FGF4 Human

Fibroblast Growth Factor-4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure

FGF4 is a non-glycosylated polypeptide composed of 182–206 amino acids, depending on the isoform. Its mature form (aa 71–206) includes a conserved FGF homology domain with a heparin-binding region near the C-terminus . The recombinant human FGF4 protein (e.g., CYT-312, 235-F4) is produced in E. coli and purified to >97% purity .

Embryonic Development

FGF4 is essential for:

  • Inner cell mass survival: Acts as an autocrine/paracrine ligand during postimplantation development .

  • Limb patterning: Collaborates with FGF8 in the apical ectodermal ridge (AER) to regulate digit number and interdigital apoptosis. Overexpression causes polysyndactyly .

  • Visceral organ left-right asymmetry: Knockdown in zebrafish disrupts liver, pancreas, and heart patterning .

Bone and Cartilage Formation

  • Endochondral ossification: FGF4 retrogene insertions in dogs (chromosomes 12/18) lead to dwarfism and intervertebral disc disease (IVDD) due to defective long bone growth .

Gestational Diabetes Mellitus (GDM)

Elevated FGF4 levels in GDM placentas correlate with hyperglycemia . In mouse models:

  • Recombinant FGF4 (rFGF4) improves maternal glucose tolerance and reduces neural tube defects (NTDs) .

  • Mechanism: Inhibits placental inflammation and neuroepithelial apoptosis independently of glucose regulation .

ModelOutcomeSource
HFD-induced GDM mice↓ Blood glucose (15–30 min GTT), ↓ NTDs
STZ-induced GDM mice↓ NTDs, ↓ placental inflammation

Tumor Progression

FGF4 is co-amplified with FGF3 in human tumors, driving angiogenesis and proliferation .

Stem Cell Biology

FGF4 is critical for:

  • Embryonic stem cell self-renewal: Included in culture media for human embryonic stem cells (hESCs) .

  • Intestinal organoid generation: Synergizes with WNT signaling to direct iPSC-derived endoderm into hindgut structures .

Tissue Expression

Data from the Human Protein Atlas :

TissueExpressionNotes
PlacentaHighElevated in GDM placentas
BrainModerateRole in neural progenitor cells
Small IntestineLowLinked to hindgut differentiation

Recombinant FGF4 Products

SupplierCatalog NumberKey FeaturesSource
Prospec BioCYT-312Lyophilized, 2xPBS formulation, non-glycosylated
R&D Systems235-F4>97% purity, cited in 76+ publications
QkineQk00414 kDa bioactive domain, AOF, EC₅₀ = 4.3 ng/mL

Quality Assurance

  • Purity: SDS-PAGE confirms single bands at 14–19.7 kDa (reduced/non-reduced) .

  • Stability: Avoid freeze-thaw cycles; store at -20°C .

Product Specs

Introduction

Fibroblast growth factor 4 (FGF4) is a protein that plays a role in cell growth, development, and repair. It is involved in a variety of biological processes, including embryonic development, wound healing, and tumor growth. FGF4 is a potent mitogen, meaning that it stimulates cell division. It also promotes cell survival and differentiation. FGF4 is overexpressed in several types of cancer, and it has been implicated in tumor progression and metastasis. FGF4 binds to and activates the fibroblast growth factor receptor (FGFR) family of receptor tyrosine kinases. Activation of FGFRs leads to the activation of downstream signaling pathways that regulate cell proliferation, survival, and differentiation. FGF4 is a potential therapeutic target for the treatment of cancer and other diseases.

Description

Recombinant human FGF4 is a single, non-glycosylated polypeptide chain containing 182 amino acids with a molecular mass of 19.8 kDa. The protein is produced in E. coli and purified using proprietary chromatographic techniques.

Physical Appearance

The appearance of this product is a sterile, filtered, white, lyophilized (freeze-dried) powder.

Formulation

This product is formulated with 2X phosphate-buffered saline (PBS) at a pH of 7.4.

Solubility

To reconstitute the lyophilized FGF4, it is recommended to dissolve it in sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can then be further diluted into other aqueous solutions.

Stability

Lyophilized FGF4 is stable at room temperature for 3 weeks. However, it is recommended to store the lyophilized product desiccated below -18°C. After reconstitution, FGF4 should be stored at 4°C for 2-7 days or below -18°C for longer-term storage. Avoid repeated freeze-thaw cycles.

