FGFR1 Human

Fibroblast Growth Factor Receptor 1 Fc Chimera Human Recombinant
Shipped with Ice Packs
In Stock

Description

Normal Physiological Roles

FGFR1 regulates:

  • Embryonic Development:

    • Craniofacial bone formation, limb induction, and neural tube development .

    • Critical for GnRH neuron migration via FGF8-FGFR1 signaling .

  • Neurological Function:

    • Survival and migration of olfactory and GnRH neurons .

  • Cellular Processes:

    • Angiogenesis, wound healing, and epithelial-mesenchymal interactions .

Cancer

FGFR1 amplification or overexpression is implicated in multiple cancers:

Cancer TypeFGFR1 Amplification (%)Protein Overexpression (%)Prognostic Impact
Lung SQCLC23% 9% No survival link
HNSCC~20% ~35% Poor OS (HR: 1.97)
BreastN/AN/APromotes invasion

Key Findings:

Genetic Disorders

FGFR1 mutations cause congenital syndromes:

SyndromeMutation TypeKey Features
Pfeiffer SyndromeActivating (P232R)Craniosynostosis, mid-face deformities
Kallmann SyndromeInactivating (R622X)Hypogonadotropic hypogonadism, anosmia
Antley-BixlerInactivating (I300T)Craniofacial abnormalities, skeletal defects

Mechanisms of Nuclear Translocation

FGFR1 exhibits dual localization:

  1. Membrane-Bound: Triggers canonical signaling (e.g., Ras/MAPK).

  2. Nuclear:

    • Pathways:

      • FGF-2/FGFR1 cotransport via importin-β .

      • Granzyme B-mediated cleavage (breast cancer) .

    • Nuclear Populations:

      • Fast mobile: Cytosolic/nucleoplasmic.

      • Slow mobile: Chromatin-bound.

      • Immobile: Nuclear matrix-associated .

Functional Impact:

  • Regulates CREB-binding protein (CBP) and RSK1 to activate genes involved in differentiation .

  • Modulates transcription of migration-related genes (e.g., PRSS27, SFN) .

Therapeutic Targeting and Challenges

  • Potential: FGFR1 inhibitors (e.g., erdafitinib) show promise in FGFR1-amplified cancers.

  • Limitations:

    • Lung Cancer: Amplification does not always correlate with protein expression .

    • Heterogeneity: Variable response rates due to compensatory pathways .

