FGFR2 Human, His is a truncated or full-length recombinant protein comprising:
Extracellular domain: Three immunoglobulin (Ig)-like domains (IgI, IgII, IgIII) for ligand binding.
Transmembrane domain: A single hydrophobic segment anchoring the protein to the cell membrane.
Intracellular tyrosine kinase domain: Phosphorylates downstream targets to initiate signaling cascades .
Alternative splicing of the IgIII domain generates two major isoforms:
Isoform | Ligand Specificity | Tissue Expression |
---|---|---|
FGFR2b | FGF1, FGF2, FGF9 | Epithelial tissues |
FGFR2c | FGF7, FGF10 | Mesenchymal tissues |
These isoforms exhibit distinct signaling profiles, influencing processes like epithelial-mesenchymal interactions and tissue repair .
FGFR2 Human, His is employed to study receptor-mediated signaling in vitro. Key functions include:
FGFR2 activates multiple pathways upon FGF binding:
Pathway | Key Components | Biological Effects |
---|---|---|
Ras/MAPK | GRB2, SOS, ERK1/2 | Proliferation, differentiation |
PI3K/AKT | PIK3R1, AKT1 | Survival, migration |
PLCγ | IP3, DAG, PKC | Ca²⁺ signaling, cell migration |
STAT | STAT1, STAT3 | Angiogenesis, immune response |
These pathways are disrupted in cancers, where FGFR2 mutations or fusions drive oncogenic signaling .
Recombinant FGFR2 Human, His is utilized in:
Oncogenic Transformation: FGFR2 knockdown in mammary epithelial cells reduces integrin β1 expression, impairing adhesion and migration .
Mutation Analysis: Truncated FGFR2 (ΔE18) variants exhibit constitutive kinase activity, promoting tumor growth in cholangiocarcinoma .
Drug Screening: AZD4547 (FGFR inhibitor) reduces FGFR2 phosphorylation and integrin β1 levels in HB2 cells .
Condition | Mechanism | Therapeutic Targeting |
---|---|---|
Apert Syndrome | S252W/P253R mutations in IgIII | Inhibitors targeting constitutive signaling |
Cholangiocarcinoma | FGFR2-BICC1 fusions | Pemigatinib (FDA-approved FGFR inhibitor) |
Bladder Cancer | S252W hotspot mutations | Erdafitinib (FGFR inhibitor) |
FGFR2 alterations (mutations, fusions) are detected in 10–16% of cholangiocarcinomas and 10% of bladder cancers .
Cancer Type | Mutation | Frequency | Functional Impact |
---|---|---|---|
CHOL | FGFR2-BICC1 fusions | 10–16% | Constitutive kinase activation |
UCEC | S252W | ~10% | Ligand-independent signaling |
BLCA | S252W | ~10% | Increased proliferation |
Construct | Expressed Region | Host System | Purity | Application |
---|---|---|---|---|
FGFR2 α (IIIc) His-tag | ECD (IgIIIc) | HEK293 | >70% | Ligand-binding assays |
FGFR2 (R612T) His-tag | Kinase domain | Sf9 cells | >70% | Kinase activity tests |
EC 2.7.10, FGFR-2, BFR-1, CD332, BBDS, CEK3, ECT1, TK14, TK25, CFD1, KSAM, JWS, Fibroblast Growth Factor Receptor 2, Keratinocyte Growth Factor Receptor, Bacteria-Expressed Kinase, EC 2.7.10.1, K-SAM, KGFR, BEK, Protein Tyrosine Kinase, Receptor Like 14, BEK Fibroblast Growth Factor Receptor, Craniofacial Dysostosis 1, Jackson-Weiss Syndrome, Pfeiffer Syndrome, Crouzon Syndrome, CD332 Antigen.