Purity

The purity of this product is greater than 95.0% as determined by SDS-PAGE analysis.

Biological Activity

The biological activity of this product was determined by measuring its ability to stimulate the proliferation of NR6R-3T3 cells. The ED50 was found to be 0.543 ng/ml, corresponding to a specific activity of 1.8 x 10^6 units/mg.

Synonyms
HBGF4, FGF-4, FGF4, KFGF, HSTF1.
Source
Escherichia Coli.
Amino Acid Sequence

GRGGAAAPTA PNGTLEAELE RRWESLVALS LARLPVAAQP KEAAVQSGAG DYLLGIKRLR RLYCNVGIGF HLQALPDGRI GGAHADTRDS LLELSPVERG VVSIFGVASR FFVAMSSKGK LYGSPFFTDE CTFKEILLPN NYNAYESYKY PGMFIALSKN GKTKKGNRVS PTMKVTHFLP RL.

Q&A

What is FGF4 and what roles does it play in human development?

FGF4 is a 21-kDa protein encoded by the FGF4 gene in humans that functions as a signaling molecule during embryonic development. It belongs to the fibroblast growth factor family, whose members possess broad mitogenic and cell survival activities . FGF4 plays vital roles in multiple developmental processes, including:

  • Regulation of embryonic development, particularly in the post-implantation phase where it facilitates survival and growth of the inner cell mass

  • Promotion of cell proliferation and differentiation

  • Development of normal limbs and cardiac valves during embryogenesis

  • Embryonic molar tooth bud development through inducing MSX1, MSX2, and MSX1-mediated expression of SDC1 in dental mesenchyme cells

  • Left-right patterning of visceral organs including liver, pancreas, and heart (based on zebrafish studies)

  • Hindgut identity during gastrointestinal development

The FGF4 protein acts through the FGF receptor family to activate several signaling cascades, ultimately influencing gene expression patterns during development. Knockout studies in mice have demonstrated developmental defects both in vivo and in vitro, highlighting FGF4's essential nature in embryogenesis .

How is FGF4 expression regulated in human embryonic stem cells?

FGF4 demonstrates a specific expression pattern in human embryonic stem cells (HESCs) that is tightly regulated to maintain pluripotency. Research indicates that FGF4 is produced by multiple undifferentiated HESC lines, suggesting an autocrine signaling mechanism .

The regulation involves:

  • Active expression in undifferentiated HESCs and early differentiated cells

  • Cessation of expression in mature differentiated cells

  • Concurrent expression of a novel FGF4 splice isoform (FGF4si) that acts as a feedback inhibitor

  • Targeted knockdown of FGF4 using siRNA increases differentiation of HESCs, demonstrating the importance of endogenous FGF4 signaling in maintaining pluripotency

This expression pattern suggests that FGF4 plays a crucial role in the self-renewal of HESCs while maintaining their pluripotent state. The interplay between FGF4 and its splice isoform appears to create a regulatory mechanism that balances growth promotion and differentiation inhibition in stem cell populations .

What are the molecular characteristics of recombinant human FGF4?

Recombinant human FGF4 protein has specific molecular and biophysical properties important for research applications:

CharacteristicDescription
Molecular Mass17 and 27 kDa (reduced and non-reduced, monomer, glycosylated)
Activity≤ 1.25 ng/mL in Balb/c 3T3 cells; 6-30 ng/mL in NIH 3T3 cells in defined media
Purity>95% for research-grade preparations
Endotoxin level<1 EU/μg for cell culture applications
Species ReactivityHuman, mouse
Storage stabilityLyophilized proteins are stable for 1 year between -20°C and -80°C

When working with recombinant FGF4, researchers should reconstitute it to approximately 0.2 mg/mL in sterile 1x PBS (pH 7.4) and avoid repeated freeze-thaw cycles to maintain bioactivity . This information is critical for experimental design involving exogenous FGF4 in cell culture systems.

How does the FGF4 splice isoform (FGF4si) antagonize FGF4 function in stem cell biology?

