Fibroblast growth factor receptor 4 (FGFR4) is a transmembrane tyrosine kinase receptor encoded by the FGFR4 gene located on chromosome 5q35.1 in humans . It belongs to the FGFR family (FGFR1–4), which regulates critical processes such as embryonic development, tissue repair, and metabolism . Structurally, FGFR4 consists of:
Three extracellular immunoglobulin (Ig)-like domains (IgI, IgII, IgIII) for ligand binding.
A transmembrane domain anchoring the receptor to the cell membrane.
An intracellular tyrosine kinase domain responsible for downstream signal transduction .
Unlike FGFR1–3, FGFR4 lacks splice variants in the IgIII domain, enabling high-affinity binding to ligands like fibroblast growth factor 19 (FGF19) .
FGFR4 regulates:
FGFR4 overexpression or dysregulation is implicated in multiple cancers:
The FGFR4 Gly388Arg (rs351855) polymorphism is a key functional variant associated with cancer progression:
Inhibitor | Mechanism | Clinical Stage | Targeted Cancers |
---|---|---|---|
BLU-9931 | Irreversible FGFR4 inhibition | Preclinical | HCC, cholangiocarcinoma |
FGF401 | Reversible covalent inhibition | Phase I/II (NCT02325739) | HCC, solid tumors |
H3B-6527 | Covalent FGFR4 binding | Phase I (NCT02834780) | HCC, gastric cancer |
BGJ398 | Pan-FGFR inhibitor (FGFR1–4) | Preclinical | CRC, breast cancer |
FGF19-neutralizing antibodies block FGFR4 activation, suppressing HCC and colon cancer growth in preclinical models .
Silencing FGFR4 enhances apoptosis in CRC cells and restores sensitivity to 5-FU/oxaliplatin .
FGFR4 upregulates anti-apoptotic proteins (Bcl-2, c-FLIP) via STAT3 signaling .
FGFR4 is a transmembrane receptor tyrosine kinase that conducts signals from fibroblast growth factors to regulate critical cellular processes. In normal physiology, FGFR4 plays essential roles in:
Cell division, growth regulation, and cellular maturation
Formation of blood vessels (angiogenesis)
Wound healing processes
Embryonic development
Muscle development and maturation of bone cells in the skull
Development and maintenance of specialized foveal cone cells in the retina
FGFR4 conducts signals by interacting with specific growth factors at the cell membrane, transmitting these signals to the nucleus where they influence gene expression. This signaling pathway enables cells to respond appropriately to environmental changes through division, migration, or differentiation into specialized cell types .
FGFR4 is a 110 kDa glycosylated transmembrane receptor tyrosine kinase. The mature human FGFR4 protein consists of:
A 348 amino acid extracellular domain (ECD) containing three immunoglobulin-like domains
A 21 amino acid transmembrane segment
A 412 amino acid cytoplasmic domain housing the tyrosine kinase catalytic region
FGFR4 Domain Structure | Amino Acid Position | Function |
---|---|---|
Extracellular Domain | Leu22-Asp369 | Ligand binding |
Ig-like Domains (three) | Within ECD | Specificity for FGF binding |
Transmembrane Segment | 21 aa length | Membrane anchoring |
Cytoplasmic Domain | 412 aa length | Signal transduction |
Tyrosine Kinase Domain | Within cytoplasmic domain | Catalytic activity |
Alternative splicing generates variant isoforms, including a potentially secreted form with substitutions in the transmembrane segment and a 65 kDa N-terminally truncated isoform lacking the signal peptide and first two Ig-like domains. This truncated form is found in pituitary adenomas and displays constitutive phosphorylation with oncogenic properties .
FGFR4 demonstrates specific expression patterns that vary between developmental stages and adult tissues:
During embryonic development: Widely expressed in multiple developing tissues
In adult tissues: Predominantly expressed in liver, kidney, and lung tissue
In pathological states: Overexpressed in various cancer types compared to normal tissue counterparts, particularly in colon cancer , rhabdomyosarcoma , astrocytoma , and advanced prostate cancer
Methodologically, researchers can assess FGFR4 expression through:
Immunohistochemistry (IHC) for protein-level detection in tissue samples
PCR-Southern blot or qRT-PCR for mRNA transcript detection
Western blot analysis for protein expression in cell or tissue lysates
Public database mining using resources like The Human Protein Atlas for comparative tissue expression data
FGFR4 expression often increases with cancer progression, as demonstrated in several tumor types:
In astrocytoma:
Grade II astrocytomas: FGFR4 negative by immunohistochemistry in all examined cases (0/7)
Grade III astrocytomas: FGFR4 positive in 26.7% of cases (4/15)
Grade IV astrocytomas (glioblastoma): FGFR4 positive in 68.4% of cases (13/19)
This pattern suggests FGFR4 protein expression correlates with malignancy grade, with increased expression in higher-grade tumors. Interestingly, FGFR4 mRNA was detected in all specimens regardless of grade, indicating post-transcriptional regulation might be involved in the differential protein expression observed across tumor grades .
For prostate cancer, immunohistochemical staining analysis has demonstrated that FGFR4 expression is significantly increased in advanced-stage tumors compared to early-stage disease or normal prostatic tissue, suggesting its potential role in disease progression .
