PDGFRA encodes a 170 kDa transmembrane protein with three domains:
Extracellular ligand-binding domain (binds PDGF isoforms: A, B, AB, C)
Transmembrane domain
Intracellular tyrosine kinase domain (activates downstream signaling)
Key signaling pathways include PI3K-AKT, MAPK/ERK, and STAT, regulating cell migration, differentiation, and survival .
Embryogenesis: Essential for cephalic closure, gastrointestinal tract development, and mesangial cell differentiation .
Hematopoiesis: Maintains hematopoietic stem cells and eosinophil regulation .
Wound Healing: Promotes fibroblast chemotaxis and tissue repair .
PDGFRA knockout mice exhibit lethal anomalies: skeletal malformations, anemia, and thrombocytopenia .
In humans, PDGFRA mutations are linked to isolated cleft palate (e.g., c.*34G>A in 3′UTR disrupts miR-140 binding) .
Gastrointestinal Stromal Tumors (GISTs): ~10% of GISTs harbor PDGFRA mutations, often mutually exclusive with KIT mutations .
Hypereosinophilic Syndromes: Fusion-driven neoplasms with eosinophilia .
Inflammatory Fibroid Polyps: Benign GI tumors with PDGFRA activation .
Imatinib: First-line tyrosine kinase inhibitor (TKI) for PDGFRA-mutant GISTs, but ineffective against D842V due to disrupted "DFG-out" conformation .
Kinase Dynamics: The D842V mutation increases ATP affinity and stabilizes the active kinase state, reducing drug binding .
Mutation | Effect |
---|---|
D842V | Destabilizes autoinhibited conformation; enhances ATP binding |
T674I | Reduces drug-receptor interaction (analogous to BCR-ABL1 T315I) |
PDGFRA is a receptor tyrosine kinase (RTK) encoded by the PDGFRA gene located on chromosome 4q12. It functions as a cell surface receptor that transmits signals into the cell through signal transduction when platelet-derived growth factor binds to it. This activation triggers phosphorylation of intracellular proteins, initiating multiple signaling pathways that control critical cellular processes including proliferation, differentiation, migration, and survival .
PDGFRA signaling is particularly important for the development of numerous cell types throughout the body. The receptor is expressed in various tissues and plays crucial roles in embryonic development, tissue homeostasis, and wound healing. In the adult organism, PDGFRA continues to function in tissue maintenance and repair processes .
PDGFRA alterations manifest in several human diseases through diverse mechanisms:
Oncogenic mutations: PDGFRA mutations are commonly found in gastrointestinal stromal tumors (GISTs), inflammatory fibroid polyps, and gliomas .
Gene amplifications: PDGFRA gene amplification occurs in approximately 11% of glioblastomas (GBMs), making it the second most frequently amplified receptor tyrosine kinase gene in these tumors after EGFR .
Gene rearrangements: Chromosomal rearrangements involving PDGFRA can cause myeloid neoplasms associated with hypereosinophilia, such as PDGFRA-associated chronic eosinophilic leukemia .
Pediatric high-grade gliomas: PDGFRA alterations have been identified in nearly 15% of pediatric high-grade glioma patients, with significantly elevated PDGFRA expression in tumors harboring PDGFRA mutation or amplification .
For reliable detection of PDGFRA expression in tissues, researchers should consider the following methodological approaches:
Transcriptional analysis:
RT-PCR for targeted gene expression analysis
RNA sequencing for comprehensive transcriptomic profiling
Protein detection:
Immunohistochemistry (IHC) using specific anti-PDGFRA antibodies
Western blotting for protein size and expression level assessment
Validation methods:
Inclusion of appropriate positive and negative controls
Preabsorption tests with recombinant PDGFRA to confirm antibody specificity
When working with mouse models, researchers studying PDGFRA expression may utilize the Pdgfratm11(EGFP)Sor/J mice (PDGFRα-eGFP mice), which express enhanced green fluorescent protein (eGFP) driven by the endogenous Pdgfra promoter, allowing for direct visualization of PDGFRα+ cells .