The novel FGF4 splice isoform (FGF4si) represents a sophisticated regulatory mechanism in stem cell biology. This isoform codes for only the amino-terminal half of FGF4 and functions as an endogenous antagonist to full-length FGF4 . The mechanisms of this antagonism include:

  • Direct countering of FGF4's growth-promoting effects on HESCs

  • Shutting down FGF4-induced Erk1/2 phosphorylation, thereby inhibiting downstream signaling

  • Differential expression patterns between FGF4 and FGF4si during differentiation:

    • Both isoforms are expressed in HESCs and early differentiated cells

    • FGF4 expression ceases in mature differentiated cells

    • FGF4si continues expression after cell differentiation

This antagonistic relationship suggests a carefully balanced feedback inhibition mechanism where FGF4 promotes undifferentiated growth while FGF4si may facilitate differentiation at later developmental stages. This represents an elegant molecular switch controlling stem cell fate decisions .

Experimentally, researchers can study this interaction by:

  • Using recombinant proteins to test the dose-response relationships

  • Selective knockdown of each isoform using targeted siRNAs

  • Monitoring downstream signaling events (particularly MAPK pathway activation)

  • Tracking expression ratios of FGF4:FGF4si during differentiation experiments

What in-silico approaches can identify pathogenic SNPs in the FGF4 gene?

Computational prediction of pathogenic non-synonymous single nucleotide polymorphisms (nsSNPs) in the FGF4 gene involves a multi-tool bioinformatic workflow. Based on recent research, the following methodological approach has proven effective :

  • SNP retrieval and initial screening:

    • Retrieve SNP data from databases like NCBI dbSNP

    • Obtain protein sequence in FASTA format from UniProtKB

    • Filter for non-synonymous SNPs that alter amino acid sequences

  • Pathogenicity prediction using multiple algorithms:

    • PROVEAN: Identifies "deleterious" variants

    • SIFT: Classifies SNPs as "damaging"

    • PolyPhen-2: Categorizes as "probably damaging" or "possibly damaging"

    • PhD-SNP and SNPs&GO: Predict "disease" associations

    • PMut: Scores >0.5 indicate pathogenicity

  • Conservation and structural analysis:

    • Use ConSurf to analyze evolutionary conservation

    • Identify conserved and exposed residues (e.g., Q97K, G106V, N164S, and N167S)

    • Employ SWISS-Model for 3D structural alteration predictions

  • Functional and network analysis:

    • Analyze protein-protein interactions using Cytoscape and STRING databases

    • Identify involvement in critical pathways (like GFR-MAPK signaling)

    • Correlate with clinical outcomes using Kaplan-Meier bioinformatics analyses

This comprehensive approach can identify high-risk nsSNPs with potential clinical significance, such as those affecting bladder cancer prognosis .

How does FGF4 coordinate with Wnt signaling in intestinal development and organoid formation?

The interplay between FGF4 and Wnt signaling represents a critical regulatory network in intestinal development. Research on human pluripotent stem cell (hPSC) differentiation into intestinal lineages has revealed several key aspects of this coordination :

These findings suggest a model where Wnt signaling drives intestinal specification, while FGF4 helps delineate boundaries between different endodermal lineages. For experimental applications, researchers should note that excessive FGF4 can be detrimental to intestinal organoid development, while Wnt modulation can direct specific intestinal cell type differentiation .

What methodological considerations are critical when using antibodies to detect FGF4 in experimental systems?

When designing experiments to detect FGF4 protein expression, several methodological considerations are essential for reliable results:

  • Antibody selection criteria:

    • Validated antibodies with confirmed specificity for human or mouse FGF4

    • Rabbit polyclonal antibodies like ab106355 have been validated for multiple applications

    • Consider cross-reactivity with the FGF4 splice isoform when interpreting results

  • Validated applications and working concentrations:

    • Western blot: 0.5-1 μg/mL concentration range

    • Immunocytochemistry/Immunofluorescence: 2.5 μg/ml

    • Appropriate positive controls (e.g., NIH 3T3 cells) should be included

  • Sample preparation considerations:

    • FGF4 can be sequestered by extracellular matrix components

    • Proper extraction methods are essential for detecting secreted FGF4

    • Consider using protein transport inhibitors to detect intracellular pools

  • Epitope availability:

    • Ensure antibody recognizes epitopes within the immunogen sequence (human FGF4)

    • Post-translational modifications may affect antibody binding

    • For glycosylated FGF4 (17 and 27 kDa forms), ensure antibody recognizes both forms

  • Detection sensitivity limitations:

    • FGF4 is often expressed at low levels, requiring sensitive detection methods

    • Consider signal amplification methods for low-abundance samples

    • Compare results across multiple detection platforms for confirmation

Proper controls, including FGF4 knockdown samples and recombinant protein standards, should be incorporated into experimental design to validate findings and ensure accurate interpretation of results.

