Expression System
Produced in Sf9 cells via baculovirus infection, ensuring proper folding and glycosylation .
Affinity Chromatography: Glutathione agarose binds GST-tagged EGFR .
Proprietary Techniques: Additional chromatography steps refine purity (>90% by SDS-PAGE) .
Component | Details |
---|---|
Buffer | 50 mM HEPES pH 7.5, 100 mM NaCl, 5 mM DTT, 15 mM reduced glutathione, 20% glycerol |
Storage | -20°C to -80°C (stable for 12 months; avoid freeze-thaw cycles) |
Substrate Phosphorylation: PolyEY or peptide substrates (e.g., ERK) are used to measure tyrosine kinase activity .
Inhibitor Screening: Erlotinib, gefitinib, and lapatinib binding kinetics are studied via SPR (surface plasmon resonance) .
Mutant Analysis: T790M/L858R variants (linked to drug resistance) are used to study targeted therapy efficacy .
Signaling Pathways: Activates RAS-RAF-MEK-ERK, PI3K-AKT, and PLCγ-PKC cascades .
Ligand-Specific Signaling
EGF and TGF-α induce distinct EGFR activation profiles:
Ligand | Autophosphorylation | Erk Activation |
---|---|---|
EGF | High | Strong |
TGF-α | Moderate | Weak |
This variation suggests ligand-dependent modulation of EGFR signaling .
Recombinant human EGFR protein expressed in Sf9 cells refers to a functionally active form of the epidermal growth factor receptor produced using a baculovirus expression system in Spodoptera frugiperda (Sf9) insect cells . This typically comprises a protein fragment corresponding to residues 668 to 1210 of human EGFR and often includes an N-terminal GST tag to facilitate purification . The resulting protein retains kinase activity and is capable of autophosphorylation and phosphorylation of appropriate substrates . The baculovirus-Sf9 system provides advantages for obtaining reasonable yields of functionally active protein suitable for enzymatic and structural studies.
The specific activity of EGFR from Sf9 cells is typically measured using peptide kinase assays with defined substrates. According to the reference data, commercially available EGFR protein expressed in this system demonstrates specific activity of approximately 66 nmol/min/mg in kinase assays using Poly(Glu:Tyr, 4:1) as substrate . Activity measurements can also include:
Western blot detection of autophosphorylation at specific tyrosine residues (e.g., Y1068)
Quantification of substrate phosphorylation rates under standardized conditions
Assessment of dose-dependent inhibition profiles using known EGFR inhibitors
These activity measurements provide critical information for determining protein quality and suitability for downstream applications such as inhibitor screening or mechanistic studies.
Human EGFR expressed in Sf9 cells via baculovirus infection exhibits several key structural features:
Feature | Specification | Notes |
---|---|---|
Expression System | Baculovirus-infected Sf9 cells | Insect cell-based expression system |
Purity | >70% by SDS-PAGE | Affinity purified |
Molecular Weight | 89-100 kDa | As determined by SDS-PAGE |
Active Region | Residues 668-1210 | Includes the intracellular kinase domain |
Tag | GST tag (N-terminus) | Facilitates purification |
Contaminants | ~26 kDa GST-binding proteins | Do not affect function |
The protein typically demonstrates enzymatic activity suitable for functional studies and maintains the structural integrity required for kinase activity . Unlike mammalian cell-expressed EGFR, the insect cell-produced protein may have differences in post-translational modifications, particularly glycosylation patterns.
Optimizing expression of active human EGFR in Sf9 cells requires careful attention to several key parameters:
Baculovirus generation and amplification: Following established protocols like the Bac-to-Bac Baculovirus Expression System for generating recombinant baculovirus, with two rounds of amplification prior to protein expression .
Culture conditions: Maintaining Sf9 cells as suspension cultures at 27°C in appropriate insect cell media such as ESF-921 Insect Cell Culture Medium .