Research Gaps and Future Directions

  1. Nuclear FGFR1 Signaling: Elucidate transcriptional targets and mechanisms.

  2. Biomarker Development: Identify predictive markers for FGFR1 inhibitor response.

  3. Syndrome Pathogenesis: Investigate isoform-specific roles in craniofacial development.

Product Specs

Introduction
The Fibroblast Growth Factor (FGF) family consists of at least 18 structurally related proteins involved in various physiological and pathological processes. These processes include cell growth, differentiation, angiogenesis, wound healing, and tumorigenesis. FGFs exert their biological effects by binding to and activating type I transmembrane tyrosine kinase receptors. Upon ligand binding, these receptors dimerize and autophosphorylate. Four distinct genes encode closely related FGF receptors: FGFR-1, -2, -3, and -4. Alternative splicing of mRNAs generates multiple forms of FGFR-1 to -3. A common splicing event in FGFR-1 and -2 produces receptors containing all three immunoglobulin-like domains (Ig domains) termed the alpha isoform. Alternatively, receptors containing only IgII and IgIII are termed the beta isoform. Only the alpha isoform has been identified for FGFR-3 and FGFR-4. Further splicing events for FGFR-1 to -3 involve the C-terminal half of the IgIII domain. These events result in FGF receptors with alternative IgIII domains (IIIb and IIIc) generated by two mutually exclusive alternative exons. A secreted FGF binding protein, the IIIa isoform, containing only the N-terminal half of the IgIII domain and some intron sequences, has also been reported for FGFR-1. Notably, mutations in FGFR-1 to -3 have been identified in patients with birth defects involving craniosynostosis.
Description
Soluble FGFR-1a (IIIc) Fc Chimera Human Recombinant, incorporating a Xa cleavage site and the Fc domain of human IgG1, is produced in a baculovirus expression system. This chimeric protein is a heterodimeric, glycosylated polypeptide chain comprising 601 amino acids with a molecular weight of 170 kDa. The purification of FGFR1 is achieved using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
CD331 was lyophilized from a sterile solution at a concentration of 1 mg/ml in 1xPBS.
Solubility
To reconstitute the lyophilized bFGF-R, it is recommended to dissolve it in sterile PBS at a minimum concentration of 100 µg/ml. The reconstituted solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized FGFR1A can be stored at room temperature for up to 3 weeks; however, for extended storage, it is recommended to store it desiccated at a temperature below -18°C. After reconstitution, FGFR1 should be stored at 4°C for a period of 2-7 days. For long-term storage, it is advisable to store it below -18°C. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of this product is greater than 90.0% as determined by SDS-PAGE analysis.
Biological Activity
The biological activity of this product is determined based on its ability to inhibit human FGF acidic-dependent proliferation in R1 cells. The ED50 for this inhibitory effect is typically in the range of 15.0-30.0 ng/ml.
Synonyms
FGFR-1, bFGF-R, C-FGR, CD331, fms-related tyrosine kinase 2, Pfeiffer syndrome, CEK, FLG, FLT2, KAL2, BFGFR, FGFBR, HBGFR, FGFR1/FGFR1OP2 FUSION GENE, FGFR1/ZNF198 FUSION GENE, FLG FGFR1/BCR FUSION GENE, FLG protein, FMS-LIKE GENE, N-sam tyrosine kinase, basic fibroblast growth factor receptor 1.
Source
Insect Cells.
Amino Acid Sequence

RPSPTLPEQAQPWGAPVEVESFLVHPGDLLQLRCRLRDDVQSINWLRDGVQL AESNRTRITGEEVEVQDSVPADSGLYACVTSSPSGSDTTYFSVNVSDALPSS EDDDDDDDSSSEEKETDNTKPNRMPVAPYWTSPEKMEKKLHAVPAAKTVKFK CPSSGTPNPTLRWLKNGKEFKPDHRIGGYKVRYATWSIIMDSVVPSDKGNYT CIVENEYGSINHTYQLDVVERSPHRPILQAGLPANKTVALGSNVEFMCKVYS DPQPHIQWLKHIEVNGSKIGPDNLPYVQILKTAGVNTTDKEMEVLHLRNVSF EDAGEYTCLAGNSIGLSHHSAWLTVLEALEERPAVMTSPLYLEDPRRASIEG RGDPEEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL NGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK

Q&A

What is the normal function of FGFR1 in human cells?

FGFR1 is a transmembrane receptor tyrosine kinase that plays crucial roles in multiple cellular processes. The protein spans the cell membrane with one end inside the cell and the other extending from the outer surface, allowing it to interact with fibroblast growth factors (FGFs) in the extracellular environment. When an FGF attaches to the FGFR1 protein, it initiates a cascade of intracellular chemical reactions that regulate cell behavior. FGFR1 signaling is involved in cell division, growth regulation, maturation, blood vessel formation (angiogenesis), wound healing, and embryonic development.

Specifically, FGFR1 is essential for the development and growth of several body structures, including:

  • Brain development

  • Craniofacial bone formation

  • Development of bones in the hands and feet

  • Formation of long bones in the arms and legs

Experimental approaches to study FGFR1 function typically involve gene knockout models, receptor-specific antibodies, and phosphorylation assays to assess downstream signaling activation.

How does FGFR1 signaling operate at the molecular level?

FGFR1 signaling occurs through a complex mechanism involving ligand binding, receptor dimerization, and activation of downstream pathways:

  • Ligand Binding and Dimerization: When FGF ligands bind to the extracellular domain of FGFR1, the receptor undergoes homo- or hetero-dimerization with other FGFRs.

  • Phosphorylation Cascade: Dimerization triggers sequential phosphorylation of specific intracellular tyrosine residues, activating the intracellular signaling cascade.