How can researchers effectively use FGF4 in stem cell culture systems?

Effective use of FGF4 in stem cell culture requires careful consideration of multiple parameters:

  • Source and quality of FGF4:

    • Use HEK293-expressed recombinant human FGF4 for highest bioactivity

    • Ensure preparations are endotoxin-free (<1 EU/μg) to prevent non-specific effects

    • Verify glycosylation status as this affects bioactivity

  • Concentration optimization:

    • Activity range varies by cell type: ≤1.25 ng/mL for Balb/c 3T3 cells; 6-30 ng/mL for NIH 3T3 cells

    • For human embryonic stem cells, determine optimal concentration through dose-response experiments

    • Be aware that excessive concentrations may promote differentiation rather than maintenance

  • Stability and storage considerations:

    • Reconstitute lyophilized protein to 0.2 mg/mL in sterile 1x PBS pH 7.4

    • Store reconstituted protein at 4°C for short-term use or at -20°C to -80°C for long-term

    • Avoid repeated freeze-thaw cycles to maintain bioactivity

  • Combinatorial signaling:

    • Consider interactions with other growth factors and signaling pathways

    • For intestinal differentiation, combine with Wnt modulators like CHIR99021

    • For maintaining pluripotency, consider the balance with other factors like bFGF

  • Monitoring efficacy:

    • Track expression of pluripotency markers (for maintenance) or lineage-specific markers (for differentiation)

    • Monitor morphological changes and proliferation rates

    • Consider measuring activation of downstream signaling (Erk1/2 phosphorylation)

  • Addressing FGF4si influence:

    • Be aware that endogenous FGF4si may counteract exogenous FGF4

    • Consider using recombinant FGF4 variants resistant to FGF4si antagonism

    • Monitor relative expression levels of FGF4 and FGF4si during experiments

These considerations will help researchers optimize FGF4 usage for specific experimental objectives in stem cell research.

What approaches can be used to study FGF4 retrogene functions in developmental disorders?

While FGF4 retrogenes have been primarily studied in canines related to short leg phenotypes and intervertebral disc disease , the methodologies can be adapted to study potential roles in human developmental disorders:

  • Genomic identification approaches:

    • Whole genome sequencing to identify potential FGF4 retrogene insertions

    • PCR-based detection methods for known insertion sites

    • Comparison of evolutionarily conserved regions across species

  • Functional characterization:

    • CRISPR-Cas9 mediated insertion of FGF4 retrogenes into model organism genomes

    • Creation of patient-specific iPSC lines with FGF4 retrogene variants

    • Differentiation of modified stem cells into relevant lineages (bone, cartilage, intervertebral disc)

  • Signaling pathway analysis:

    • Investigate effects on endochondral ossification pathways

    • Analyze FGF receptor specificity and signaling dynamics

    • Study interactions with other developmental signaling pathways (Hedgehog, BMP)

  • Phenotypic assessment:

    • Micro-CT analysis of skeletal development in model organisms

    • Histological analysis of growth plate structure and organization

    • Measurement of proliferation and differentiation in chondrocyte cultures

  • Clinical correlation studies:

    • Analyze human patient cohorts with skeletal dysplasias for FGF4 retrogene presence

    • Perform genotype-phenotype correlation studies

    • Develop genetic screening tools for at-risk populations

This multi-faceted approach can help translate findings from veterinary genetics to human developmental disorders involving similar pathways, particularly those affecting skeletal development and intervertebral disc health.

How can organoid models be optimized to study FGF4 function in intestinal development?

Organoid models offer powerful tools for studying FGF4's role in intestinal development, but require careful optimization:

  • Differentiation protocol optimization:

    • Begin with high-quality hPSC-derived definitive endoderm

    • Use Wnt agonist CHIR99021 for optimal CDX2 (intestinal marker) induction

    • Consider timing and concentration of FGF4 exposure, as it inhibits both formation and maturation of intestinal organoids at later stages

  • 3D culture system design:

    • hPSC-derived hindgut cells can form self-renewing organoid structures containing all major intestinal cell types

    • Unlike adult intestinal organoids, exogenous R-spondin1 may be dispensable due to the presence of a mesenchymal compartment

    • Matrix composition affects organoid formation efficiency and maturation

  • Growth factor modulation:

    • WNT3A increases expression of Paneth cell marker Lysozyme and can direct specific cell type differentiation

