FGFR3 Human

Fibroblast Growth Factor Receptor 3 Fc Chimera Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Functional Domains

FGFR3 is part of the FGFR family, sharing structural homology with FGFR1, FGFR2, and FGFR4. Its structure includes:

  • Extracellular Domain: Three immunoglobulin-like domains (D1–D3) for ligand binding.

  • Transmembrane Segment: A hydrophobic helix anchoring the receptor to the cell membrane.

  • Intracellular Domain: A tyrosine kinase domain responsible for signal transduction .

Alternative splicing generates isoforms with distinct tissue-specific expression (e.g., cartilage, brain, intestine) .

DomainFunctionKey Ligands
ExtracellularBinds FGF ligands (e.g., FGF1, FGF2, FGF18)Acidic and basic FGFs
TransmembraneMembrane localizationN/A
IntracellularPhosphorylates downstream targets (e.g., MAPK, PI3K)N/A

Genetic and Molecular Mechanisms

FGFR3 regulates bone growth by inhibiting chondrocyte proliferation and promoting differentiation. Gain-of-function mutations disrupt this balance, leading to skeletal abnormalities.

Key Mutations and Their Effects

MutationDiseaseMechanismReference
G380R (Gly380Arg)AchondroplasiaStabilizes receptor dimerization; ligand-independent activation
N540K (Asn540Lys)HypochondroplasiaReduced ligand affinity; partial receptor activation
K650M (Lys650Met)SADDAN syndromeConstitutive activation; severe skeletal and neurological defects
Y373C (Tyr373Cys)Thanatophoric dysplasiaCysteine-rich extracellular domain; aberrant ligand binding

In achondroplasia, the G380R mutation accounts for >99% of cases, causing rhizomelic shortening and craniofacial abnormalities .

Skeletal Dysplasias

ConditionIncidenceKey FeaturesGenetic Basis
Achondroplasia1:25,000–30,000Short limbs, macrocephaly, spinal stenosisFGFR3 G380R
Hypochondroplasia1:15,000–40,000Mild short stature, brachydactylyFGFR3 N540K or others
Thanatophoric DysplasiaRareSevere shortening, lethal in type IIFGFR3 K650M, Y373C

Cancers and Skin Disorders

ConditionFGFR3 RoleMutation TypePrognosis
Bladder CancerFusion proteins (FGFR3-TACC3) drive proliferationSomatic mutations or fusionsBetter prognosis in some cases
GlioblastomaFGFR3-TACC3 fusions promote mitogenesisSomatic fusionsImproved survival in subset
Epidermal NevusSomatic mutations cause keratinocyte overgrowthFGFR3 R248C (somatic)Benign but disfiguring

Current Research Directions

StrategyTargetCompoundOutcomeReference
Inhibition of FGFR3Tyrosine kinase domainPheophorbide a (Pa)Reduces FGFR3 stability; alleviates bone defects in mice
Neutralizing AntibodiesExtracellular domainAnti-FGFR3 AbBlocks ligand binding; enhances pancreatic ductal proliferation
Gene EditingFGFR3 G380R mutationCRISPR/Cas9Preclinical models for achondroplasia correction

Mouse Models

Humanized FGFR3 G380R mice recapitulate achondroplasia phenotypes, including craniosynostosis and kyphosis . These models enable testing of therapies like porphyrins, which improve bone growth in vivo .

Pancreatic Epithelial Regulation

FGFR3 inhibits ductal cell proliferation in the pancreas. In FGFR3 knockout mice:

  • Islet areas increase by 2x due to enhanced progenitor cell expansion .

  • Neutralizing FGFR3 antibodies double BrdU+ ductal cells in regenerating pancreata .

Cancer Pathogenesis

In bladder cancer, FGFR3 mutations occur in 15–30% of cases, often as R248C substitutions. These mutations correlate with low-grade, papillary tumors and better survival rates .