  • Pathway Activation: FGFR1 signaling intersects with several major cellular pathways, particularly:

    • PI3K/Akt pathway

    • MAPK pathway

    • Wnt signaling

    • Hedgehog pathway

    • Bone morphogenic protein (BMP) signaling

These pathways collectively regulate gene transcription that controls cellular processes such as proliferation, differentiation, and survival. Experimental investigation of FGFR1 signaling typically employs phospho-specific antibodies, pathway inhibitors, and reporter assays to dissect the contribution of individual downstream components.

What are the known FGF ligands that interact with FGFR1?

FGFR1 can interact with multiple FGF ligands, each with different binding affinities depending on the receptor isoform. There are 18 known human FGF ligands that can potentially interact with FGFR family members.

For FGFR1 specifically, the binding affinity varies by receptor splice variant:

  • FGFR1b isoform: Higher affinity for FGF1, FGF3, FGF7, FGF10, and FGF22

  • FGFR1c isoform: Higher affinity for FGF1, FGF2, FGF4, FGF6, FGF8, FGF9, and FGF20

The specificity of these interactions is determined by both the structural complementarity between ligands and receptors and tissue-specific expression patterns. Methodologically, binding studies using surface plasmon resonance (SPR), co-immunoprecipitation assays, and fluorescently-labeled ligands are commonly employed to characterize ligand-receptor interactions.

How do mutations in FGFR1 lead to inherited disorders?

FGFR1 mutations can cause several hereditary conditions, primarily through gain-of-function or loss-of-function mechanisms:

Loss-of-Function Mutations:

  • Hypogonadotropic hypogonadism with or without anosmia (Kallmann syndrome/KAL2): This autosomal dominant condition results from loss of FGFR1 function, affecting the development and migration of GnRH-producing neurons. These neurons are essential for controlling the production of reproductive hormones, explaining the hypogonadism phenotype.

Gain-of-Function Mutations:

  • Craniosynostosis syndromes (including Pfeiffer, Crouzon, and Jackson-Weiss syndromes): These conditions involve premature fusion of skull bones due to enhanced FGFR1 signaling.

Experimental approaches to study these mutations include:

  • Patient-derived cells for signaling studies

  • CRISPR/Cas9-mediated introduction of specific mutations in cell lines

  • Transgenic animal models expressing human mutation variants

  • Structural biology approaches to understand how mutations alter receptor conformation and function

What is the role of FGFR1 in neural development and GnRH signaling?

FGFR1 plays a critical role in neural development, particularly in the formation, survival, and migration of neurons in several brain regions. The protein is especially important for:

  • GnRH Neuronal Development: FGFR1 signaling is essential for neurons that produce gonadotropin-releasing hormone (GnRH). These neurons originate in the olfactory placode and migrate to the hypothalamus during development.

  • Hormonal Regulation: GnRH controls the production of several other hormones that direct sexual development before birth and during puberty, explaining why FGFR1 mutations can affect reproductive function.

  • Olfactory Processing: FGFR1 appears to be involved in the processing of smells by specialized olfactory neurons, which explains why some patients with FGFR1 mutations experience anosmia (loss of smell).

Research methodologies to investigate FGFR1's role in neural development include lineage tracing experiments, conditional knockout models, and ex vivo neuronal cultures to assess migration, differentiation, and functional connectivity.

Why don't individuals with hereditary FGFR1 mutations show increased cancer risk despite similar mutations occurring in tumors?

This represents a fascinating paradox in FGFR1 biology. Despite some activating FGFR1 mutations being identical between hereditary syndromes and cancers, individuals with FGFR1-related syndromes don't generally show increased cancer incidence.

Several hypotheses explain this phenomenon:

  • Developmental Compensation: The presence of germline mutations throughout development may activate compensatory mechanisms that protect against neoplastic transformation.

  • Tissue Context Dependency: The effect of FGFR1 mutations likely depends on the cellular context, with different outcomes in different tissues.

  • Temporal Factors: Germline mutations present from embryonic development versus somatic mutations acquired later in life may trigger different cellular responses.

