FGFR2 Human

Fibroblast Growth Factor Receptor 2 Fc Chimera Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to FGFR2 Human

FGFR2 (Fibroblast Growth Factor Receptor 2) is a transmembrane receptor tyrosine kinase encoded by the FGFR2 gene on chromosome 10 in humans. It plays critical roles in embryonic development, tissue repair, and angiogenesis. FGFR2 signals via binding to fibroblast growth factors (FGFs), triggering downstream pathways like MAPK, PI3K/AKT, and PLCγ, which regulate cell proliferation, differentiation, and survival .

Molecular Structure and Isoforms

FGFR2 consists of an extracellular domain (three immunoglobulin-like domains), a transmembrane segment, and an intracellular tyrosine kinase domain. Alternative splicing generates two major isoforms:

IsoformTissue ExpressionLigand Binding
FGFR2IIIbEpithelial tissues (e.g., skin, organs)FGF-1, -3, -7, -10, -22
FGFR2IIIcMesenchymal tissues (e.g., bone, craniofacial regions)FGF-1, -2, -4, -6, -8, -9, -17, -18

These isoforms differ in ligand specificity, influencing tissue-specific signaling .

Biological Functions

FGFR2 is essential for:

  • Embryonic Development: Craniofacial bone formation, limb patterning, and Wolffian duct maintenance .

  • Tissue Repair: Wound healing and angiogenesis via endothelial cell proliferation .

  • Cell Signaling: Activation of MAPK/ERK and PI3K/AKT pathways to regulate cell survival and migration .

Clinical Significance in Genetic Disorders

Craniosynostosis Syndromes
Gain-of-function FGFR2 mutations disrupt normal osteoblast differentiation, causing premature skull suture fusion. Key syndromes include:

SyndromeAssociated MutationMechanism
Apert SyndromeSer252Trp, Pro253Arg (exon 7)Loss of ligand specificity, constitutive activation
Crouzon/PfeifferExon 10 mutations (Ala391Glu)Constitutive receptor dimerization

Other Disorders

  • LADD Syndrome: Loss-of-function mutations impair gland formation and skeletal development .

  • Cleft Lip/Palate: Identified as a susceptibility locus via chromosomal linkage studies .

Role in Oncology

FGFR2 is implicated in multiple cancers through amplifications, mutations, and fusions.

Cancer-Specific Alterations

Cancer TypeAlterationFrequencyImpact
Gastric AdenocarcinomaAmplification, IIIb overexpression5–10% AKT/mTOR pathway activation, drug resistance
CholangiocarcinomaFusions (e.g., FGFR2-BICC1)~15% Constitutive kinase activity, gemcitabine resistance
Endometrial/UterineMissense mutations (S252W)12% Autocrine FGF signaling, PI3K/AKT activation

Therapeutic Vulnerabilities

  • FGFR2 Amplification: Linked to poor prognosis in gastric cancer but responsive to inhibitors like AZD4547 and infigratinib .

  • Fusions: FGFR2-BICC1 and FGFR2-TACC3 drive oncogenic signaling; targeted by PROTACs (e.g., LC-MB12) .

Small-Molecule Inhibitors

DrugTargetClinical PhaseKey Findings
AZD4547FGFR1-3 tyrosine kinasePhase II17.9% ORR in FGFR2-amplified gastric cancer
InfigratinibFGFR1-3Phase II14% ORR in FGFR2-altered cholangiocarcinoma
RLY-4008FGFR2 (selective)Phase II52% ORR in FGFRi-naïve cholangiocarcinoma

Antibody-Based Therapies

DrugMechanismClinical PhaseKey Findings
BemarituzumabAnti-FGFR2IIIb mAbPhase III53% ORR when combined with FOLFOX in gastric cancer
FPA144Anti-FGFR2IIIb mAbPhase IBlocks FGF7/10/22 binding, clinical trials ongoing

Survival Outcomes in FGFR2-Targeted Trials

CancerTreatmentORRMedian OSReference
Gastric (FGFR2b+)Bemarituzumab + FOLFOX53%19.2 mo
CholangiocarcinomaRLY-400852%8.2 mo
EndometrialAZD454714%5.6 mo