Product Specs

Introduction
The Fibroblast Growth Factor (FGF) family consists of at least 18 structurally related proteins, playing diverse roles in physiological and pathological processes such as cell growth, differentiation, angiogenesis, wound healing, and tumorigenesis. FGFs exert their biological activities by binding to and activating a family of type I transmembrane tyrosine kinase receptors known as FGFRs. Upon ligand binding, FGFRs dimerize and undergo autophosphorylation, initiating downstream signaling cascades. Currently, four distinct genes encoding highly similar FGF receptors, FGFR-1 to -4, have been identified. Alternative splicing of mRNAs gives rise to multiple isoforms of FGFR-1 to -3. A common splicing event affects FGFR-1 and -2, resulting in receptors that either contain all three immunoglobulin-like domains (IgI, IgII, and IgIII), referred to as the alpha isoform, or only IgII and IgIII, termed the beta isoform. For FGFR-3 and FGFR-4, only the alpha isoform has been observed. Further splicing events in FGFR-1 to -3, specifically involving the C-terminal region of the IgIII domain encoded by two mutually exclusive exons, generate FGF receptors with variant IgIII domains (IIIb and IIIc). Additionally, a IIIa isoform of FGFR-1 has been reported, which acts as a secreted FGF binding protein and comprises the N-terminal half of the IgIII domain along with some intronic sequences. Notably, mutations in FGFR-1 to -3 have been linked to birth defects characterized by craniosynostosis.
Description
Soluble FGFR3 Human recombinant, a heterodimeric, glycosylated polypeptide chain containing 593 amino acids with a molecular weight of 170 kDa, is produced in Fc Chimera by fusing the protein with the Fc portion of human IgG1 using a Xa cleavage site.
Physical Appearance
The product appears as a sterile, white, lyophilized (freeze-dried) powder.
Formulation
FGFR3 is lyophilized from a sterile solution at a concentration of 1 mg/ml in phosphate-buffered saline (PBS).
Solubility
To reconstitute the lyophilized FGFR-3, it is recommended to dissolve it in sterile PBS at a concentration of at least 50 µg/ml. The reconstituted solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized FGFR3, while stable at room temperature for up to 3 weeks, should be stored desiccated at a temperature below -18°C. After reconstitution, FGFR3 should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to freeze the reconstituted FGFR3 below -18°C after adding a carrier protein, such as 0.1% HSA or BSA. Avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of FGFR3 is greater than 90%, as determined by: (a) Silver stain analysis and (b) SDS-PAGE analysis.
Synonyms
Achondroplasia, Thanatophoric Dwarfism, CD333, ACH, CEK2, JTK4, HSFGFR3EX, FGFR3.
Source
Insect Cells.
Amino Acid Sequence
ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGDP RRASIEGRGD PEEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK. 

Q&A

What is the expression pattern of FGFR3 during human germ cell development?

FGFR3 demonstrates a dynamic expression pattern during human germ cell development. It is prominently expressed by human primordial germ cells (PGCs) during the first and second trimester of development, with expression beginning as early as 5 weeks post-fertilization (pf) at the time of gonadal colonization . As development progresses, FGFR3 expression becomes repressed specifically when PGCs differentiate into primordial oocytes and as meiotic germ cells enter prophase I of meiosis I, marked by the expression of meiosis-specific genes SPO11 and SYCP1 .

Single-cell RNA sequencing (scRNA-seq) data from both 10x Genomics and SMART-seq platforms confirm that FGFR3 mRNA is predominantly expressed in PGCs and retinoic acid-responsive meiotic germ cells, with expression decreasing during meiotic progression . Immunofluorescence studies show that more than 90% of TFAP2C+/VASA low PGCs express FGFR3 protein, while approximately 60% of TFAP2C-/VASA+ meiotic germ cells in cortical cords maintain FGFR3 expression .

How does FGFR3 expression differ between embryonic ovarian and testicular tissues?