RPSFSLVEDT TLEPEEPPTK YQISQPEVYV AAPGESLEVR CLLKDAAVIS WTKDGVHLGP NNRTVLIGEY LQIKGATPRD SGLYACTASR TVDSETWYFM VNVTDAISSG DDEDDTDGAE DFVSENSNNK RAPYWTNTEK MEKRLHAVPA ANTVKFRCPA GGNPMPTMRW LKNGKEFKQE HRIGGYKVRN QHWSLIMESV VPSDKGNYTC VVENEYGSIN HTYHLDVVER SPHRPILQAG LPANASTVVG GDVEFVCKVY SDAQPHIQWI KHVEKNGSKY GPDGLPYLKV LKHSGINSSN AEVLALFNVT EADAGEYICK VSNYIGQANQ SAWLTVLPKQ QAPGREKEIT ASPDYLELEP KSCDKTHTCP PCPAPELLGG PSVFLFPPKP KDTLMISRTP EVTCVVVDVS HEDPEVKFNW YVDGVEVHNA KTKPREEQYN STYRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPSRDE LTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT QKSLSLSPGK HHHHHH.
FGFR2 is a receptor tyrosine kinase encoded by the FGFR2 gene located on chromosome 10 in humans. The protein's structure consists of an extracellular region composed of three immunoglobulin domains, a single hydrophobic membrane-spanning segment, and a cytoplasmic tyrosine kinase domain. The extracellular portion interacts with fibroblast growth factors, initiating downstream signaling cascades that influence cellular mitogenesis and differentiation. The amino acid sequence of FGFR2 is highly conserved both between family members and throughout evolutionary history, highlighting its biological significance .
FGFR2 plays critical roles in embryonic development and tissue repair, particularly in bone formation and blood vessel development. On a molecular level, FGFR2 mediates cell division, growth, and differentiation through ligand binding and receptor dimerization, which activates the tyrosine kinase domains to initiate intracellular signaling cascades. These signaling pathways regulate numerous developmental processes and contribute to tissue homeostasis in mature organisms . Disruptions in FGFR2 signaling are associated with several developmental disorders and pathological conditions, including various types of cancer.
FGFR2 has two major naturally occurring isoforms, FGFR2IIIb and FGFR2IIIc, which result from alternative splicing of the third immunoglobulin-like domain. FGFR2IIIb is predominantly expressed in ectoderm-derived tissues and endothelial organ linings (such as skin and internal organs), while FGFR2IIIc is primarily found in mesenchymal tissues, including craniofacial bone. The tissue-specific distribution of these isoforms is functionally significant, as mutations in FGFR2IIIc are associated with craniosynostosis, a condition characterized by premature fusion of cranial sutures . The isoforms also differ in their ligand binding preferences, contributing to tissue-specific responses to various fibroblast growth factors.
FGFR2 expression patterns and genetic alterations vary significantly across different cancer types. Analysis of large comprehensive datasets covering over 10,000 tumor samples across more than 30 cancer types reveals distinct patterns of FGFR2 abnormal expression, methylation, and alterations (mutations, fusions, amplifications, and deletions). For example, cholangiocarcinoma (CHOL) shows the highest frequency of FGFR2 fusions, while uterine corpus endometrial carcinoma (UCEC) exhibits the highest number of FGFR2 mutations, with S252W and N549K being the most frequently mutated positions . These cancer-specific patterns have important implications for diagnosis, prognosis, and therapeutic strategies.
Optimizing expression and purification of His-tagged FGFR2 requires careful consideration of several factors:
Expression System Selection:
Bacterial systems (E. coli): Suitable for cytoplasmic domains, but may struggle with full-length receptor due to glycosylation requirements
Mammalian cell lines: Preferred for full-length FGFR2 with proper post-translational modifications
Insect cell systems: Good compromise between yield and post-translational modifications
Purification Strategy:
Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-NTA resins
Consider adding imidazole gradient elution to reduce non-specific binding
Secondary purification steps (ion exchange, size exclusion chromatography) to achieve higher purity
Buffer Optimization:
Include appropriate detergents for membrane-bound regions
Add stability enhancers such as glycerol or specific ligands
Consider phosphatase inhibitors to preserve phosphorylation states
The inclusion of rigorous quality control steps, including Western blotting and activity assays, is essential to confirm that the purified protein maintains its native conformation and function.