Mutational Hotspots in FGFR2

DomainCancer TypeMutationFunction
Exon 7Apert SyndromeSer252TrpConstitutive activation
Exon 10Crouzon SyndromeAla391GluLigand-independent dimerization
Exon 18CholangiocarcinomaTruncationsLoss of negative regulatory domains

Product Specs

Introduction
The Fibroblast Growth Factor (FGF) family consists of at least 18 structurally similar proteins with diverse roles in physiological and pathological cellular processes. These processes include cell growth, differentiation, angiogenesis, wound healing, and tumorigenesis. FGFs exert their biological activities by binding to and activating type I transmembrane tyrosine kinase receptors known as FGF receptors (FGFRs). Activation leads to receptor dimerization and autophosphorylation. Currently, four distinct genes encoding highly similar FGFRs (FGFR-1 to -4) have been identified. Alternative splicing of mRNAs from FGFR-1 to -3 genes generates multiple receptor isoforms. A common splicing event affects FGFR-1 and -2, resulting in two isoforms: the alpha isoform, containing all three immunoglobulin-like domains (IgI, IgII, and IgIII), and the beta isoform, containing only IgII and IgIII. In contrast, FGFR-3 and FGFR-4 have only been observed as the alpha isoform. Further splicing events for FGFR-1 to -3 occur in the C-terminal half of the IgIII domain. These events involve two mutually exclusive alternative exons, leading to FGF receptors with distinct IgIII domains (IIIb and IIIc). Additionally, a secreted FGF-binding protein called the IIIa isoform has been described for FGFR-1. This isoform consists of the N-terminal half of the IgIII domain and some intronic sequences. Mutations in FGFR-1 to -3 have been linked to birth defects characterized by craniosynostosis.
Description
Soluble FGFR-2a (IIIc) Fc Chimera Human Recombinant is a heterodimeric, glycosylated polypeptide chain with a molecular weight of 170 kDa. It comprises 602 amino acids and includes a Xa cleavage site fused with the Fc region of human IgG1. The protein is produced in a baculovirus expression system and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
CD332 was lyophilized from a sterile solution with a concentration of 1 mg/ml. The solution does not contain any additives.
Solubility
To reconstitute the lyophilized FGFR-2, it is recommended to dissolve it in sterile PBS at a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions.
Stability
Lyophilized FGFR2A remains stable at room temperature for up to 3 weeks. However, it is recommended to store it desiccated below -18°C for long-term storage. Once reconstituted, FGFR2 should be stored at 4°C for 2-7 days. For future use, it can be stored below -18°C. To ensure long-term stability during storage, it is advisable to add a carrier protein like HSA or BSA at a concentration of 0.1%. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is determined by SDS-PAGE analysis and is greater than 90.0%.
Biological Activity
The biological activity of the protein is assessed based on its ability to inhibit human FGF-2-dependent proliferation in HUVE cells. The ED50 for this effect is typically in the range of 15 - 30 ng/ml.
Synonyms
Keratinocyte growth factor receptor 2, CD332, FGFR2.
Source
Insect Cells.
Amino Acid Sequence

RPSFSLVEDTTLEPEEPPTKYQISQPEVYVAAPGESLEVRCLLKDAAVISWT KDGVHLGPNNRTVLIGEYLQIKGATPRDSGLYACTASRTVDSETWYFMVNVT DAISSGDDEDDTDGAEDFVSENSNNKRAPYWTNTEKMEKRLHAVPAANTVKF RCPAGGNPMPTMRWLKNGKEFKQEHRIGGYKVRNQHWSLIMESVVPSDKGNY TCVVENEYGSINHTYHLDVVERSPHRPILQAGLPANASTVVGGDVEFVCKVY SDAQPHIQWIKHVEKNGSKYGPDGLPYLKVLKAAGVNTTDKEIEVLYIRNVT FEDAGEYTCLAGNSIGISFHSAWLTVLPAPGREKEITASPDYLEDPRRASIE GRGDPEEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSL TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK

10x Buffer With Mgso4
default

Q&A

What is FGFR2 and what are its primary functions in human biology?