FGFR3 expression has been documented in both embryonic ovarian and testicular tissues, though with distinct patterns. In the ovary, FGFR3 is predominantly expressed by PGCs and becomes progressively downregulated as germ cells enter meiosis . Single-cell RNA-seq analyses of human embryonic and fetal ovaries (6-16 weeks pf) show FGFR3 mRNA enrichment in the germ cell population .

In the testis, FGFR3 protein has been previously demonstrated to be dynamically expressed during fetal testicular germ cell development . While both ovarian and testicular tissues show FGFR3 expression in germ cells, the developmental trajectory differs due to the sex-specific timing of meiotic entry. The role of FGFR3 signaling in sex-specific germ cell development represents an important area for further investigation.

Is FGFR3 expression restricted to germ cells in developing gonads?

While FGFR3 is predominantly expressed in germ cells, it is not exclusively restricted to them. Single-cell RNA-seq analysis reveals that rare somatic cells in developing gonads also express FGFR3 mRNA, though typically at lower levels than germ cells . Specifically, a subset of pre-granulosa cells (expressing FOXL2) and endothelial cells (expressing PECAM1) show detectable FGFR3 expression in 8 and 13 week pf ovaries .

How can FGFR3 be used as a marker for isolating human primordial germ cells?

FGFR3 has emerged as a valuable cell surface marker for isolating and enriching human primordial germ cells. Fluorescence-activated cell sorting (FACS) using antibodies recognizing FGFR3, followed by single-cell RNA sequencing, has been shown to effectively enrich for PGCs from prenatal ovarian tissue . This approach offers significant advantages over previous methods that relied on less specific markers.

The methodology involves:

  • Tissue dissociation of prenatal ovaries (typically 7-14 weeks pf)

  • Incubation with fluorophore-conjugated anti-FGFR3 antibodies

  • FACS isolation of FGFR3-positive cells

  • Confirmation of PGC identity through scRNA-seq analysis

This technique is particularly valuable for researchers studying human germ cell development, as it allows for the isolation of a relatively pure population of PGCs from complex tissues. The enrichment of FGFR3-positive cells significantly increases the proportion of PGCs in the sorted population, enabling more precise molecular and functional analyses .

What are the technical considerations when using FGFR3 for fluorescence-activated cell sorting of germ cells?

When implementing FGFR3-based FACS for germ cell isolation, researchers should consider several technical factors:

  • Antibody selection: Choose antibodies that recognize extracellular domains of FGFR3 to ensure detection of surface expression in living cells.

  • Potential somatic cell contamination: While FGFR3 enriches for PGCs, it's important to recognize that rare FGFR3-positive somatic cells may be included in the sorted population. Single-cell RNA sequencing of sorted cells reveals that approximately 2-5% of FGFR3-positive cells may be pre-granulosa or endothelial cells .

  • Developmental timing: FGFR3 expression changes during development, with higher expression in pre-meiotic PGCs compared to meiotic germ cells . Therefore, the enrichment efficiency may vary depending on the developmental stage of the tissue.

  • Validation approach: Confirm the identity of sorted cells using orthogonal methods, such as immunofluorescence for PGC markers (TFAP2C, NANOG, POU5F1) or scRNA-seq to verify the cellular composition of the sorted population .

  • Comparison with in vitro models: While FGFR3 is expressed by in vivo PGCs, its expression pattern may differ in PGC-like cells (PGCLCs) differentiated from human pluripotent stem cells, which should be considered when comparing in vitro and in vivo germ cell development .

What sequence homology exists between human FGFR3 and mouse Fgfr3, and how does this impact experimental design?

Sequence homology between human FGFR3 and mouse Fgfr3 is an important consideration when designing cross-species experiments or developing mouse models of human FGFR3-related conditions. The proteins share significant structural and functional similarities, which is why mouse models can be valuable for studying FGFR3-related human pathologies.