  • Additional Genetic Events: Cancer development typically requires multiple genetic hits beyond the initial FGFR1 mutation.

This area remains under investigation, with approaches including comparative transcriptomics between syndrome-affected cells and cancer cells with identical FGFR1 mutations, and development of tissue-specific conditional knockin models.

How does FGFR1 signaling affect glucose metabolism and insulin sensitivity?

Recent research has unveiled an important role for FGFR1 in metabolic regulation, particularly glucose homeostasis:

A 2024 study published in the Journal of the Endocrine Society demonstrated that individuals with rare deleterious FGFR1 variants exhibit metabolic alterations compared to non-carrier controls. Specifically:

  • FGFR1-mutation carriers displayed higher β-cell function coupled with lower insulin sensitivity compared to controls

  • These findings suggest that impaired FGFR1 signaling may contribute to an early insulin resistance phase of diabetes pathogenesis

  • The study utilized a frequently sampled intravenous glucose tolerance test to evaluate these parameters

This human genetic evidence supports previous animal studies showing that activation of FGFR1 signaling improves metabolic health, while inactivation leads to diabetes in mice. The findings point to the FGFR1 signaling pathway as a potential therapeutic target for metabolic disorders.

Methodologically, researchers investigating FGFR1's metabolic functions employ glucose tolerance tests, hyperinsulinemic-euglycemic clamps, metabolic tracer studies, and tissue-specific conditional knockout models to dissect pathway-specific effects.

What experimental approaches best reveal the intersection between FGFR1's reproductive and metabolic functions?

The dual role of FGFR1 in both reproduction and metabolism represents an important research area. Effective experimental approaches include:

  • Recall-by-genotype Studies: Identifying individuals with FGFR1 variants and comprehensively phenotyping both their reproductive and metabolic parameters, as exemplified by Stamou et al. (2024).

  • Tissue-Specific Conditional Models: Using Cre-lox technology to delete or modify FGFR1 in specific tissues (hypothalamus, pancreatic β-cells, adipose tissue) to dissect tissue-specific contributions.

  • Metabolic Challenge Tests: Subjecting FGFR1-variant models to metabolic challenges (high-fat diet, glucose/insulin tolerance tests) while monitoring reproductive parameters.

  • Single-cell Transcriptomics: Identifying cell populations at the intersection of metabolic and reproductive regulation that express FGFR1 and downstream effectors.

  • Hormonal Profiling: Comprehensive analysis of both reproductive hormones (GnRH, LH, FSH) and metabolic hormones (insulin, glucagon, incretins) in FGFR1-variant models.

These approaches collectively help researchers understand how FGFR1 integrates metabolic status with reproductive function, a relationship that has been known clinically but whose molecular basis remains incompletely understood.

What are the mechanisms of FGFR1 dysregulation in human cancers?

FGFR1 dysregulation contributes to cancer development and progression through several mechanisms:

  • Gene Amplification: Increased FGFR1 gene copy number, particularly common in:

    • Lung squamous cell carcinoma

    • Breast cancer

    • Head and neck squamous cell carcinoma

  • Overexpression: Elevated FGFR1 protein levels without gene amplification, suggesting transcriptional or post-transcriptional regulation.

  • Activating Mutations: Somatic mutations that enhance receptor activity or ligand binding affinity.

  • Chromosomal Rearrangements: The 8p11 myeloproliferative syndrome involves chromosomal translocations creating fusion proteins with constitutive FGFR1 activation.

  • Autocrine/Paracrine Signaling Loops: Cancer cells may produce both FGFR1 and its ligands, creating a self-stimulatory growth circuit.

Importantly, FGFR1 amplification doesn't always correlate with protein expression, highlighting the need for comprehensive biomarker analysis beyond genomic testing. Research approaches include fluorescence in situ hybridization (FISH) for amplification detection, immunohistochemistry for protein expression, and functional assays to characterize pathway activation.

How do different cancer types exhibit varying dependencies on FGFR1 signaling?