FGFR2 is a member of the fibroblast growth factor receptor family, characterized by an extracellular portion composed of three immunoglobulin-like domains, a hydrophobic membrane-spanning segment, and a cytoplasmic tyrosine kinase domain. This transmembrane receptor spans the cell membrane with one end remaining inside the cell while the other projects from the outer surface, allowing it to interact with specific growth factors and transduce signals across the membrane . The protein plays crucial roles in multiple biological processes including cell proliferation, differentiation, migration, and survival . During embryonic development, FGFR2 is particularly important for bone formation, directing immature cells to become bone cells and form structures of the head, hands, feet, and other tissues . Additionally, FGFR2 remains active throughout life, participating in processes such as angiogenesis (formation of blood vessels), wound healing, and bone remodeling, where old bone is broken down and replaced by new bone .

How is the FGFR2 gene structured and what isoforms exist?

The FGFR2 gene in humans is a protein-coding gene located in the genome with multiple exons that can undergo alternative splicing to generate different isoforms of the protein . Several slightly different versions (isoforms) of the FGFR2 protein exist, and these isoforms demonstrate tissue-specific distribution patterns that can change throughout growth and development . The alternative splicing particularly affects the third immunoglobulin-like domain, resulting in receptor variants with different ligand-binding specificities and affinities . The FGFR2 gene contains regions that encode the extracellular immunoglobulin-like domains, the transmembrane domain, and the intracellular tyrosine kinase domain . These different isoforms are functionally significant as they can respond to different FGF ligands and are expressed in tissue-specific patterns, allowing for precise regulation of FGF signaling in different cellular contexts and developmental stages .

What signaling pathways are activated downstream of FGFR2?

When fibroblast growth factors bind to FGFR2, the receptor undergoes dimerization and autophosphorylation of its tyrosine residues in the intracellular domain, triggering a cascade of downstream signaling events . The activated FGFR2 initiates several major signaling pathways, including the RAS-MAPK (mitogen-activated protein kinase) pathway, which is critical for cell proliferation and differentiation . Additionally, FGFR2 activates the PI3K-AKT pathway, which promotes cell survival and metabolism, as well as the PLCγ (phospholipase C gamma) pathway, which regulates calcium signaling and protein kinase C activation . The STAT (signal transducer and activator of transcription) pathway may also be activated, contributing to gene expression regulation . These signaling cascades ultimately lead to changes in gene expression profiles that instruct cells to undergo specific changes, such as proliferation, differentiation, migration, or survival, depending on the cellular context and the specific FGF ligand involved .

What role does FGFR2 play in embryonic development?

FGFR2 serves as a key regulator during embryonic development, functioning as a critical signaling molecule that orchestrates proper tissue and organ formation . During early development, FGFR2 signaling directs the commitment of certain progenitor cells to become bone-forming cells, playing an essential role in craniofacial development and the formation of limbs . The receptor mediates epithelial-mesenchymal interactions that are fundamental to organogenesis, with the IIIb isoform being particularly important for mesenchymal-epithelial signaling during mouse organogenesis, suggesting a similar critical role in human development . FGFR2 signaling also regulates cellular proliferation and differentiation in multiple developing tissues, including the brain, skin, lungs, and other organs . Disruptions in FGFR2 signaling during embryonic development can lead to severe developmental abnormalities, particularly affecting bone formation and growth, which manifests in various congenital disorders associated with FGFR2 mutations .

What human disorders are associated with FGFR2 mutations?