FGFR2 plays a critical role in regulating epithelial cell interactions with the extracellular matrix (ECM) through mechanisms involving integrin expression and function. Experimental knockdown of FGFR2 in HB2 mammary epithelial cells results in decreased protein levels of specific integrins, including α2, α5, and the mature form of β1, while leaving α1, α3, and α6 unchanged. Additionally, treatment with the FGFR inhibitor AZD4547 significantly decreases integrin β1 protein levels after 48 and 72 hours, coinciding with pronounced inhibition of FGFR signaling . These findings suggest a molecular link between FGFR2 activity and epithelial cell-ECM communication, with FGFR2 loss and concomitant integrin β1 degradation responsible for deregulating these interactions.
Integrin | Effect of FGFR2 Knockdown | Timeframe |
---|---|---|
α1 | No significant change | N/A |
α2 | Decreased protein levels | N/A |
α3 | No significant change | N/A |
α5 | Decreased protein levels | N/A |
α6 | No significant change | N/A |
β1 (mature) | Decreased protein levels | N/A |
β1 (with AZD4547) | Significant decrease | 48-72 hours |
Researchers investigating FGFR2-mediated cell adhesion and migration can employ multiple complementary approaches:
Genetic Manipulation:
CRISPR/Cas9-mediated FGFR2 knockout or knockdown
Overexpression of wild-type or mutant FGFR2 constructs
Domain-specific mutations to isolate functional regions
Functional Assays:
Adhesion assays on different ECM components (collagen, fibronectin, laminin)
Wound healing assays to assess collective cell migration
Single-cell tracking for detailed migration parameters
Transwell migration and invasion assays
Molecular Analysis:
Co-immunoprecipitation to identify FGFR2-integrin interactions
Western blotting to quantify integrin expression levels and activation states
Immunofluorescence to visualize focal adhesion formation and cytoskeletal organization
Pharmacological Approaches:
Specific FGFR2 inhibitors (e.g., AZD4547) to block signaling
Integrin-blocking antibodies to dissect downstream effects
Pathway-specific inhibitors to determine signaling mechanisms
These approaches, used in combination, can provide comprehensive insights into how FGFR2 regulates cellular interactions with the extracellular environment.
FGFR2 alterations in cancer include mutations, fusions, and amplifications, with distinct patterns across different cancer types. In cholangiocarcinoma, FGFR2 fusions are the predominant alteration and are highly targetable, with FDA-approved drugs showing efficacy against these alterations . In contrast, uterine corpus endometrial carcinoma exhibits a high frequency of point mutations, particularly S252W and N549K, which have oncogenic effects but show less response to targeted therapies .
The alteration type significantly influences therapeutic outcomes:
Cancer Type | Predominant Alteration | Therapeutic Implication |
---|---|---|
Cholangiocarcinoma | FGFR2 fusions | Highly targetable; FDA-approved drugs available |
Endometrial Carcinoma | S252W and N549K mutations | Oncogenic effect; less responsive to therapy |
Various Cancers | Amplifications | Variable response to inhibitors |
Clinical trials targeting FGFR2 rearrangements in cholangiocarcinoma have shown objective remission rates of 14-35% and disease control rates of 75-83%, with progression-free survival of 5.8-6.9 months . The FDA has approved pemigatinib, an FGFR inhibitor, for treating cholangiocarcinomas with FGFR2 rearrangements that have failed prior chemotherapy .
In genomic analyses, FGFR2 fusions involve various partner genes, with FGFR2-BICC1 being the most common fusion type in cholangiocarcinoma. The most frequent concomitant genetic alterations with FGFR2 rearrangements include BAP1, CDKN2A, and CDKN2B mutations, while KRAS and IDH1 mutations appear to be mutually exclusive with FGFR2 rearrangements . Understanding these molecular and clinical associations is crucial for patient stratification and treatment selection in precision oncology approaches.
