue clearing methods combined with light-sheet microscopy enable whole-organ imaging of FGF1 distribution and activity. These approaches could help explain the distinct temporal effects observed between peripheral and central FGF1 administration—peripheral injection triggers acute glucose lowering within hours while central administration produces effects after about a week that persist beyond 16 weeks . Spatially resolved transcriptomics and proteomics can map FGF1-induced changes across tissues with subregional resolution, potentially revealing localized signaling hubs that coordinate broader metabolic responses.
Elucidating the mechanistic differences between FGF1, FGF2, and FGF21 in metabolic regulation requires integrated experimental approaches that examine receptor specificity, tissue tropism, and downstream signaling pathways. Comparative binding and activation studies using surface plasmon resonance, biolayer interferometry, and cell-based receptor activation assays can systematically quantify the affinities and activation potentials of each FGF for different receptor isoforms. The findings that FGF1 and FGF2 both influence central regulation of food intake but only FGF1 displays significant glucose-lowering effects after peripheral injection highlight the need for detailed receptor specificity analysis. Domain-swapping experiments between these FGFs can identify the structural determinants responsible for their distinct metabolic activities, building upon the success of FGF1-FGF21 chimeric proteins with enhanced stability and maintained function .
Tissue-specific knockout models of FGF receptors can determine which tissues mediate the metabolic effects of each growth factor. For example, adipose tissue has been identified as a primary target of FGF1 through the FGF1-FGFR1 signaling cascade , but comparative studies are needed to determine if FGF2 and FGF21 act through the same or different target tissues. Parallel metabolic phenotyping in response to each FGF should include comprehensive assessment of glucose homeostasis, insulin sensitivity, energy expenditure, and substrate utilization across multiple tissues. The remarkable sustained effect of central FGF1 administration (glucose lowering for beyond 16 weeks after a single intracerebroventricular injection) should be directly compared with central FGF2 and FGF21 administration to determine if this is a unique property of FGF1.
Molecular profiling using RNA-seq, proteomics, and phosphoproteomics across multiple tissues and time points can identify shared and distinct signaling pathways activated by each FGF. Attention should be given to the role of cofactors—FGF1 requires heparin/heparan sulfate for function, while FGF21 operates in an endocrine manner without requiring heparan sulfate . These mechanistic insights are crucial for optimizing therapeutic applications and potentially developing next-generation FGF-based metabolic regulators with enhanced efficacy and specificity.
Fibroblast Growth Factor-acidic (FGF-acidic), also known as FGF-1, is a member of the fibroblast growth factor family, which is involved in a wide range of biological processes including cell growth, development, and tissue repair. The human recombinant form of FGF-acidic consists of 154 amino acids and is produced using recombinant DNA technology.
FGF-acidic is a heparin-binding protein that plays a crucial role in cell proliferation, differentiation, and survival. It interacts with high-affinity transmembrane receptors, known as fibroblast growth factor receptors (FGFRs), to initiate a cascade of downstream signaling pathways. These pathways are essential for various cellular processes such as angiogenesis, wound healing, and embryonic development.
The human recombinant form of FGF-acidic is produced using Escherichia coli (E. coli) expression systems. The gene encoding FGF-acidic is inserted into a plasmid vector, which is then introduced into E. coli cells. These cells are cultured under specific conditions to express the FGF-acidic protein. The protein is then purified through various chromatographic techniques to achieve high purity and biological activity.