Mutations in the FGFR2 gene are associated with numerous autosomal dominantly inherited disorders, primarily affecting skeletal and craniofacial development . Craniosynostosis syndromes, characterized by premature fusion of skull bones, represent a significant category of FGFR2-related disorders and include Crouzon syndrome (characterized by cranial and facial abnormalities), Pfeiffer syndrome (featuring craniosynostosis with broad thumbs and great toes), and Apert syndrome (involving craniosynostosis with syndactyly of hands and feet) . Other FGFR2-associated conditions include Jackson-Weiss syndrome, Beare-Stevenson cutis gyrata syndrome, and Saethre-Chotzen syndrome, each with distinctive clinical presentations but sharing underlying mechanisms of aberrant FGFR2 signaling . Recently, mosaic FGFR2 variants have been identified in patients with neurocutaneous syndromes, expanding the phenotypic spectrum to include cutaneous nevi, ocular malformations, macrocephaly, and developmental delay . Additionally, somatic mutations in FGFR2 have been implicated in various cancers, including gastric cancer, highlighting the oncogenic potential of dysregulated FGFR2 signaling .

How do different types of FGFR2 mutations affect protein function and disease phenotypes?

FGFR2 mutations can affect protein function through various mechanisms, resulting in distinct disease phenotypes depending on the location and nature of the mutation . Gain-of-function mutations, which cause constitutive activation of the receptor independent of ligand binding, are commonly associated with craniosynostosis syndromes and typically affect the extracellular domain or the region regulating tyrosine kinase activity . For example, mutations in the tyrosine kinase domain, such as the p.(Asn549Lys) variant, can result in increased tyrosine phosphorylation and constitutive receptor activation, leading to aberrant downstream signaling . Mutations affecting the ligand-binding specificity domain can alter the receptor's interaction with different FGF ligands, potentially resulting in inappropriate activation or inhibition of specific signaling pathways . Genotype-phenotype correlations have been observed, with specific mutations consistently associated with certain syndromes; for instance, particular mutations in the region affecting ligand binding and specificity are associated with Apert syndrome, while others affecting tyrosine kinase activity are linked to Pfeiffer syndrome . The varied clinical manifestations reflect the complex role of FGFR2 in multiple developmental processes and tissues, with different mutations disrupting distinct aspects of receptor function and downstream signaling .

What are the recommended methods for studying FGFR2 expression in human tissues?

Researchers investigating FGFR2 expression in human tissues should employ a multi-modal approach combining molecular, cellular, and imaging techniques to comprehensively characterize expression patterns . Quantitative reverse transcription PCR (qRT-PCR) provides precise measurement of FGFR2 mRNA levels, allowing researchers to quantify expression changes across different tissues or experimental conditions while distinguishing between different isoforms through careful primer design . Immunohistochemistry and immunofluorescence represent valuable approaches for visualizing FGFR2 protein distribution within tissue sections, enabling assessment of spatial expression patterns, subcellular localization, and co-expression with other proteins of interest . Western blotting serves as a standard method for detecting and quantifying FGFR2 protein levels in tissue or cell lysates, with phospho-specific antibodies additionally providing information about receptor activation status . For higher resolution analysis, RNA in situ hybridization can reveal cell-specific expression patterns within complex tissues, while single-cell RNA sequencing offers unprecedented insights into cellular heterogeneity of FGFR2 expression across different cell populations . When studying human tissues, researchers should carefully validate antibodies and probes for specificity, as the high sequence homology between FGFR family members can lead to cross-reactivity issues .

What experimental systems are most appropriate for functional analysis of FGFR2 mutations?

Multiple experimental systems can be employed for functional analysis of FGFR2 mutations, each offering distinct advantages for investigating different aspects of receptor biology and pathology . Cell-based systems represent a fundamental approach, with transient transfection of wild-type or mutant FGFR2 constructs into appropriate cell lines (such as HEK293T cells) enabling analysis of protein expression, stability, phosphorylation status, and downstream signaling pathway activation . For more physiologically relevant studies, patient-derived primary cells carrying FGFR2 mutations or CRISPR/Cas9-edited cell lines provide valuable models that retain the endogenous genomic context and cellular environment . Animal models, particularly genetically engineered mice carrying specific FGFR2 mutations, offer powerful systems for studying the developmental consequences and tissue-specific effects of FGFR2 variants in an intact organism, with mouse models successfully recapitulating many features of human FGFR2-associated disorders . Emerging three-dimensional culture systems, including organoids derived from patient cells or engineered to harbor specific FGFR2 mutations, bridge the gap between simple cell culture and complex animal models, allowing for analysis of FGFR2 function in a structured tissue-like environment . Researchers should select experimental systems based on their specific research questions, considering factors such as physiological relevance, technical feasibility, and the particular aspect of FGFR2 biology being investigated .