Structure-based computational approaches offer powerful tools for developing novel FGFR2 inhibitors. An integrated bioinformatics workflow typically includes:
Virtual Screening:
Structure-based virtual screening of compound libraries (e.g., PubChem database)
Application of Tanimoto similarity thresholds (e.g., 80%) to identify structural analogs of known inhibitors
Molecular docking to predict binding modes and affinities
Filtering and Selection Criteria:
Drug-likeness properties assessment
Binding affinity evaluation
Docking score analysis
Selectivity profiling
Advanced Simulation Techniques:
All-atom molecular dynamics (MD) simulations (e.g., 200 ns duration)
Essential dynamics analysis
Principal component analysis (PCA)
Free energy landscapes (FELs)
One successful application of this approach identified promising FGFR2 inhibitors from 2336 compounds in the PubChem database, with a compound (PubChem CID:507883) demonstrating particularly favorable characteristics . The docking protocol validation through retrospective procedures, such as redocking co-crystallized ligands, ensures the reliability of the computational predictions.
Evaluating the specificity of FGFR2 inhibitors against other FGFR family members requires a multi-faceted approach:
Computational Methods:
Comparative molecular docking against all FGFR family members
Binding site analysis to identify unique structural features of FGFR2
Molecular dynamics simulations to assess inhibitor stability in different FGFR binding pockets
MM-GBSA or FEP calculations to compare binding energies across FGFR family
Biochemical Assays:
In vitro kinase assays with purified recombinant FGFR1-4 proteins
Determination of IC50 values for each family member
Calculation of selectivity indices (ratio of IC50 values)
Cellular Systems:
Cell lines with differential expression of FGFR family members
Phosphorylation assays of downstream targets
Cellular phenotypic assays (proliferation, migration, etc.)
Structural Biology:
X-ray crystallography or cryo-EM of inhibitor-bound FGFR2
Comparison with structures of other FGFR family members
By combining these approaches, researchers can develop comprehensive selectivity profiles and understand the molecular basis for any observed differences in inhibitor efficacy across the FGFR family, guiding further optimization efforts toward improved specificity.
FGFR2 mutations are a significant cause of craniosynostosis, a condition characterized by premature fusion of cranial sutures. In a comprehensive genomic screening of 259 patients with craniosynostosis (where mutations in other genes like FGFR1, FGFR3, and TWIST had been excluded), 85 independent FGFR2 mutations were detected . These mutations predominantly affect the FGFR2IIIc isoform, which is expressed in mesenchyme including craniofacial bone .
The mutations typically occur in specific hotspots of the FGFR2 gene and can be categorized based on their effects on protein function:
Ligand-binding domain mutations: These alter ligand binding specificity or affinity, potentially causing inappropriate activation of the receptor.
Transmembrane domain mutations: These can lead to constitutive dimerization and activation of the receptor independent of ligand binding.
Tyrosine kinase domain mutations: These may enhance catalytic activity or alter substrate specificity.
Splicing mutations: These can affect the balance between different FGFR2 isoforms, disrupting the tissue-specific expression patterns.
The functional consequences of these mutations include increased ligand binding affinity, ligand-independent activation, altered downstream signaling, and disrupted cross-talk with other signaling pathways, ultimately affecting the normal development of craniofacial structures.
Studying FGFR2 mutations in developmental contexts requires models that can recapitulate the complex cellular interactions and temporal dynamics of embryonic development. Several complementary approaches include:
Cell-Based Models:
Primary cranial osteoblasts or mesenchymal stem cells
Induced pluripotent stem cells (iPSCs) derived from patients with FGFR2 mutations
Differentiation protocols to generate relevant cell types (osteoblasts, chondrocytes)
Organoid Models:
Cranial suture organoids
Bone developmental organoids
Co-culture systems with multiple cell types
Animal Models:
Genetically engineered mouse models carrying specific FGFR2 mutations
Conditional knockout or knockin approaches for tissue-specific effects
CRISPR/Cas9-mediated introduction of mutations in developing embryos
Systems for Functional Analysis:
Live imaging of suture development in animal models
Micro-CT analysis of craniofacial bone development
Single-cell RNA sequencing to identify cell-specific effects
Phosphoproteomics to map altered signaling pathways
These models provide complementary insights, with cell-based systems offering detailed molecular analyses, organoids capturing 3D tissue architecture, and animal models revealing systemic developmental effects and phenotypic outcomes of FGFR2 mutations.
