EGFR Human Sf9

Epidermal Growth Factor Receptor Sf9 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to EGFR Human Sf9

EGFR Human Sf9 refers to recombinant human epidermal growth factor receptor (EGFR) expressed in Spodoptera frugiperda (Sf9) insect cells. This system produces EGFR fragments spanning amino acids 672–1210, which include the kinase domain and cytoplasmic tail . The protein is typically fused with a glutathione S-transferase (GST) tag at the N-terminus to facilitate purification via affinity chromatography . Key applications include studying kinase activity, receptor-ligand interactions, and the effects of oncogenic mutations .

Key Features

PropertyDescriptionSource
Amino Acid Range672–1210 (cytoplasmic domain, including kinase and C-terminal tail)
Molecular Weight~89,171 Da (GST-tagged)
PurificationProprietary chromatographic techniques (e.g., GST affinity)
Specific Activity30 pmol/µg·min (kinase activity measured via ATP/NADH coupled assay)
Purity≥50% (suitable for SDS-PAGE and functional studies)

Key Applications:

  • Kinase Activity Assays: Measures ATP-dependent phosphorylation of tyrosine residues .

  • Inhibitor Screening: Tests drug efficacy against wild-type (WT) and mutant EGFR variants .

  • Mutation Analysis: Studies structural and functional impacts of oncogenic mutations (e.g., T790M, L858R) .

Functional and Mechanistic Studies

EGFR Human Sf9 is critical for elucidating receptor signaling and drug resistance mechanisms:

  1. Ligand-Dependent Activation:

    • EGF vs. TGF-α: EGF induces stronger EGFR autophosphorylation and downstream ERK activation than TGF-α in cells expressing full-length EGFR .

    • Structural Coupling: Ligand binding modulates extracellular domain conformation, transmitting signals to the kinase domain via Domain IV and transmembrane helices .

  2. Oncogenic Mutants:

    • T790M/L858R: Confers resistance to first-generation EGFR inhibitors (e.g., gefitinib/erlotinib) while maintaining sensitivity to newer inhibitors like TAK-285 .

    • Exon20 Insertions (e.g., V769_D770insASV, D770_N771insSVD): Alters ATP-binding pocket geometry, reducing inhibitor affinity .

Comparison of Key Mutants

MutationImpact on Drug SensitivityKinase ActivityReferences
T790MConfers resistance to gefitinib/erlotinibReduced inhibitor binding affinity
L858REnhances sensitivity to gefitinib/erlotinibHyperactive kinase signaling
L858R/T790MDual resistance to first-/second-gen inhibitorsCompromised ATP-binding pocket
V769_D770insASVReduced affinity for ATP/substrateAltered Km (500 nM ATP)
D770_N771insSVDHyperactive kinase activityElevated kcat (1.2 µM ATP)

Kinetic Data for EGFR Variants

EGFR VariantKm (ATP, µM)kcat (min⁻¹)Specific Activity (pmol/µg·min)Source
WTNot reportedNot reported30 (assumed from )
L858R30 nMNot reportedNot reported
L858R/T790M60 nMNot reportedNot reported
V769_D770insASV500 nMNot reportedNot reported

Note: Kinetic parameters vary based on assay conditions and substrate specificity. WT EGFR specific activity is inferred from GST-tagged constructs .

Ligand-Specific Signaling

PropertyEGF-Stimulated EGFRTGF-α-Stimulated EGFRReferences
AutophosphorylationHighModerate
ERK PhosphorylationStrongWeak
Structural CouplingDomain IV leg stabilizationDomain IV leg flexibility

Key Insight: EGF stabilizes the extracellular domain’s Domain IV leg, enhancing transmembrane helix proximity and kinase activation .

Common Applications

  1. Kinase Activity Assays:

    • Substrate: Poly-[Glu⁴Tyr] peptide.

    • Detection: ATP/NADH coupled system (A340 absorbance) .

  2. Inhibitor Screening:

    • Irreversible Inhibitors: Poziotinib for active enzyme quantification .

    • Reversible Inhibitors: TAK-285 and neratinib for mutant EGFR binding studies .

  3. Purification:

    • Affinity Chromatography: GST-tag capture followed by thrombin cleavage .