    • Careful titration of FGF4 is essential as it can inhibit intestinal organoid formation and maturation

    • Consider temporal modulation of signaling to mimic developmental progression

  • Assessment methods:

    • Immunohistochemistry for intestinal cell type markers (enterocytes, goblet cells, enteroendocrine cells, Paneth cells)

    • qRT-PCR analysis of stage-specific intestinal genes

    • Functional assays (barrier function, absorption, secretion)

    • Single-cell RNA sequencing to identify cell populations and lineage trajectories

  • Patient-specific applications:

    • Similar hindgut and organoid cultures can be established from human induced pluripotent stem cells

    • This approach enables patient-specific intestinal tissue models for disease modeling in vitro

    • Consider disease-relevant functional readouts when designing experiments

These optimization strategies can help researchers develop more physiologically relevant intestinal organoid models to study FGF4's complex role in intestinal development and disease.

Why might FGF4 knockdown experiments yield inconsistent results?

Inconsistent results in FGF4 knockdown experiments can arise from several methodological challenges:

By addressing these factors, researchers can improve consistency in FGF4 knockdown experiments and better interpret the resulting phenotypes.

How can contradictory findings about FGF4's role in differentiation be reconciled?

The literature contains seemingly contradictory findings regarding FGF4's role in differentiation processes. These can be reconciled through several considerations:

  • Context-dependent signaling:

    • FGF4 promotes self-renewal in human embryonic stem cells

    • Yet it inhibits formation and maturation of intestinal organoids

    • Different cellular contexts alter downstream signaling outcomes

  • Biphasic dose-response:

    • Low concentrations may promote maintenance while higher doses induce differentiation

    • Carefully controlled dose-response experiments are essential

    • Consider measuring both phospho-ERK (acute signaling) and transcriptional responses

  • Temporal specificity:

    • FGF4's effects change based on developmental timing

    • Early exposure may block hepatic lineage to permit intestinal development

    • Later exposure inhibits intestinal organoid formation and maturation

    • Design experiments with precise temporal control of FGF4 exposure

  • Interaction with parallel signaling pathways:

    • FGF4 effects are modulated by concurrent signaling (e.g., Wnt, BMP)

    • Wnt signaling dominates intestinal commitment while FGF4 plays a complementary role

    • Analyze pathway interactions rather than isolated effects

  • Splice isoform balance:

    • The ratio of FGF4 to FGF4si affects net signaling outcome

    • FGF4 promotes undifferentiated growth while FGF4si may promote differentiation

    • Measure both isoforms when interpreting experimental results

By considering these factors, researchers can develop more nuanced models of FGF4 function that accommodate apparently contradictory findings across different experimental systems.

What emerging technologies could advance our understanding of FGF4 signaling dynamics?

Several cutting-edge technologies show promise for deepening our understanding of FGF4 signaling:

  • Single-cell multi-omics approaches:

    • Single-cell RNA-seq to map FGF4 and receptor expression at cellular resolution

    • Single-cell ATAC-seq to identify chromatin accessibility changes downstream of FGF4

    • Spatial transcriptomics to visualize FGF4 signaling gradients in developing tissues

  • Live-cell signaling reporters:

    • FRET-based sensors for real-time visualization of FGF4-induced ERK activation

    • Fluorescent fusion proteins to track FGF4 and FGF4si localization and trafficking

    • Optogenetic control of FGF receptor activation for precise temporal manipulation

  • Advanced organoid technologies:

    • Multi-lineage organoids to study FGF4's role in tissue interactions

    • Microfluidic organ-on-chip systems with controlled gradient formation

    • Bioprinting approaches to create complex 3D tissue architectures with defined FGF4 signaling

  • Computational modeling approaches:

    • Systems biology models of FGF4 signaling networks

    • Machine learning to predict FGF4 response patterns from multi-omics data

    • In silico prediction of pathogenic variants and their functional consequences

  • Genome editing with precise control:

    • Base editing for studying specific FGF4 variants without disrupting gene structure

    • Prime editing for introducing complex modifications at endogenous loci

    • CRISPR activation/repression systems to modulate endogenous FGF4 expression

These emerging technologies will enable researchers to move beyond static snapshots of FGF4 function toward dynamic, systems-level understanding of its role in development and disease.

How might FGF4 research inform regenerative medicine approaches?