Cancer dependence on FGFR1 signaling shows significant heterogeneity across tumor types:

Methodologically, researchers assess FGFR1 dependency through:

  • CRISPR/Cas9 knockout screens

  • Small molecule inhibitor sensitivity profiling

  • Phospho-proteomic analysis of downstream pathway activation

  • Patient-derived xenograft models to evaluate therapeutic responses

Understanding the precise nature of FGFR1 dependency in each cancer type is critical for developing effective targeted therapies and identifying appropriate biomarkers for patient selection.

What strategies show promise for targeting FGFR1 in cancer therapy?

Several therapeutic approaches targeting FGFR1 in cancer are under investigation:

  • Small Molecule Tyrosine Kinase Inhibitors:

    • Pan-FGFR inhibitors (e.g., erdafitinib, infigratinib)

    • Selective FGFR1 inhibitors

    • Dual FGFR/VEGFR inhibitors

  • Antibody-Based Approaches:

    • Anti-FGFR1 monoclonal antibodies

    • Antibody-drug conjugates targeting FGFR1

  • Novel Modalities:

    • FGFR1 degraders (PROTACs)

    • RNA-based therapeutics (siRNA, antisense oligonucleotides)

  • Combination Strategies:

    • FGFR1 inhibitors plus immune checkpoint inhibitors

    • FGFR1 inhibitors plus endocrine therapy in breast cancer

    • FGFR1 inhibitors plus chemotherapy

Clinical trials are ongoing, including for lung cancer patients with FGFR1 amplification (e.g., NCT01004224). Key research challenges include:

  • Identifying robust biomarkers beyond gene amplification

  • Understanding resistance mechanisms

  • Mitigating toxicities associated with FGFR inhibition

  • Developing rational combination regimens

Research methodologies include high-throughput drug screening, combination synergy studies, and integrated genomic and proteomic analyses to identify biomarkers of response.

What are the optimal experimental models for studying FGFR1 signaling specificity?

Researching FGFR1 signaling specificity requires sophisticated experimental models that can distinguish among complex receptor-ligand interactions and downstream effects:

  • Receptor Isoform-Specific Models:

    • CRISPR-engineered cell lines expressing single FGFR1 splice variants (e.g., FGFR1b or FGFR1c)

    • Isoform-specific knockin/knockout mice to assess in vivo specificity

  • Ligand-Receptor Interaction Platforms:

    • Proximity ligation assays to detect specific FGF-FGFR1 interactions in situ

    • Bioluminescence/fluorescence resonance energy transfer (BRET/FRET) systems to measure real-time binding dynamics

    • Surface plasmon resonance for quantitative binding affinity determination

  • Pathway Tracing Methods:

    • Phospho-proteomic approaches to map signaling cascades

    • Temporal signaling analysis using rapid inhibitor washout techniques

    • Optogenetic control of FGFR1 activation for precise temporal resolution

  • Tissue-Specific Models:

    • Organoid cultures expressing endogenous FGFR1

    • Conditional tissue-specific FGFR1 manipulation in vivo

    • Patient-derived primary cells maintaining physiological receptor levels

When designing these experiments, researchers must carefully consider the expression levels of both receptor and ligands, as overexpression can alter specificity profiles compared to physiological conditions.

How can researchers resolve contradictory findings on FGFR1 function between model systems?

Contradictory findings across different model systems studying FGFR1 are common and require systematic approaches to resolution:

  • Standardization of Experimental Conditions:

    • Consistent expression levels of FGFR1 and ligands

    • Defined matrix components when studying extracellular interactions

    • Standardized readouts for pathway activation

  • Comprehensive Context Analysis:

    • Characterization of cell-type specific co-receptors and modulators

    • Assessment of compensatory mechanisms in different models

    • Evaluation of cross-talk with other signaling pathways

  • Multi-system Validation Approaches:

    • Parallel studies in cell lines, primary cells, and in vivo models

    • Cross-validation between human samples and model systems

    • Computational models integrating data across platforms

  • Genetic Background Considerations:

    • Use of isogenic cell lines differing only in FGFR1 status

    • Backcrossing animal models to control genetic background

    • Analysis of human genetic variation when comparing patient samples

  • Temporal Resolution:

    • Distinguishing acute versus chronic FGFR1 signaling effects

    • Developmental stage-specific analyses

    • Aging effects on FGFR1 signaling in model systems

When conflicts arise, researchers should systematically eliminate variables by designing experiments that directly compare conditions side-by-side rather than relying on historical controls from different laboratory environments.