Two key polymorphisms in the FGFR4 gene have been extensively studied for their association with cancer susceptibility:
G388R polymorphism (Glycine to Arginine substitution at position 388):
V10I polymorphism (Valine to Isoleucine substitution at position 10):
Bioinformatic analysis using Polyphen2 predicts that the G388R mutation damages FGFR4 protein function, which may explain its clinical associations. Although these polymorphisms produce no apparent effects in healthy individuals, they may accelerate disease progression in cancer patients .
FGFR4 contributes to cancer progression through multiple mechanisms:
Anti-apoptotic signaling:
Chemoresistance:
Prognostic impact:
Cancer-specific mechanisms:
Researchers employ multiple complementary approaches to investigate FGFR4 function:
Genetic manipulation:
RNA interference (RNAi) using short hairpin RNA (shRNA) for FGFR4 silencing
CRISPR-Cas9 genome editing for knockout studies
Overexpression systems using viral vectors
Pharmacological inhibition:
Protein interaction studies:
Signaling pathway analysis:
Western blotting for phosphorylation status
Reporter assays for transcription factor activation
Immunofluorescence for protein localization
A methodologically rigorous approach combines multiple techniques to establish both necessity and sufficiency of FGFR4 in biological processes.
Successful FGFR4 targeting requires consideration of several methodological factors:
Selecting appropriate model systems:
Cell lines with documented FGFR4 expression (colon cancer, rhabdomyosarcoma, hepatocellular carcinoma)
Patient-derived xenografts for in vivo studies
Genetic mouse models with FGFR4 modifications
Validation of targeting efficiency:
Confirming knockdown/knockout at both mRNA and protein levels
Assessing specificity for FGFR4 vs. other FGFR family members
Dose-response relationships for pharmacological inhibitors
Functional readouts:
Cell viability assays (MTT, CellTiter-Glo)
Apoptosis measurements (caspase activation, Annexin V)
Cell cycle analysis
Colony formation for long-term effects
Migration and invasion assays for metastatic potential
Downstream signaling analysis:
STAT3 phosphorylation status
c-FLIP expression
Other pathway components (MAPK, PI3K/AKT)
When designing FGFR4 targeting experiments, researchers should consider targeting not only FGFR4 itself but also its interaction with co-receptors like beta-Klotho, which significantly enhances FGFR4 signaling specificity and capacity .
FGFR4 functions within a complex network of interactions that modulate its activity:
Co-receptor interactions:
Ligand specificity:
FGFR4 binds preferentially to specific FGF family members:
Metabolic regulation:
Methodologically, researchers can investigate these interactions through co-immunoprecipitation, proximity ligation assays, FRET/BRET techniques, and functional studies with selective ligands or co-receptor knockdowns.
Developing selective FGFR4 inhibitors faces several significant challenges:
Structural homology:
High sequence similarity between FGFR family members (FGFR1-4)
Conserved ATP-binding pocket across tyrosine kinases
Difficult to achieve selectivity without off-target effects
Isoform considerations:
Context-dependent functions:
Resistance mechanisms:
Potential compensatory upregulation of other FGFR family members
Activation of alternative signaling pathways
Selection for resistant cellular subpopulations
Researchers should consider combination approaches targeting both FGFR4 and key downstream effectors (like STAT3) or employing synthetic lethality strategies to overcome these challenges.
FGFR4 shows significant potential as a prognostic biomarker in multiple cancer types:
Astrocytoma:
Prostate cancer:
Colorectal cancer:
Implementation methodology should include:
Standardized immunohistochemical staining protocols
Defined scoring systems with validated cutoffs
Integration with other established prognostic markers
Prospective validation in clinical cohorts
Several therapeutic approaches targeting FGFR4 are being investigated:
Small molecule inhibitors:
Biologics:
RNA-based therapeutics:
siRNA/shRNA approaches demonstrated efficacy in preclinical models
Potential for antisense oligonucleotide development
mRNA destabilization strategies
Downstream pathway inhibition:
Patient selection strategies:
FGFR4 consists of three extracellular immunoglobulin-like domains, a single transmembrane helix, and an intracellular tyrosine kinase domain . The extracellular domains are responsible for binding to fibroblast growth factors (FGFs), which are a family of 18 glycoproteins . Upon binding to FGFs, FGFR4 undergoes dimerization and autophosphorylation, leading to the activation of downstream signaling pathways that regulate cellular processes .
The FGFR4 Fc Chimera is a recombinant protein that combines the extracellular domain of FGFR4 with the Fc region of human immunoglobulin G1 (IgG1) . This fusion protein is designed to enhance the stability and solubility of FGFR4, making it suitable for various research applications. The Fc region also allows for easy purification using protein A or G affinity chromatography .
The recombinant FGFR4 Fc Chimera is typically expressed in mammalian cell lines, such as NS0 cells . The protein is then purified using affinity chromatography, followed by size-exclusion chromatography to ensure high purity and homogeneity . The final product is lyophilized and can be reconstituted in phosphate-buffered saline (PBS) for use in experiments .
FGFR4 Fc Chimera is widely used in research to study the role of FGFR4 in various biological processes and diseases. It is particularly useful in investigating the mechanisms of FGFR4 signaling and its involvement in cancer . FGFR4 is upregulated in multiple tumors, including pituitary, breast, pancreatic, hepatocellular, prostate, and gynecological cancers . It functions as an oncogene in breast cancer and is implicated in drug resistance in colorectal cancer . Therefore, FGFR4 Fc Chimera serves as a valuable tool for developing targeted therapies and understanding cancer progression .