What techniques are available for assessing FGFR2 phosphorylation and activation status?

Several sophisticated techniques enable researchers to assess FGFR2 phosphorylation and activation status with varying degrees of sensitivity, specificity, and contextual information . Immunoprecipitation followed by western blotting with phosphotyrosine-specific antibodies represents a standard approach for detecting FGFR2 activation, as demonstrated in studies characterizing the functional consequences of the p.(Asn549Lys) variant . This technique involves isolating FGFR2 protein (often using epitope tags such as FLAG) and subsequently probing for phosphorylated tyrosine residues, allowing for quantitative comparison between wild-type and mutant receptors or between different treatment conditions . Phospho-specific antibodies targeting particular phosphorylation sites within FGFR2 can be used directly in western blotting, immunohistochemistry, or flow cytometry to assess activation of specific regulatory sites without the need for immunoprecipitation . For higher throughput analysis, phospho-protein arrays or mass spectrometry-based phosphoproteomics enable comprehensive profiling of phosphorylation events across multiple sites and downstream effectors simultaneously . Live-cell imaging approaches using fluorescent biosensors can provide real-time visualization of FGFR2 activation dynamics in living cells, offering insights into the spatial and temporal aspects of receptor signaling that are not captured by biochemical methods . When interpreting results, researchers should consider that different mutations may affect distinct phosphorylation sites and downstream pathways, necessitating a comprehensive approach to fully characterize receptor activation status .

How do different FGF ligands interact with FGFR2 and what determines binding specificity?

The interaction between FGF ligands and FGFR2 is governed by complex structural and molecular determinants that establish binding specificity and regulate downstream signaling outcomes . FGFR2 demonstrates variable affinity for different FGF ligands, with binding specificity largely determined by the third immunoglobulin-like domain (IgIII domain) in the extracellular portion of the receptor, which exists in alternatively spliced isoforms (IIIb and IIIc) with distinct ligand binding properties . The FGFR2-IIIb isoform preferentially binds FGF7, FGF10, and FGF22, while the FGFR2-IIIc isoform exhibits higher affinity for FGF2, FGF4, FGF6, FGF9, and FGF20, enabling tissue-specific responses to different FGF ligands . Heparan sulfate proteoglycans (HSPGs) serve as essential cofactors in FGF-FGFR2 interactions, stabilizing the ligand-receptor complex and influencing binding specificity through the formation of ternary complexes . The binding of FGF ligands to FGFR2 induces receptor dimerization, bringing the intracellular kinase domains into proximity and enabling transphosphorylation, with different FGF ligands potentially inducing distinct conformational changes that may influence downstream signaling outcomes . Understanding these complex interaction dynamics is crucial for interpreting how mutations in FGFR2 affect ligand binding and for developing therapeutic strategies targeting specific FGF-FGFR2 interactions .

What are the current challenges in reconciling conflicting data about FGFR2 function in different tissues?

Researchers studying FGFR2 face several challenges in reconciling apparently conflicting data about receptor function across different tissues and biological contexts . The tissue-specific expression patterns of FGFR2 isoforms represent a significant source of complexity, as the predominant isoform can vary dramatically between epithelial (primarily expressing FGFR2-IIIb) and mesenchymal (primarily expressing FGFR2-IIIc) tissues, leading to divergent functional consequences of FGFR2 signaling in different cellular environments . The dynamic regulation of FGFR2 during development and in pathological states further complicates interpretation, as receptor function can change during developmental transitions, tissue repair, or disease progression, resulting in seemingly contradictory observations depending on the specific temporal context examined . The complex interplay between FGFR2 and other signaling pathways contributes additional layers of complexity, with cross-talk between FGF signaling and other pathways such as WNT, Hedgehog, and Notch potentially modulating FGFR2 function in a context-dependent manner . Methodological differences across studies, including variations in experimental systems, analytical techniques, and specific cell types or tissues examined, can also lead to apparent discrepancies that reflect technical factors rather than true biological differences . Addressing these challenges requires integrated approaches that consider the specific isoforms, developmental stages, and cellular contexts being studied, emphasizing the importance of precisely defining experimental conditions when comparing results across different studies .