When developing experimental models, researchers have successfully replaced mouse Fgfr3 with human FGFR3 cDNA to generate humanized mouse models . This approach ensures that the human receptor is expressed under the control of the endogenous mouse Fgfr3 promoter, maintaining physiologically relevant expression patterns while allowing assessment of human-specific FGFR3 functions or mutations .

For experimental design, researchers should consider:

  • Whether species-specific antibodies or detection methods are required

  • Potential differences in downstream signaling pathways

  • Whether human-specific mutations (such as G380R in achondroplasia) would have equivalent effects in the mouse protein

The successful generation of mice expressing human FGFR3 under mouse Fgfr3 regulatory elements demonstrates the feasibility of this approach for studying human FGFR3 biology in vivo .

How has the G380R FGFR3 mutation been modeled in mice to study achondroplasia?

The G380R mutation in FGFR3 represents the most common genetic cause of achondroplasia (ACH) in humans. To create a human-relevant ACH mouse model, researchers have generated knock-in mice in which the endogenous mouse Fgfr3 gene was replaced with human FGFR3 cDNA containing the G380R mutation . This model offers significant advantages over previous approaches by expressing the actual human mutant protein rather than a mouse homolog.

The experimental approach involved:

  • Designing a targeting construct containing human FGFR3 G380R cDNA

  • Incorporating the endogenous mouse Fgfr3 promoter, intron 1, and 5′/3′ untranslated regions to ensure proper expression control

  • Generating both heterozygous (FGFR3^ACH/+) and homozygous (FGFR3^ACH/ACH) mice

  • Validating expression by Southern blotting and PCR of genomic DNA

  • Confirming transcription using RT-PCR with gene-specific primers

This model successfully recapitulates the key human ACH phenotypes, including:

  • Growth retardation

  • Disproportionate shortening of the limbs

  • Round head and midface hypoplasia

  • Kyphosis (developing progressively during postnatal growth)

  • Premature fusion of cranial sutures

  • Low bone density

Importantly, the severity of these phenotypes corresponds to the copy number of activated FGFR3, with homozygous mice showing more severe manifestations than heterozygous counterparts .

What phenotypic differences exist between heterozygous and homozygous FGFR3^G380R mice?

The FGFR3^G380R mouse model demonstrates a clear gene dosage effect, with distinct phenotypic differences between heterozygous (FGFR3^ACH/+) and homozygous (FGFR3^ACH/ACH) animals:

Heterozygous (FGFR3^ACH/+) mice:

  • Show more severe growth retardation

  • Develop pronounced kyphosis earlier (typically by 2 weeks of age)

  • Exhibit significantly lower survival rates at birth and higher mortality before 4 weeks of age

  • Most die around 1 year of age

  • Display protrusion of the lower incisors due to skull changes affecting incisor alignment

Homozygous (FGFR3^ACH/ACH) mice:

  • Show intermediate body weights and lengths between wild-type and heterozygous mice

  • About 90% develop kyphosis before 1 month of age

  • Have better survival rates than heterozygous mice

This gene dosage relationship indicates that the severity of achondroplasia-like phenotypes directly correlates with the level of activated FGFR3^G380R expression. Interestingly, the heterozygous condition actually produces more severe phenotypes than the homozygous state for some parameters, suggesting complex effects of FGFR3 signaling on skeletal development .

The control mice expressing non-mutated human FGFR3 (FGFR3^WT/+ or FGFR3^WT/WT) displayed phenotypes identical to wild-type mice, confirming that the observed abnormalities are specifically associated with the G380R mutation rather than expression of human FGFR3 itself .

How do skeletal phenotypes in FGFR3^G380R mice compare with human achondroplasia patients?