Infection parameters: Using optimal virus-to-cell ratios (typically 1:20 v/v) and harvest timing (approximately 72 hours post-infection) .
Stabilizing additives: Addition of 2 μM Erlotinib (an EGFR inhibitor) during expression can improve protein stability and yield by preventing excessive autophosphorylation and subsequent degradation .
Expression enhancers: Supplementation with 8 mM sodium butyrate can significantly enhance recombinant protein expression levels .
This methodological approach has been successfully employed to generate sufficient quantities of active EGFR for both structural and functional studies as described in the research literature.
EGFR expressed in Sf9 cells is suitable for numerous research applications:
Kinase activity assays: For measuring autophosphorylation and substrate phosphorylation, enabling quantitative assessment of enzyme kinetics .
Inhibitor screening and characterization: For evaluating binding and inhibitory properties of small molecules, antibodies, and peptides targeting different conformational states of EGFR .
Mutational analysis: For studying the effects of clinically relevant mutations (e.g., L834R, T766M) on kinase activity, inhibitor sensitivity, and signaling outputs .
Structural studies: For understanding conformational changes upon ligand binding or mutation, particularly when combined with techniques like X-ray crystallography or cryo-EM .
Protein-protein interaction studies: For investigating EGFR heterodimerization with other ErbB family members and interaction with downstream signaling partners .
The recombinant protein's activity and stability make it particularly valuable for biochemical and biophysical studies that require purified, active enzyme.
Different ligands can induce distinct activation patterns in EGFR, as evidenced by comparative studies with EGF and TGF-α:
EGF induces stronger EGFR autophosphorylation than TGF-α when tested with purified EGFR protein and in cellular assays . This differential activation extends to downstream signaling events, with EGF stimulating higher levels of ERK phosphorylation than TGF-α at equivalent concentrations .
The molecular mechanism underlying these differences involves ligand-specific conformational changes that propagate from the extracellular ligand-binding domain to the transmembrane region. Research has demonstrated that:
Mutations disrupting the connection between Domain III and Domain IV (e.g., W492G) eliminate differential activation by EGF versus TGF-α .
Modifications to the linker between the extracellular module and transmembrane helix (e.g., replacing Asn615-Pro620 with a flexible GGSGGS sequence) compromise ligand-specific activation differences .
These findings suggest that different EGFR ligands induce distinct conformational states that differentially regulate kinase activity, providing a molecular basis for ligand-specific cellular responses.
EGFR expressed in Sf9 cells provides a valuable system for studying cancer-associated mutations, particularly those found in lung cancer:
Comparative kinase activity: The system allows direct comparison of wild-type and mutant EGFR kinase activity using purified proteins under controlled conditions .
"Superacceptor" activity analysis: Research has revealed that lung-cancer-associated EGFR mutants (e.g., L834R, T766M) function as "superacceptors" that hyperphosphorylate coexpressed wild-type EGFR or ErbB-2, a phenomenon that can be studied using differentially tagged recombinant proteins .
Directional preference in heterodimerization: Studies with mutated EGFR variants demonstrate specific directional preferences in heterodimer formation, which can be analyzed using strategically designed mutations like I682Q and V924R that preferentially function as donors or acceptors, respectively .
Downstream signaling effects: Co-expression of mutant EGFR with wild-type EGFR or ErbB-2 leads to synergistic enhancement of downstream signaling pathways, particularly ERK activation, which exceeds what would be expected from simple additive effects .
This research approach has revealed that lung cancer-associated EGFR mutations not only increase intrinsic kinase activity but also substantially alter interactions with wild-type receptors, potentially explaining their potent oncogenic effects.
Several experimental approaches can detect conformational changes in EGFR upon ligand binding:
Strategic mutational analysis: Mutations at key coupling points between domains (e.g., W492G between Domain III and Domain IV) can disrupt conformational transmission pathways, allowing researchers to map the structural changes propagating from ligand binding .