How does FGFR2 signaling interact with other major developmental pathways?

FGFR2 signaling exhibits complex interactions with other major developmental pathways, creating an integrated network that coordinates multiple aspects of embryonic development and tissue homeostasis . The RAS-MAPK pathway represents a critical node of integration, as FGFR2 activation stimulates this signaling cascade, which is also regulated by other growth factor receptors and developmental signals, allowing for coordinated responses to multiple inputs during development . This explains why disorders caused by mutations in FGFR2 share phenotypic features with RASopathies, as demonstrated by the initial clinical suspicion of Schimmelpenning–Feuerstein–Mims syndrome (a RASopathy) in a patient ultimately diagnosed with an FGFR2 mosaic variant . FGFR2 signaling also intersects with the WNT pathway, another fundamental developmental signaling system, with evidence suggesting reciprocal regulation and convergence on common downstream targets involved in cell fate decisions and morphogenesis . Cross-talk with the Notch signaling pathway influences cell differentiation and boundary formation during development, while interactions with the Hedgehog pathway affect patterning and skeletal development . These pathway interactions exhibit tissue-specific and developmental stage-specific characteristics, contributing to the precise spatial and temporal control of cellular responses during development . Understanding these complex interactions is essential for interpreting the pleiotropic effects of FGFR2 mutations and for developing therapeutic strategies that consider the broader signaling network rather than FGFR2 in isolation .

What genetic screening approaches are most effective for identifying FGFR2 variants in clinical samples?

Multiple genetic screening approaches can be employed for identifying FGFR2 variants in clinical samples, with the selection of method depending on the clinical context, suspected variant type, and available resources . Targeted sequencing of the FGFR2 gene represents a focused approach for patients with clinical features suggestive of FGFR2-related disorders, such as craniosynostosis syndromes, allowing for deep coverage of coding regions and splice sites to detect single nucleotide variants and small insertions or deletions . Whole exome sequencing (WES) offers a more comprehensive approach that captures variants across all protein-coding regions, making it particularly valuable for cases with atypical presentations or when differential diagnoses span multiple genes, as illustrated by the successful identification of a mosaic FGFR2 variant in a patient initially suspected of having a RASopathy . For detecting low-level mosaicism, deep sequencing with high coverage (>100×) is essential, as demonstrated in cases where pathogenic variants were present in only a fraction of cells and might be missed by standard-depth sequencing . Analyzing multiple tissue types can also be crucial for mosaic variants, as exemplified by a case where an FGFR2 variant was detected in DNA from affected skin tissue but not in blood, highlighting the importance of appropriate tissue sampling . When interpreting genetic findings, functional characterization of novel variants through experimental approaches is valuable for establishing pathogenicity, particularly for variants of uncertain significance .

What are the recommended protocols for functional characterization of novel FGFR2 variants?

Comprehensive functional characterization of novel FGFR2 variants requires a multi-faceted approach that evaluates different aspects of receptor biology and signaling . Expression analysis represents an initial step, involving transfection of wild-type and mutant FGFR2 constructs into appropriate cell lines followed by western blot analysis to compare protein expression levels and stability, as demonstrated in studies of the p.(Asn549Lys) variant where similar expression levels indicated that the variant did not affect protein stability . Phosphorylation assays are crucial for evaluating tyrosine kinase activity, typically involving immunoprecipitation of FGFR2 followed by western blotting with phosphotyrosine-specific antibodies to assess baseline and ligand-induced phosphorylation, with results from such assays revealing that the p.(Asn549Lys) variant caused strongly increased tyrosine phosphorylation compared to wild-type FGFR2 . Downstream signaling analysis should examine activation of pathways such as MAPK, PI3K/AKT, and PLCγ through western blotting for phosphorylated forms of key signaling proteins, providing insights into how the variant affects signal transduction beyond the receptor itself . Structural modeling complements biochemical approaches by predicting how amino acid substitutions might affect protein conformation and function, as illustrated by protein structure simulations of the p.(Asn549Lys) variant that suggested constitutive receptor activation . For comprehensive characterization, these approaches should be integrated with cell-based functional assays examining phenotypic outcomes such as proliferation, differentiation, or migration relevant to the biological roles of FGFR2 .