What are the technical challenges in measuring FGFR1 activation status in patient samples?

Assessing FGFR1 activation in patient samples presents several technical challenges:

  • Tissue Preservation Issues:

    • Phosphorylation status rapidly changes during sample collection

    • Conventional fixation methods can affect phospho-epitope detection

    • Freezing artifacts may disrupt membrane receptor organization

  • Heterogeneity Challenges:

    • Mixed cell populations dilute signal from FGFR1-positive cells

    • Spatial heterogeneity requires analysis of multiple regions

    • Temporal variation in signaling requires standardized collection timing

  • Specificity Problems:

    • Cross-reactivity between FGFR family members

    • Limited availability of phospho-site specific antibodies

    • Difficulty distinguishing receptor isoforms

  • Quantification Limitations:

    • Narrow dynamic range of immunohistochemistry

    • Sample amount constraints for phospho-proteomic approaches

    • Lack of universal standards for quantification

Methodological solutions include:

  • Rapid sample preservation techniques (e.g., cold formalin fixation)

  • Phosphatase inhibitors during sample collection

  • Multiplexed analysis combining receptor levels with downstream markers

  • Single-cell approaches to address heterogeneity

  • Proximity ligation assays to detect specific receptor-ligand complexes

These technical considerations are crucial when designing biomarker studies for FGFR1-targeted therapies.

How might single-cell technologies advance our understanding of FGFR1 biology?

Single-cell technologies hold tremendous potential for resolving complex questions in FGFR1 biology:

  • Heterogeneity Resolution:

    • Single-cell RNA sequencing can identify distinct cell populations with unique FGFR1 expression patterns

    • Cell-specific co-expression of FGFR1 with ligands, co-receptors, and modulators

    • Identification of rare cell populations with extreme FGFR1 dependency

  • Spatial Context Analysis:

    • Spatial transcriptomics and proteomics can map FGFR1 signaling networks within tissue architecture

    • Analysis of ligand gradients and their relationship to receptor activation

    • Mapping of cell-cell communication networks involving FGFR1

  • Temporal Dynamics:

    • Single-cell trajectory analysis to understand how FGFR1 signaling influences cell fate decisions

    • Real-time single-cell imaging of FGFR1 activation in live tissues

    • Temporal analysis of signaling during development or disease progression

  • Multi-omic Integration:

    • Combined single-cell genomics, transcriptomics, and proteomics to link FGFR1 genetic variants to functional outcomes

    • Correlation of FGFR1 expression with epigenetic states at single-cell resolution

    • Metabolomic profiling to connect FGFR1 signaling with metabolic phenotypes

These technologies will help resolve long-standing questions about cell-type specificity of FGFR1 function and the precise mechanisms underlying the pleiotropic effects of FGFR1 signaling across different physiological and pathological contexts.

What emerging regulatory mechanisms for FGFR1 warrant further investigation?

Several emerging regulatory mechanisms for FGFR1 deserve focused research attention:

  • Non-coding RNA Regulation:

    • microRNAs targeting FGFR1 mRNA

    • Long non-coding RNAs modulating FGFR1 transcription or mRNA stability

    • Circular RNAs acting as miRNA sponges affecting FGFR1 expression

  • Alternative Splicing Machinery:

    • Factors controlling FGFR1 isoform expression

    • Tissue-specific splicing regulators

    • Splicing alterations in disease contexts

  • Post-translational Modifications:

    • Beyond phosphorylation: ubiquitination, SUMOylation, glycosylation patterns

    • Crosstalk between different PTMs on FGFR1

    • Enzymes responsible for these modifications as potential therapeutic targets

  • Receptor Trafficking Dynamics:

    • Endosomal signaling of internalized FGFR1

    • Recycling versus degradation pathways

    • Spatial compartmentalization of signaling

  • Extracellular Matrix Interactions:

    • Role of specific heparan sulfate proteoglycan structures

    • Matrix-sequestered versus free FGF ligands

    • FGFBP1 and other FGF-binding proteins as signaling modulators

Research methodologies should include CRISPR screens to identify regulatory factors, advanced imaging to track receptor trafficking, and structural biology approaches to understand conformational changes associated with different regulatory states.