PDGFRA alterations contribute to glioma development through multiple mechanisms that enhance tumor cell proliferation, survival, and invasiveness:
Genetic alterations:
Gene amplification occurs in 11% of glioblastomas, creating multiple copies of the PDGFRA gene
Intragenic deletions, such as PDGFRAΔ8,9 (deletion of exons 8 and 9), have been identified in 40% of PDGFRA-amplified GBMs
Complex rearrangements between PDGFRA and neighboring genes like KDR can create fusion proteins with constitutive kinase activity
Histological correlations:
PDGFRA amplification significantly correlates with oligodendroglioma histological features (p=0.0075), suggesting a role in determining tumor cell phenotype
Tumors with PDGFRA amplification often display increased PDGFRA and Olig2 immunopositivity
Signal activation:
PDGFRA gene rearrangements create constitutively phosphorylated tyrosine kinases that activate downstream signaling pathways independent of ligand binding
This aberrant activation drives uncontrolled cell proliferation and survival mechanisms in tumor cells
Recent research has identified PDGFRA as a promising therapeutic target for pediatric high-grade gliomas, particularly H3K27M diffuse midline gliomas (DMG), which are aggressive malignant brain tumors with poor prognosis:
Prevalence of alterations:
A multicenter study led by Dr. Mariella Filbin analyzed genomic data from 217 pediatric high-grade glioma samples and identified PDGFRA alterations in nearly 15% of patients
Transcriptomic data revealed significantly elevated PDGFRA expression in tumors with PDGFRA mutation or amplification
Oncogenic role:
Elevated PDGFRA expression has been shown to drive growth of DMG tumors
PDGFRA appears to play a multifaceted role in the pathogenesis of both adult and pediatric high-grade gliomas
Therapeutic implications:
The study provides the first real-world clinical data supporting the use of PDGFRA inhibitors in treating certain pediatric patients with high-grade gliomas
Tyrosine kinase inhibitors targeting PDGF receptors, such as imatinib, have shown preclinical validation for conditions involving PDGFRA alterations
Proper analysis of PDGFRA gene amplification in tumor samples requires a multi-method approach for reliable results:
Quantitative PCR (qPCR):
Design primers specific to different regions of the PDGFRA gene
Use reference genes (e.g., 18S) for normalization
Consider gene amplification when average DNA quantities are greater than 4-fold relative to reference samples
Array comparative genomic hybridization (aCGH):
Provides high-resolution detection of copy number variations
Can identify complex amplicons and rearrangements
Particularly useful for detecting intragenic alterations within PDGFRA
Fluorescence in situ hybridization (FISH):
Offers direct visualization of gene amplification in tissue sections
Enables assessment of tumor heterogeneity
Can detect focal amplifications that might be missed by other methods
Next-generation sequencing (NGS):
Provides comprehensive analysis of genomic alterations
Can detect both amplifications and mutations simultaneously
Allows for analysis of the entire PDGFRA locus and adjacent regions
Data interpretation considerations:
Complex amplicons may involve adjacent genes (e.g., KDR)
Gene rearrangements often accompany amplifications
The pattern of amplification across different exons can suggest potential functional consequences
Several experimental models are available for investigating PDGFRA function, each with specific advantages for different research questions:
Cell culture models:
Primary cells expressing endogenous PDGFRA (e.g., fibroblasts, glial cells)
Cell lines with characterized PDGFRA expression levels
Genetically modified cells with PDGFRA knockdown, knockout, or overexpression
Patient-derived cells harboring specific PDGFRA mutations or amplifications
Animal models:
PDGFRα-eGFP mice (Pdgfratm11(EGFP)Sor/J) that express eGFP in nuclei of PDGFRα+ cells, enabling visualization and isolation of cells expressing PDGFRA
Conditional knockout mice for tissue-specific PDGFRA deletion
Patient-derived xenograft models that maintain the genetic characteristics of human tumors
CRISPR-engineered mice carrying specific PDGFRA mutations found in human diseases
Ex vivo systems:
Organoid cultures that better represent tissue architecture and cellular interactions
Tissue explants maintaining the native microenvironment of PDGFRA-expressing cells
Co-culture systems to study interactions between PDGFRA+ cells and other cell types
Recent research has identified links between PDGFRA expression and cellular senescence, providing methodological approaches for investigating this relationship:
Senescence induction methods:
Irradiation-induced senescence models (particularly effective in nucleus pulposus cells)
Oxidative stress-induced senescence
Replicative senescence through extended passaging
Key findings from recent research:
PDGFRA gene expression is significantly reduced in irradiation-induced senescent nucleus pulposus (NP) cells compared to non-irradiated cells