What model systems best recapitulate human FGFR2-related disorders for translational research?

Several model systems can effectively recapitulate aspects of human FGFR2-related disorders, each offering distinct advantages for translational research depending on the specific research questions and applications . Genetically engineered mouse models carrying specific FGFR2 mutations have successfully reproduced many phenotypic features of human craniosynostosis syndromes and other FGFR2-associated disorders, providing valuable platforms for studying disease pathogenesis, developmental consequences, and potential therapeutic interventions in an intact mammalian system . The high conservation of FGFR2 between mice and humans, including shared isoforms and expression profiles, contributes to the translational relevance of mouse models, with induced mutations in mice resulting in phenotypes highly similar to those observed in humans with comparable mutations . Patient-derived induced pluripotent stem cells (iPSCs) offer alternative models that maintain the exact genetic background of affected individuals, enabling differentiation into relevant cell types such as osteoblasts, chondrocytes, or neural crest cells to study tissue-specific manifestations of FGFR2 mutations . Three-dimensional organoid cultures derived from patient cells or engineered with specific FGFR2 mutations represent emerging systems that can capture complex tissue architecture and cellular interactions, potentially bridging the gap between simpler in vitro models and complex in vivo systems . When selecting model systems, researchers should consider factors such as phenotypic fidelity, genetic background, developmental stage, and specific aspects of FGFR2 biology being investigated, recognizing that complementary use of multiple models may provide the most comprehensive insights for translational applications .

What are emerging therapeutic approaches targeting FGFR2 signaling in developmental disorders and cancer?

Several innovative therapeutic approaches targeting FGFR2 signaling are emerging, with applications for both developmental disorders and cancer driven by dysregulated FGFR2 activity . Small molecule tyrosine kinase inhibitors (TKIs) that target the ATP-binding pocket of the FGFR2 kinase domain represent a major therapeutic strategy, with both pan-FGFR inhibitors and more selective FGFR2-specific inhibitors in development for cancer treatment and potentially applicable to developmental disorders caused by FGFR2 gain-of-function mutations . Monoclonal antibodies directed against FGFR2 offer an alternative approach, blocking ligand binding and receptor activation with potentially greater specificity than TKIs, although their efficacy for developmental disorders would depend on timing of administration and ability to reach target tissues . RNA-based therapeutics, including antisense oligonucleotides and RNA interference approaches, could selectively reduce expression of mutant FGFR2 alleles or modulate alternative splicing of FGFR2 isoforms, potentially offering precision approaches for specific mutations . For developmental disorders, the critical window for intervention presents a significant challenge, necessitating prenatal diagnosis and potentially in utero treatment approaches for conditions that manifest during embryonic development . The emerging understanding of mosaic FGFR2 variants in neurocutaneous conditions suggests potential for postnatal interventions in some cases, as these disorders may involve ongoing signaling dysregulation beyond embryonic development . Future therapeutic development will likely benefit from advances in delivery technologies, including targeted nanoparticles and protein engineering approaches to improve tissue-specific drug delivery .

How might single-cell technologies advance our understanding of FGFR2 biology?