The FGFR3^G380R mouse model closely recapitulates the skeletal phenotypes observed in human achondroplasia patients, making it an excellent model for studying the disorder. Key comparative features include:

Similarities with human ACH:

  • Rhizomelic dwarfism (disproportionate shortening of proximal limbs)

  • Rounded skull morphology

  • Midface hypoplasia present at birth

  • Kyphosis developing during postnatal growth

  • Craniosynostosis (premature fusion of cranial sutures)

  • Low bone density

Developmental progression:
In both humans and the mouse model, certain phenotypes (such as kyphosis) develop postnatally rather than being present at birth. The FGFR3^G380R mice show ACH phenotypes at birth that become more pronounced during postnatal skeletal development .

What makes this model particularly valuable is that it expresses the actual human FGFR3 protein with the precise mutation found in human patients, rather than a mouse homolog. This feature is crucial for testing therapeutic approaches designed specifically to target human FGFR3 signaling, such as antibodies or small molecules that might not interact identically with mouse Fgfr3 .

What experimental approaches are most effective for studying FGFR3 signaling in human cells?

Studying FGFR3 signaling in human cells can be approached through several complementary methodologies:

  • Primary tissue analysis:

    • Single-cell RNA sequencing of tissues expressing FGFR3 (embryonic gonads, growth plates)

    • Immunohistochemistry with phospho-specific antibodies to detect activated signaling components

    • Laser capture microdissection to isolate FGFR3-expressing cells for molecular analysis

  • Cell culture systems:

    • Human cell lines expressing wild-type or mutant FGFR3

    • Primary cells isolated using FGFR3-based FACS

    • Induced pluripotent stem cell (iPSC) derivatives modeling FGFR3-dependent lineages

  • Signaling pathway analysis:

    • Phospho-proteomics to identify downstream effectors

    • Inhibitor studies targeting specific branches of FGFR3 signaling

    • CRISPR/Cas9-mediated genome editing to modify FGFR3 or pathway components

  • Functional readouts:

    • Cell proliferation, differentiation, and survival assays

    • Transcriptional reporter assays for FGFR3-responsive elements

    • Morphological and cytoskeletal analyses

When designing experiments to study FGFR3 signaling, it's important to consider the cellular context, as FGFR3 may activate different downstream pathways depending on the cell type and developmental stage. Additionally, the specific FGFR3 ligands present in the experimental system can significantly influence receptor activation and signaling outcomes.

How can researchers distinguish between normal and pathological FGFR3 signaling in experimental systems?

Distinguishing between normal and pathological FGFR3 signaling requires careful experimental design and multiple analytical approaches:

  • Quantitative analysis of signaling intensity:

    • Comparing phosphorylation levels of FGFR3 and downstream effectors

    • Measuring the kinetics of pathway activation and deactivation

    • Assessing nuclear translocation of signaling components

  • Pathway specificity assessment:

    • Analyzing which downstream pathways are preferentially activated (MAPK, STAT, PLCγ)

    • Comparing the balance between different signaling branches

    • Identifying pathway-specific transcriptional responses

  • Cellular outcome evaluation:

    • Growth plate chondrocyte proliferation versus differentiation rates

    • Cell cycle progression analysis

    • Extracellular matrix production and organization

  • Comparative studies:

    • Parallel analysis of wild-type and mutant FGFR3 (e.g., G380R)

    • Dose-response studies with varying levels of receptor activation

    • Comparison with other FGFR3 mutations causing different disorders

  • In vivo validation:

    • Using the FGFR3^G380R mouse model to confirm in vitro findings

    • Analyzing tissue-specific effects in different FGFR3-expressing lineages

    • Correlating molecular findings with phenotypic outcomes

What are the recommended approaches for validating potential therapeutic targets for FGFR3-related disorders?