Flexible linker insertions: Replacing rigid segments with flexible linkers (e.g., substituting Asn615-Pro620 with GGSGGS) or inserting additional flexible segments (e.g., GGSGGSGGS between Pro617 and Lys618) to disrupt conformational coupling provides insights into how structural changes propagate through the receptor .
Differential activation readouts: Monitoring differences in autophosphorylation patterns and downstream signaling (particularly ERK phosphorylation) between different ligands allows indirect assessment of conformational states .
Structural biology techniques: Although not explicitly mentioned in the search results, complementary approaches like hydrogen-deuterium exchange mass spectrometry, FRET-based sensors, and electron microscopy can provide direct visualization of conformational states.
These approaches have revealed that EGFR signaling involves precisely coordinated conformational changes that couple ligand binding to kinase activation, with different ligands inducing distinct conformational states that result in different signaling outputs.
Several sophisticated strategies have been developed to study EGFR heterodimerization with other ErbB family members, particularly ErbB-2:
Co-expression with size differentiation: Expressing differentially sized constructs (e.g., full-length EGFR with truncated ErbB-2 or vice versa) allows separation by SDS-PAGE to independently assess expression levels and phosphorylation status of each protein .
Directional preference analysis: Using mutations that bias receptors toward donor (I682Q) or acceptor (V924R) roles in the asymmetric dimer enables investigation of directional preferences in heterodimerization .
Cross-phosphorylation assessment: Analyzing how wild-type or mutant EGFR variants affect phosphorylation of co-expressed ErbB-2 provides insights into trans-activation mechanisms .
Inhibitor response profiling: Examining how specific inhibitors (e.g., WZ-4002 for T766M mutations) affect heterodimer signaling provides mechanistic insights and potential therapeutic strategies .
Downstream pathway analysis: Monitoring activation of multiple downstream pathways (ERK, Akt, STAT3) reveals how heterodimers influence signaling specificity and intensity .
These approaches have demonstrated that lung cancer-associated EGFR mutations not only affect homodimerization but also dramatically alter heterodimerization with ErbB-2, potentially contributing to their oncogenic potency.
Sf9-expressed EGFR provides a valuable platform for developing conformation-specific inhibitors with potential advantages over conventional inhibitors:
Targeting specific conformational states: Rather than completely inhibiting EGFR activity, antibodies or peptides that bind specific regions (e.g., Domain IV legs) could stabilize conformations that permit dimerization but reduce signaling intensity .
Overcoming resistance mechanisms: This approach might circumvent resistance mechanisms that emerge in response to conventional inhibitors that completely block EGFR signaling, as partial modulation might be less likely to trigger compensatory adaptations .
Structure-guided design: The detailed structural understanding of EGFR activation mechanisms enables rational design of molecules targeting specific interfaces or conformational states .
Differential ligand responses: Understanding how different ligands induce distinct conformational states provides opportunities to develop inhibitors that selectively block responses to specific ligands .
This research direction represents a paradigm shift from simply inhibiting kinase activity toward more nuanced approaches that modulate receptor conformation and signaling intensity, potentially leading to more effective and resistance-resistant therapeutic strategies.
When assessing EGFR inhibitors using Sf9-expressed protein, several experimental design considerations are crucial:
Protein quality and activity: Ensuring consistent specific activity (e.g., 66 nmol/min/mg) across batches is essential for reliable inhibitor assessment .
Assay conditions optimization: Developing standardized conditions for buffer composition, substrate concentration, ATP concentration, and reaction time that provide an appropriate dynamic range.
Conformational state consideration: Since inhibitors may preferentially target specific conformational states, understanding which states are favored by the recombinant protein is critical .
Validation in cellular contexts: Confirming findings from purified protein assays in cellular systems expressing full-length EGFR to account for cellular factors affecting inhibitor efficacy .
Mutation-specific effects: For inhibitors targeting mutant EGFR, evaluating effects on both the target mutant and potentially co-expressed wild-type EGFR is essential given the "superacceptor" activity of certain mutants .