Advanced technologies for studying FGFR2 signaling dynamics in real-time include:
FRET-Based Biosensors:
Genetically encoded sensors for monitoring FGFR2 conformational changes
Sensors for downstream signaling events (MAPK, PI3K, PLCγ activation)
Single-molecule FRET to track individual receptor behavior
Live-Cell Imaging Techniques:
Lattice light-sheet microscopy for high-resolution 3D imaging
Total internal reflection fluorescence (TIRF) microscopy for membrane dynamics
Fluorescence recovery after photobleaching (FRAP) to measure receptor mobility
Optogenetic Approaches:
Light-controlled activation of FGFR2 signaling
Spatiotemporal control of receptor dimerization
Reversible modulation of specific signaling branches
Single-Cell Technologies:
Mass cytometry (CyTOF) for high-dimensional analysis of signaling states
Microfluidic platforms for single-cell signaling analysis
Live-cell RNA imaging to track transcriptional responses
These methodologies enable researchers to dissect the complex spatiotemporal dynamics of FGFR2 signaling with unprecedented resolution, revealing how signal transduction changes over time, varies between individual cells, and responds to different ligands or inhibitors.
Multi-omics approaches provide a comprehensive view of FGFR2 biology by integrating data from various molecular levels:
Genomics and Epigenomics:
Whole genome/exome sequencing to identify genetic alterations
DNA methylation analysis to understand epigenetic regulation
ATAC-seq for chromatin accessibility profiling
Transcriptomics:
RNA-seq for global expression changes
Single-cell RNA-seq for cell-type-specific responses
Spatial transcriptomics to map expression patterns in tissue context
Proteomics and Post-translational Modifications:
Global proteome analysis
Phosphoproteomics to map signaling networks
Ubiquitylation and SUMOylation profiling
Interactomics:
Affinity purification-mass spectrometry to identify protein interactions
Proximity labeling (BioID, APEX) for spatial interaction mapping
Cross-linking mass spectrometry for structural insights
Metabolomics:
Targeted and untargeted metabolite profiling
Stable isotope tracing for metabolic flux analysis
Integrative Computational Approaches:
Network analysis to connect multi-omics datasets
Machine learning for pattern recognition
Systems biology modeling of signaling dynamics
The integration of these approaches has revealed that DNA methylation is associated with FGFR2 expression in several cancers, and that FGFR2 alterations correlate with distinct patterns of gene expression and pathway activation . Such multi-omics studies provide a systems-level understanding of FGFR2 biology that cannot be achieved through single-omics approaches alone.
Current therapeutic strategies targeting FGFR2 in cancer treatment include:
Small Molecule Inhibitors:
Selective FGFR inhibitors (e.g., pemigatinib, infigratinib)
Multi-kinase inhibitors with FGFR activity (e.g., dovitinib, ponatinib)
Irreversible FGFR inhibitors in development
Clinical Applications:
FDA-approved pemigatinib for FGFR2 fusion-positive cholangiocarcinoma that has failed prior chemotherapy
Clinical trials showing objective remission rates of 14-35% and disease control rates of 75-83% for advanced cholangiocarcinomas with FGFR2 fusions
Progression-free survival of 5.8-6.9 months in targeted therapy trials
Combination Approaches:
FGFR inhibitors with immune checkpoint inhibitors
Combinations with cytotoxic chemotherapy
Strategies to overcome resistance mechanisms
Emerging Approaches:
Antibody-drug conjugates targeting FGFR2
Proteolysis targeting chimeras (PROTACs) for FGFR2 degradation
Allosteric modulators of FGFR2 function
These strategies are being evaluated in multiple clinical trials, with particular focus on tumors with specific FGFR2 alterations. The success of pemigatinib in cholangiocarcinoma has established FGFR2-targeted therapy as a viable approach for precision oncology, with ongoing efforts to expand its applications to other cancer types with relevant FGFR2 alterations.