Validating potential therapeutic targets for FGFR3-related disorders requires a systematic, multi-level approach:

  • Target identification and validation:

    • Identify components of the FGFR3 signaling pathway through phospho-proteomics or transcriptomics

    • Validate targets using CRISPR/Cas9-mediated knockout or RNAi in relevant cell types

    • Confirm that target inhibition counteracts the effects of FGFR3 hyperactivation

  • In vitro proof-of-concept studies:

    • Test candidate compounds in cell lines expressing mutant FGFR3

    • Assess effects on proliferation, differentiation, and FGFR3 signaling

    • Determine dose-response relationships and potential off-target effects

  • Ex vivo tissue models:

    • Culture growth plate cartilage from FGFR3^G380R mice with candidate compounds

    • Assess restoration of normal chondrocyte maturation and organization

    • Measure growth parameters and extracellular matrix production

  • In vivo preclinical testing:

    • Administer candidates to FGFR3^G380R mice at different developmental stages

    • Monitor skeletal growth parameters, bone density, and cranial suture closure

    • Evaluate dose-dependent effects and treatment duration requirements

  • Safety and specificity assessment:

    • Evaluate effects on wild-type FGFR3 signaling in non-target tissues

    • Assess potential impacts on other FGF receptors (FGFR1, FGFR2, FGFR4)

    • Determine developmental stage-specific safety profiles

The FGFR3^G380R mouse model offers a particularly valuable tool for assessing therapeutic approaches before human clinical trials, as it expresses the human FGFR3 protein with the exact mutation found in achondroplasia patients . This allows for testing of compounds designed specifically to target human FGFR3, such as human-specific antibodies or aptamers that might not interact with mouse Fgfr3.

How do other FGFR3 mutations differ from G380R in their effects on receptor function and cellular outcomes?

While the G380R mutation is the most common cause of achondroplasia, numerous other FGFR3 mutations have been identified in skeletal dysplasias with varying phenotypic severity. Understanding the mechanistic differences between these mutations is crucial for developing targeted therapies:

  • Activation mechanism differences:

    • G380R (achondroplasia) causes ligand-independent dimerization through the transmembrane domain

    • K650E (thanatophoric dysplasia type II) directly activates the kinase domain

    • R248C (thanatophoric dysplasia type I) creates an unpaired cysteine in the extracellular domain

  • Signaling intensity variation:

    • Different mutations can produce varying levels of receptor activation

    • More severe conditions (thanatophoric dysplasia) typically show stronger activation than milder conditions (achondroplasia)

    • The relationship between activation level and phenotypic outcome is not always linear

  • Pathway specificity:

    • Different mutations may preferentially activate certain downstream pathways

    • This could explain why some mutations affect certain tissues more than others

  • Receptor trafficking and turnover:

    • Some mutations may affect receptor internalization and degradation

    • Altered receptor half-life can significantly impact signaling duration and intensity

Future research should systematically compare different FGFR3 mutations in identical experimental systems to directly assess these differences. This would involve creating parallel mouse models with different human FGFR3 mutations using the same knock-in approach used for the G380R model .

What is the interplay between FGFR3 signaling and other developmental pathways in human germ cell and skeletal development?

FGFR3 signaling does not operate in isolation but interacts with numerous other developmental pathways. Understanding these interactions is critical for comprehending the full impact of FGFR3 dysregulation:

In germ cell development:

  • FGFR3 expression in primordial germ cells coincides with key developmental transitions, suggesting potential crosstalk with pathways governing PGC specification and meiotic entry

  • The downregulation of FGFR3 as PGCs enter meiosis indicates possible interactions with retinoic acid signaling, which is known to trigger meiotic initiation

  • The sex-specific dynamics of FGFR3 expression suggest interactions with pathways that regulate sexual differentiation of germ cells

In skeletal development:

  • FGFR3 signaling interacts with Indian hedgehog (IHH) and parathyroid hormone-related protein (PTHrP) signaling in the growth plate

  • Bone morphogenetic protein (BMP) and WNT signaling pathways likely modulate the effects of FGFR3 activation

  • The severity of phenotypes in FGFR3^G380R mice suggests that compensatory mechanisms may be overwhelmed by hyperactive FGFR3 signaling

Future research should employ systems biology approaches to map these interactions comprehensively, potentially through:

  • Parallel single-cell transcriptomics of wild-type and FGFR3 mutant tissues

  • Proteomic analysis of signaling complexes formed in the presence of normal versus mutant FGFR3

  • Genetic interaction studies combining FGFR3 mutations with modifications to interacting pathways

How can single-cell multi-omics approaches advance our understanding of FGFR3 biology and pathology?