How can systems biology approaches integrate diverse FGFR1 datasets to generate new hypotheses?

Systems biology offers powerful frameworks for integrating complex FGFR1 data and generating testable hypotheses:

  • Network Modeling Approaches:

    • Construction of FGFR1 signaling networks incorporating protein-protein interactions, genetic dependencies, and transcriptional responses

    • Prediction of key nodes and bottlenecks in signaling

    • Identification of feedback and feed-forward loops regulating response dynamics

  • Multi-scale Integration:

    • Linking molecular-level FGFR1 signaling to cellular behaviors and tissue-level phenotypes

    • Mathematical modeling of ligand diffusion and gradient formation

    • Connecting genomic variation to functional outcomes through pathway analysis

  • Comparative Systems Analysis:

    • Cross-species comparison of FGFR1 networks to identify evolutionarily conserved modules

    • Comparison between normal and disease states to identify critical alterations

    • Analysis across tissue types to identify context-specific network components

  • Therapeutic Response Prediction:

    • In silico prediction of cellular responses to FGFR1 targeted therapies

    • Identification of potential resistance mechanisms through network perturbation analysis

    • Rational design of combination therapies based on network vulnerability points

  • Data Integration Platforms:

    • Development of resources that integrate genomic, transcriptomic, proteomic, and clinical data related to FGFR1

    • Machine learning approaches to identify patterns across diverse datasets

    • Interactive visualization tools to explore complex FGFR1 relationships

These systems approaches can help resolve apparent contradictions in the literature and generate novel hypotheses about FGFR1 function that might be overlooked in more reductionist experimental paradigms.

Product Science Overview

Introduction

Fibroblast Growth Factor Receptor 1 (FGFR1) is a crucial member of the fibroblast growth factor receptor family, which plays a significant role in various cellular processes, including cell growth, differentiation, angiogenesis, and wound healing . The FGFR1 Fc Chimera is a recombinant protein that combines the extracellular domain of FGFR1 with the Fc region of human immunoglobulin G (IgG), enhancing its stability and functionality .

Structure and Function

FGFR1 consists of an extracellular region with three immunoglobulin-like domains, a single hydrophobic membrane-spanning segment, and a cytoplasmic tyrosine kinase domain . The extracellular portion interacts with fibroblast growth factors (FGFs), initiating a cascade of downstream signals that influence mitogenesis and differentiation .

The Fc Chimera variant of FGFR1 is engineered to include the Fc region of human IgG, which provides several advantages:

  • Increased Stability: The Fc region enhances the stability of the recombinant protein, making it more suitable for various applications .
  • Improved Purification: The Fc region allows for easier purification using protein A or G affinity chromatography .
  • Enhanced Biological Activity: The Fc Chimera can inhibit FGF-dependent proliferation of cells, making it useful in research and therapeutic applications .
Applications

The FGFR1 Fc Chimera has several applications in biomedical research and therapeutic development:

  • Cancer Research: FGFR1 is implicated in various cancers, and the Fc Chimera can be used to study its role in tumorigenesis and as a potential therapeutic target .
  • Developmental Biology: FGFR1 is involved in limb induction and other developmental processes, making the Fc Chimera a valuable tool for studying these pathways .
  • Drug Development: The Fc Chimera can be used in high-throughput screening assays to identify potential inhibitors of FGFR1 signaling .
Production and Purification

The recombinant FGFR1 Fc Chimera is typically produced in mammalian cell lines, such as NS0 or CHO cells, to ensure proper folding and post-translational modifications . The protein is purified using affinity chromatography, and its purity is assessed by SDS-PAGE and other analytical techniques .

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.