These considerations help ensure that findings with Sf9-expressed EGFR translate effectively to more complex cellular and in vivo contexts, ultimately improving the predictive value of in vitro inhibitor screening.
While the search results don't explicitly address this question, significant differences in post-translational modifications exist between Sf9-expressed and mammalian-expressed EGFR that researchers should consider:
Glycosylation patterns: Sf9 cells produce simpler, high-mannose type N-glycosylation patterns compared to the complex N-linked glycosylation with terminal sialic acids found in mammalian cells.
Phosphorylation states: Basal phosphorylation patterns differ due to the distinct kinase and phosphatase environments in insect versus mammalian cells.
Lipid modifications: Differences may exist in lipid modifications that affect membrane association and protein-protein interactions.
These differences should be considered when interpreting results, particularly for studies involving:
Antibody recognition of conformational epitopes
Protein-protein interactions influenced by glycosylation
Membrane association properties
Stability and solubility characteristics
For applications where mammalian-type modifications are critical, researchers might consider alternative expression systems or engineered Sf9 cells with humanized post-translational modification pathways.
Sf9-expressed EGFR preparations typically contain endogenous insect GST-binding proteins (~26 kDa) as contaminants . While these do not affect function, researchers can implement several strategies to minimize their impact:
Optimized purification protocols: Sequential chromatography steps including ion exchange after initial affinity purification can reduce contaminants.
Alternative tags: Using tags other than GST (e.g., His, FLAG, or Strep) may avoid co-purification of GST-binding proteins.
Size exclusion chromatography: As a final polishing step to separate EGFR (89-100 kDa) from smaller contaminants.
Control experiments: Including appropriate negative controls to distinguish between effects due to EGFR versus potential contaminants.
Western blot analysis: Using EGFR-specific antibodies for detection in functional studies rather than relying solely on total protein staining.
These approaches can help ensure experimental results reflect EGFR-specific activities rather than effects from contaminating proteins, particularly for sensitive applications like high-throughput screening or structural studies.
The Epidermal Growth Factor Receptor (EGFR) is a critical component in the regulation of cell growth, survival, proliferation, and differentiation. It is a member of the ErbB family of receptor tyrosine kinases, which includes four closely related receptors: EGFR (ErbB1), ErbB2 (HER2/neu), ErbB3 (HER3), and ErbB4 (HER4) . These receptors are involved in various cellular processes and have significant implications in cancer biology when their expression or activity is dysregulated .
EGFR is a transmembrane glycoprotein with an extracellular ligand-binding domain, a single hydrophobic transmembrane segment, and an intracellular tyrosine kinase domain . The receptor is activated upon binding to specific ligands such as epidermal growth factor (EGF), transforming growth factor-alpha (TGF-α), and others . Ligand binding induces receptor dimerization, either as homodimers or heterodimers with other ErbB family members, leading to autophosphorylation of tyrosine residues in the cytoplasmic domain . This autophosphorylation triggers a cascade of downstream signaling pathways, including the MAPK, PI3K/AKT, and JAK/STAT pathways, which regulate various cellular responses .
The recombinant expression of human EGFR in Sf9 cells (a cell line derived from the fall armyworm Spodoptera frugiperda) is a common method for producing active EGFR protein for research purposes . The baculovirus expression system used in Sf9 cells allows for high-level expression of recombinant proteins with proper post-translational modifications, which are essential for the biological activity of EGFR . The recombinant EGFR expressed in Sf9 cells is often fused with tags such as GST (Glutathione S-transferase) to facilitate purification and detection .
Recombinant EGFR is widely used in biochemical and structural studies to understand its function and regulation. It is also employed in drug discovery and development, particularly in screening for inhibitors that can block EGFR signaling in cancer cells . The availability of active recombinant EGFR enables researchers to study the receptor’s interactions with ligands, other proteins, and small-molecule inhibitors in a controlled environment .