Single-cell multi-omics approaches offer unprecedented opportunities to understand FGFR3 biology and pathology with high resolution:

  • Integrative single-cell analyses:

    • Combining scRNA-seq with single-cell ATAC-seq to correlate FGFR3 expression with chromatin accessibility

    • Integrating single-cell proteomics to assess post-transcriptional regulation

    • Implementing spatial transcriptomics to maintain tissue context information

  • Developmental trajectory mapping:

    • Analyzing FGFR3 expression dynamics across developmental time points

    • Identifying branch points where FGFR3 influences cell fate decisions

    • Comparing normal and pathological developmental trajectories

  • Cell-cell interaction analysis:

    • Examining how FGFR3-expressing cells communicate with neighboring populations

    • Identifying secondary effects of FGFR3 dysregulation on the tissue microenvironment

    • Modeling signaling networks across multiple cell types

  • Response to therapeutic interventions:

    • Measuring cell type-specific responses to FGFR3-targeted therapies

    • Identifying resistant cell populations and compensatory mechanisms

    • Optimizing treatment timing based on developmental trajectories

The application of these approaches to samples from both the FGFR3^G380R mouse model and human patient tissues could provide unprecedented insights into the molecular basis of FGFR3-related disorders and identify new therapeutic targets.

Product Science Overview

Introduction

Fibroblast Growth Factor Receptor 3 (FGFR3) is a member of the fibroblast growth factor receptor family, which plays a crucial role in various cellular processes including cell growth, differentiation, angiogenesis, wound healing, and tumorigenesis . FGFR3 is a transmembrane tyrosine kinase receptor that belongs to the immunoglobulin (Ig) superfamily and binds to fibroblast growth factors (FGFs) . The receptor is involved in cell survival, proliferation, migration, and differentiation .

Structure and Isoforms

FGFR3 consists of an extracellular ligand-binding domain, a single transmembrane domain, and an intracellular tyrosine kinase domain . The extracellular domain contains three immunoglobulin-like (Ig-like) loops, a short cluster of conserved acidic amino acids, and a heparin-binding domain . Alternative splicing in the Ig-like loop III results in IIIb and IIIc isoforms of FGFR1-3, which change the receptor specificity . FGFR3 IIIc binds many FGFs including FGF-1, -2, -4, -6, -8, -9, -18, -19, and -20, while FGFR IIIb only binds to FGF-1, -9, and -16 .

FGFR3 Fc Chimera

The FGFR3 Fc Chimera is a recombinant protein that combines the extracellular domain of FGFR3 with the Fc region of human IgG1 . This fusion protein is designed to enhance the stability and solubility of FGFR3, making it suitable for various research applications . The Fc region also allows for easy purification and detection using Fc-specific antibodies .

Preparation and Production

The recombinant FGFR3 Fc Chimera is typically produced in mammalian cell lines such as NS0 or 293E cells . The protein is expressed with a linker sequence and a C-terminal human IgG1, resulting in a disulfide-linked homodimer . The recombinant protein is then purified using affinity chromatography and characterized by SDS-PAGE to ensure high purity and correct molecular weight .

Applications and Significance

FGFR3 Fc Chimera is widely used in research to study the role of FGFR3 in various cellular processes and diseases . It is particularly useful in cancer research, as FGFR3 is one of the most mutated genes in human cancers . The recombinant protein can be used to inhibit FGF-dependent proliferation of cells, making it a valuable tool for studying the signaling pathways involved in cancer and other diseases .

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.