PDGF-AB/BB treatment can modulate gene expression in senescent cells, affecting clusters related to cell cycle regulation, mesenchymal cell differentiation, and response to reduced oxygen levels
PDGFRA appears to be a top hub gene in treated NP cells according to network analysis
Analytical approaches:
Transcriptomic analysis to identify changes in PDGFRA and related gene expression
Network analysis to determine the position of PDGFRA in senescence-associated gene networks
Functional assays measuring senescence-associated phenotypes (e.g., β-galactosidase activity, cell cycle arrest)
Assessment of senescence-associated secretory phenotype (SASP) in relation to PDGFRA expression
Investigating PDGFRA signaling pathways requires specialized techniques to capture the complex dynamics of receptor activation and downstream effects:
Receptor activation analysis:
Phospho-specific antibodies to detect activated PDGFRA
Immunoprecipitation followed by western blotting for phosphotyrosine detection
Proximity ligation assays to visualize protein-protein interactions in situ
FRET/BRET techniques for real-time monitoring of receptor activation
Downstream signaling assessment:
Phospho-proteomics to identify phosphorylation events triggered by PDGFRA activation
Kinase activity assays for key downstream effectors
Transcriptional profiling to identify genes regulated by PDGFRA signaling
Pathway inhibitors to dissect specific branches of PDGFRA-activated pathways
Functional readouts:
Proliferation assays (e.g., EdU incorporation, Ki67 staining)
Migration assays (e.g., wound healing, transwell)
Survival assays under stress conditions
Differentiation markers to assess cell fate decisions
The relationship between PDGFRA mutations and clinical outcomes varies across different human diseases:
Gastrointestinal stromal tumors (GISTs):
PDGFRA mutations are found in approximately 5-10% of GISTs
D842V mutation in exon 18 is associated with resistance to imatinib therapy
Non-D842V mutations generally confer better prognosis and response to targeted therapies
Glioblastoma (GBM):
PDGFRA amplification occurs in 11% of GBMs, making it the second most frequent RTK gene amplified in these tumors
PDGFRA amplification is associated with oligodendroglioma histological features
The PDGFRAΔ8,9 mutant is found in 40% of PDGFRA-amplified GBMs
Pediatric high-grade gliomas:
PDGFRA alterations are present in nearly 15% of pediatric high-grade glioma patients
Elevated PDGFRA expression is a key driver of diffuse midline glioma (DMG) tumor growth
Recent clinical data supports targeting PDGFRA as a therapeutic approach in these devastating tumors
PDGFRA-associated chronic eosinophilic leukemia:
Often involves FIP1L1-PDGFRA fusion gene resulting from deletion of material from chromosome 4
Patients typically respond well to imatinib therapy
Response to therapy correlates with the specific fusion partner and breakpoint location
Several approaches are being employed to target PDGFRA in cancer therapy, with varying degrees of success:
Tyrosine kinase inhibitors (TKIs):
Imatinib has been preclinically validated for conditions involving PDGFRA alterations
Crenolanib demonstrates activity against the imatinib-resistant D842V mutation
Avapritinib is highly potent against PDGFRA exon 18 mutations
Dasatinib and nilotinib show activity against certain PDGFRA mutants
Resistance mechanisms:
Point mutations in the kinase domain (especially gatekeeper mutations)
Activation of alternative signaling pathways
Increased drug efflux via ABC transporters
Epigenetic alterations affecting drug sensitivity
Emerging therapeutic strategies:
Antibody-drug conjugates targeting PDGFRA
Combination therapies targeting multiple RTKs simultaneously
PROTAC-based approaches for PDGFRA degradation
Immunotherapies directed against PDGFRA-altered cells
Effective clinical trial design for targeting PDGFRA alterations requires careful consideration of several factors:
Patient selection criteria:
Comprehensive molecular profiling to identify specific PDGFRA alterations
Stratification based on mutation type, amplification status, or fusion variants
Consideration of co-occurring genomic alterations that may affect response
Analysis of PDGFRA expression levels in addition to genetic alterations
Biomarker development:
Establish reliable assays for detecting PDGFRA alterations in tissue and liquid biopsies
Develop pharmacodynamic markers of target engagement
Identify predictive biomarkers of response and resistance
Monitor clonal evolution during treatment
Trial design considerations:
Basket trials enrolling patients based on PDGFRA alterations across tumor types
Umbrella trials testing multiple agents against different PDGFRA alterations
Incorporation of adaptive designs to modify treatment based on early response signals
Inclusion of crossover options to maintain patient access to experimental therapies
Several cutting-edge technologies hold promise for advancing PDGFRA research:
Single-cell technologies:
Single-cell RNA sequencing to define heterogeneity in PDGFRA expression
Single-cell proteomics to characterize PDGFRA signaling at the individual cell level
Spatial transcriptomics to map PDGFRA expression within tissue architecture
CRISPR screening at single-cell resolution to identify modifiers of PDGFRA function
Advanced imaging:
Super-resolution microscopy for visualizing PDGFRA distribution and clustering
Live-cell imaging with fluorescent biosensors to track PDGFRA activation in real-time
Intravital microscopy to observe PDGFRA+ cells in their native environment in vivo
Correlative light and electron microscopy to link PDGFRA localization with ultrastructure
Computational approaches:
AI-driven analysis of PDGFRA signaling networks
Systems biology modeling of PDGFRA pathway dynamics
Integration of multi-omics data to comprehensively characterize PDGFRA function
Virtual screening and molecular dynamics simulations for drug discovery
Despite extensive research, several aspects of PDGFRA biology remain underexplored:
Non-canonical functions:
Nuclear localization and potential transcriptional regulation
Ligand-independent signaling mechanisms
Interactions with non-traditional binding partners
Potential roles in cellular metabolism regulation
Developmental roles:
Fine-grained spatiotemporal mapping of PDGFRA expression during embryogenesis
Cell fate decisions influenced by PDGFRA signaling
Interaction with other developmental pathways (e.g., Hedgehog, Wnt)
Role in tissue repair and regeneration in adults
Microenvironmental interactions:
Communication between PDGFRA+ cells and immune components
Role in extracellular matrix remodeling and tissue architecture
Contribution to formation of specialized niches (e.g., perivascular, subepithelial)
Response to mechanical forces and tissue stiffness
Translational opportunities:
Exploration of PDGFRA as a target in diseases beyond cancer
Development of PDGFRA-targeted cellular therapies
Investigation of PDGFRA in aging and age-related diseases
Potential for PDGFRA modulation in regenerative medicine approaches
Platelet-Derived Growth Factor Receptor Alpha (PDGFRA) is a cell surface tyrosine kinase receptor that plays a crucial role in various cellular processes. It is encoded by the PDGFRA gene and is part of the receptor tyrosine kinase (RTK) family, specifically the class III subfamily . PDGFRA is involved in the regulation of cell growth, development, and differentiation, making it a significant focus of research in developmental biology and oncology.
PDGFRA is a type I transmembrane glycoprotein that consists of an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain . The receptor is activated upon binding to its ligands, which are members of the platelet-derived growth factor (PDGF) family. These ligands include PDGF-AA, PDGF-BB, and PDGF-CC . Upon ligand binding, PDGFRA dimerizes, leading to autophosphorylation and activation of its kinase activity. This activation triggers downstream signaling pathways that regulate various cellular functions such as proliferation, migration, and survival .
PDGFRA is integrally involved in embryonic development, angiogenesis, and organogenesis . It induces fibroblast proliferation and migration, which are essential processes in wound healing and tissue repair . PDGFRA is expressed in various cell types, including epithelial cells, muscle cells, osteosarcoma cells, and neuronal progenitor cells . Its role in organ development and wound healing highlights its importance in maintaining tissue homeostasis.
Mutations and aberrant activation of PDGFRA have been implicated in several diseases, including idiopathic hypereosinophilic syndrome, gastrointestinal stromal tumors (GISTs), and other cancers . In GISTs, PDGFRA mutations are often mutually exclusive with KIT mutations and are considered a hallmark of the disease . These mutations can lead to constitutive activation of the receptor, driving uncontrolled cell proliferation and tumor growth. Additionally, PDGFRA gene fusions are associated with eosinophilia and are classified as a distinct disorder by the World Health Organization .
Given its role in various cancers, PDGFRA is a target for therapeutic interventions. Tyrosine kinase inhibitors (TKIs) such as imatinib have been developed to inhibit the activity of PDGFRA and are used in the treatment of GISTs . However, resistance to TKIs can occur due to secondary mutations in the PDGFRA gene. Research is ongoing to develop new therapeutic strategies to overcome resistance and improve patient outcomes.
Recombinant human PDGFRA is produced using recombinant DNA technology, which involves the insertion of the PDGFRA gene into a suitable expression system, such as bacterial or mammalian cells. This allows for the large-scale production of the receptor for research and therapeutic purposes. Recombinant PDGFRA is used in various studies to understand its structure, function, and role in disease, as well as to develop and test new therapeutic agents.