Single-cell technologies offer unprecedented opportunities to dissect the complex roles of FGFR2 in development, homeostasis, and disease at cellular resolution, potentially resolving longstanding questions about tissue-specific functions and heterogeneous responses . Single-cell RNA sequencing (scRNA-seq) enables comprehensive profiling of FGFR2 expression and its downstream targets across thousands of individual cells, revealing cell type-specific expression patterns, identifying previously unrecognized FGFR2-expressing cell populations, and characterizing cellular heterogeneity in response to FGFR2 signaling or in FGFR2-related disorders . Spatial transcriptomics methods that preserve tissue architecture while profiling gene expression can map FGFR2 signaling activities within complex tissues, potentially illuminating how FGFR2 functions at tissue boundaries and in specific microenvironmental contexts . Single-cell ATAC-seq (Assay for Transposase-Accessible Chromatin) can reveal how FGFR2 signaling alters chromatin accessibility and regulatory element usage in different cell types, providing insights into epigenetic mechanisms underlying cell type-specific responses to FGFR2 activation . For developmental disorders, these technologies could characterize the cellular consequences of FGFR2 mutations with unprecedented resolution, potentially identifying specific cell populations that drive pathogenesis and revealing compensation mechanisms in unaffected cells . In mosaic conditions, single-cell approaches offer particular value for distinguishing between cells carrying mutant FGFR2 and wild-type cells within the same tissue, enabling precise characterization of cell-autonomous and non-cell-autonomous effects of FGFR2 mutations .

What are the key unanswered questions about FGFR2 biology that require further investigation?

Despite decades of research, several fundamental questions about FGFR2 biology remain unanswered, representing important areas for future investigation with implications for both basic science and clinical applications . The precise mechanisms determining tissue-specific outcomes of FGFR2 signaling remain incompletely understood, including how the same mutation can affect different tissues with varying severity, and what factors modulate these tissue-specific responses beyond the expression patterns of receptor isoforms . The temporal dynamics of FGFR2 signaling during development and in pathological states requires further elucidation, particularly regarding how signaling requirements change across developmental stages and how these dynamics influence disease progression and potential therapeutic windows . The contribution of FGFR2 to cellular plasticity and fate decisions in adult tissues remains an emerging area of investigation, with implications for tissue homeostasis, regeneration, and carcinogenesis . The molecular basis for phenotypic variability among individuals carrying identical FGFR2 mutations represents another key question, including the identification of genetic and environmental modifiers that influence disease penetrance and expressivity . From a translational perspective, developing effective therapeutic strategies that can correct aberrant FGFR2 signaling without disrupting essential physiological functions remains a significant challenge, particularly for developmental disorders where the timing of intervention is critical . Addressing these questions will require integrated approaches combining advanced genetic technologies, high-resolution imaging, sophisticated model systems, and computational methods to fully unravel the complex biology of this multifunctional receptor .

Product Science Overview

Structure and Isoforms

FGFR2 exists in multiple isoforms due to alternative splicing of its mRNA. The two main isoforms are FGFR2 alpha (IIIb) and FGFR2 beta (IIIc), which differ in their ligand-binding specificities and tissue distribution . The alpha isoform contains all three immunoglobulin (Ig) domains, while the beta isoform contains only IgII and IgIII domains .

FGFR2 Fc Chimera

The FGFR2 Fc Chimera is a recombinant fusion protein that combines the extracellular domain of FGFR2 with the Fc region of human IgG1 . This fusion enhances the stability and solubility of the receptor, making it suitable for various research applications. The Fc region also allows for easy purification using protein A or G affinity chromatography .

Production and Purification

The FGFR2 Fc Chimera is typically produced in insect cells using a baculovirus expression system . The recombinant protein is then purified using proprietary chromatographic techniques to achieve high purity levels (>90%) as determined by SDS-PAGE . The lyophilized protein is stable at room temperature for several weeks but should be stored at -18°C for long-term storage .

Biological Activity

The biological activity of the FGFR2 Fc Chimera is determined by its ability to inhibit FGF-dependent proliferation of various cell types . For example, it has been shown to inhibit the proliferation of NR6R-3T3 mouse fibroblast cells and human umbilical vein endothelial (HUVE) cells . The effective dose (ED50) for these effects is typically in the range of 1-30 ng/mL .

Applications

The FGFR2 Fc Chimera is widely used in research to study the mechanisms of FGF signaling and its role in various physiological and pathological processes . It is also used in drug discovery and development to screen for potential inhibitors of FGFR2 signaling .

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.