FGF4 research has several promising applications in regenerative medicine:

  • Directed differentiation protocols:

    • Precise temporal control of FGF4 signaling can guide stem cell differentiation

    • Understanding the interplay between FGF4 and Wnt signaling improves intestinal differentiation protocols

    • Manipulating FGF4:FGF4si ratios may provide novel control points for differentiation processes

  • Tissue-specific organoid development:

    • FGF4's role in intestinal development informs protocols for patient-specific intestinal organoids

    • These models can be used for disease modeling and drug screening

    • Optimization of FGF4 signaling may improve organoid maturation and functionality

  • Cell therapy applications:

    • Knowledge of FGF4's role in maintaining stemness informs expansion protocols for therapeutic cells

    • Understanding FGF4's tissue-specific effects guides appropriate growth factor cocktails for different applications

    • FGF4-based approaches may improve cell engraftment and function

  • Biomaterial design:

    • FGF4-loaded scaffolds could provide sustained signaling for tissue regeneration

    • Engineered FGF4 variants with enhanced stability or receptor specificity

    • Spatially controlled release systems to create developmental signaling gradients

  • Disease-targeting approaches:

    • Targeting pathogenic FGF4 variants identified through in-silico analysis

    • Development of splice-switching oligonucleotides to modulate FGF4:FGF4si ratios

    • Small molecule modulators of specific FGF4 signaling branches

By translating basic FGF4 research findings into therapeutic strategies, researchers can develop more effective regenerative medicine approaches for conditions ranging from gastrointestinal disorders to skeletal dysplasias.

How can findings across different experimental systems be integrated into a comprehensive model of FGF4 function?

Integrating diverse experimental findings into a coherent model of FGF4 function requires consideration of several key principles:

  • Developmental stage specificity:

    • FGF4 function varies dramatically across developmental stages

    • Early roles in embryonic stem cell maintenance

    • Mid-stage roles in lineage specification and boundary formation

    • Later roles in tissue maturation and organogenesis

  • Tissue context dependency:

    • FGF4 promotes limb development through the sonic hedgehog (SHH) signaling pathway

    • It drives hindgut identity during gastrointestinal development

    • FGF4 and FGF8 cooperate in limb development but function independently in visceral organ left-right patterning

  • Signaling integration nodes:

    • Interaction with Wnt/β-catenin signaling in intestinal development

    • Coordination with multiple pathways in the GFR-MAPK signaling network

    • Feedback mechanisms through splice variants like FGF4si

  • Evolutionary conservation and divergence:

    • Core developmental functions are conserved across species

    • Species-specific adaptations exist, such as FGF4 retrogenes in canines

    • Human-specific regulatory mechanisms may involve the FGF4si splice variant

  • Pathological implications:

    • Deregulation affects processes from development to cancer progression

    • nsSNPs may serve as potential biomarkers for conditions like bladder cancer

    • Understanding normal function informs therapeutic targeting strategies

Product Science Overview

Structure and Expression

FGF-4 is a heparin-binding growth factor and is expressed during embryonic development and in several tissues during adult life . The human recombinant form of FGF-4 is typically produced in human 293 cells and is a monomeric glycoprotein with an apparent molecular mass of 17 and 27 kDa . It binds to fibroblast growth factor receptors FGFR1c, FGFR2c, FGFR3c, and FGFR4, with varying affinities .

Biological Functions

FGF-4 is known for its mitogenic activity, promoting the proliferation of fibroblasts and endothelial cells . It is a potent promoter of angiogenesis, which is the formation of new blood vessels, and is believed to be critical in embryonic limb development . Additionally, FGF-4 plays a significant role in cell differentiation and migration .

Applications in Research and Medicine

Recombinant human FGF-4 is widely used in cell culture, differentiation studies, and functional assays . It is particularly important in the self-renewal of human embryonic stem cells and may aid in their differentiation . FGF-4 is also used to study FGFR and SHH signaling pathways .

Pathological Roles

While FGF-4 is essential for normal development and tissue regeneration, it can also have pathological roles. Increased levels of FGF-4 have been found in several human tumors, indicating its potential role as an oncogene in tumor growth . Altered levels of FGF-4 during embryonic development can result in abnormal limb formation .

Quality and Stability

Recombinant human FGF-4 is produced under stringent quality control measures to ensure high purity and biological activity. It is typically endotoxin-free and has a purity of over 95% . The protein is stable when stored at -20°C to -80°C and should be reconstituted in sterile 1x PBS pH 7.4 for use .

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.