Optimizing patient selection for FGFR2-targeted therapies requires a comprehensive approach:
Molecular Testing Strategies:
Comparison of detection methods shows that FISH and NGS provide concordant results for FGFR2 rearrangements (kappa value=0.696, p<0.01), while IHC shows discordant results
NGS offers comprehensive detection of various FGFR2 alterations (mutations, fusions, amplifications)
Liquid biopsy approaches for non-invasive monitoring
Clinicopathological Features:
Concomitant Genomic Alterations:
Biomarker Development:
Downstream signaling activation markers
Expression signatures predictive of response
Resistance-associated alterations
By integrating these approaches, researchers can develop evidence-based strategies for identifying patients most likely to benefit from FGFR2-targeted therapies, improving clinical outcomes while minimizing unnecessary treatments in patients unlikely to respond.
Emerging areas in FGFR2 research include:
Structural Biology and Biophysics:
Cryo-EM studies of full-length FGFR2 in different activation states
Investigation of FGFR2 complexes with co-receptors and modulators
Single-molecule studies of receptor dynamics and clustering
Systems Biology Approaches:
Quantitative modeling of FGFR2 signaling networks
Integration of multi-omics data to build predictive models
Network-based approaches to understand context-dependent signaling
Developmental and Stem Cell Biology:
Role of FGFR2 in tissue-specific stem cell maintenance and differentiation
Single-cell analysis of FGFR2 function during embryonic development
Organoid models to study FGFR2 in tissue morphogenesis
Immune System Interactions:
Cross-talk between FGFR2 signaling and immune cell function
Implications for cancer immunotherapy combinations
Role in inflammatory conditions and wound healing
Novel Therapeutic Approaches:
Allosteric modulators of FGFR2 activity
Targeted protein degradation strategies
RNA-based therapeutics for isoform-specific modulation
These emerging areas promise to deepen our understanding of FGFR2 biology and open new avenues for therapeutic intervention in FGFR2-associated diseases.
Technological advances are poised to transform FGFR2-targeted drug development in several key areas:
Artificial Intelligence and Machine Learning:
AI-driven drug design targeting FGFR2-specific binding pockets
Predictive modeling of drug resistance mechanisms
Virtual screening of billions of compounds with improved accuracy
Advanced Structural Biology:
AlphaFold and similar tools for more accurate protein structure prediction
Cryo-EM for visualization of FGFR2 in different conformational states
Fragment-based drug discovery approaches
High-throughput Functional Screening:
CRISPR-based genetic screens for resistance mechanisms
Phenotypic screening in 3D organoid models
Patient-derived models for personalized drug testing
Precision Medicine Implementation:
Digital pathology with AI-assisted interpretation
Integration of genomic, transcriptomic, and proteomic biomarkers
Real-world evidence collection through digital health platforms
Novel Therapeutic Modalities:
Bifunctional degraders (PROTACs) targeting FGFR2
mRNA-based therapies for transient FGFR2 modulation
Gene editing approaches for permanent correction of pathogenic FGFR2 mutations
These technological advances will enable more rapid development of highly selective FGFR2 inhibitors, better prediction of clinical responses, and more effective strategies to overcome resistance mechanisms, ultimately improving outcomes for patients with FGFR2-driven diseases.
FGFR2 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 of FGFR2 interacts with FGFs, initiating a cascade of downstream signals that influence mitogenesis and differentiation . FGFR2 is known to bind to acidic, basic, and keratinocyte growth factors, depending on the isoform .
The recombinant form of FGFR2, tagged with a His (histidine) tag, is produced using various expression systems, such as baculovirus . The His tag facilitates the purification and detection of the protein. The recombinant FGFR2 is often used in research to study its role in cellular processes and its involvement in various diseases .
Mutations in the FGFR2 gene are associated with several genetic disorders, including Crouzon syndrome, Pfeiffer syndrome, Apert syndrome, Jackson-Weiss syndrome, Beare-Stevenson cutis gyrata syndrome, Saethre-Chotzen syndrome, and syndromic craniosynostosis . These mutations can lead to abnormal receptor function, resulting in altered cellular signaling and developmental abnormalities .
Recombinant FGFR2 with a His tag is widely used in research to investigate its role in various physiological and pathological processes. It is particularly valuable in studying cancer, as FGFR2 is implicated in several types of cancer, including lung and breast cancers . Researchers use recombinant FGFR2 to explore its involvement in angiogenesis, mitogenesis, osteogenesis, myogenesis, carcinogenesis, and tissue repair after injury .