Critical Factors in EGFR Human Sf9 Studies

FactorImpact on ExperimentsMitigation Strategy
Protein StabilityLoss of kinase activity upon freeze-thaw cyclesStore at -20°C to -80°C
Phosphorylation BiasAutophosphorylation in Sf9 lysates may skew dataUse reducing agents (e.g., DTT)
Mutation ArtifactsN-terminal GST-tag may alter kinase domain conformationValidate with full-length EGFR

Product Specs

Introduction

The epidermal growth factor receptor (EGFR) subfamily of receptor tyrosine kinases consists of four members: EGFR (also called HER1, ErbB1, or ErbB), ErbB2 (Neu, HER2), ErbB3 (HER3), and ErbB4 (HER4). These type I transmembrane glycoproteins share a structure that includes an extracellular domain with two cysteine-rich domains (responsible for ligand binding) separated by a spacer region, and a cytoplasmic domain containing a membrane-proximal tyrosine kinase domain and a C-terminal tail with multiple tyrosine autophosphorylation sites. The human EGFR gene encodes a precursor protein of 1210 amino acids (aa), comprising a 24 aa signal peptide, a 621 aa extracellular domain, a 23 aa transmembrane domain, and a 542 aa cytoplasmic domain. EGFR binds to several ligands of the EGF family, including EGF, amphiregulin, TGF-alpha, betacellulin, epiregulin, HB-EGF, and neuregulin-2, without requiring a co-receptor. Ligand binding triggers EGFR homodimerization and heterodimerization with ErbB2, leading to kinase activation, tyrosine phosphorylation, and downstream cell signaling. EGFR can also heterodimerize with ligand-activated ErbB3 or ErbB4. EGFR signaling is involved in regulating various biological processes, such as cell proliferation, differentiation, motility, and apoptosis, and plays a role in carcinogenesis.

Description
This product consists of the extracellular domain of the human EGFR (amino acids 25-647), excluding the signal peptide, which is cleaved during protein production in insect cells. The resulting protein has an approximate molecular weight of 85 kDa. A Strep-tag is fused to the C-terminus of the EGFR for purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
ErbB1 was lyophilized from a sterile solution at a concentration of 1 mg/ml in 1x PBS with a pH of 7.4.
Solubility
Reconstitute the lyophilized EGFR in sterile PBS at a concentration of at least 100 µg/ml. The reconstituted protein can be further diluted in other aqueous solutions.
Stability
Lyophilized EGFR is stable at room temperature for up to 3 weeks but should be stored desiccated below -18°C. After reconstitution, store EGFR at 4°C for 2-7 days. For long-term storage, freeze at -18°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms
Epidermal growth factor receptor, EC 2.7.10.1, Receptor tyrosine-protein kinase ErbB-1, ERBB, mENA, ERBB1, EGFR.
Source
Sf9 Insect Cells.
Amino Acid Sequence
LEEKKV CQGTSNKLTQ LGTFEDHFLS LQRMFNNCEV VLGNLEITYV QRNYDLSFLK TIQEVAGYVL IALNTVERIP LENLQIIRGN MYYENSYALA VLSNYDANKT GLKELPMRNL QEILHGAVRF SNNPALCNVE SIQWRDIVSS DFLSNMSMDF QNHLGSCQKC DPSCPNGSCW GAGEENCQKL TKIICAQQCS GRCRGKSPSD CCHNQCAAGC TGPRESDCLV CRKFRDEATC KDTCPPLMLY NPTTYQMDVN PEGKYSFGAT CVKKCPRNYV VTDHGSCVRA CGADSYEMEE DGVRKCKKCE GPCRKVCNGI GIGEFKDSLS INATNIKHFK NCTSISGDLH ILPVAFRGDS FTHTPPLDPQ ELDILKTVKE ITGFLLIQAW PENRTDLHAF ENLEIIRGRT KQHGQFSLAV VSLNITSLGL RSLKEISDGD VIISGNKNLC YANTINWKKL FGTSGQKTKI ISNRGENSCK ATGQVCHALC SPEGCWGPEP RDCVSCRNVS RGRECVDKCK LLEGEPREFV ENSECIQCHP ECLPQAMNIT CTGRGPDNCI QCAHYIDGPH CVKTCPAGVM GENNTLVWKY ADAGHVCHLC HPNCTYGCTG PGLEGCPTNG PKIPSIAASW SHPQFEK.

Q&A

What is EGFR and why is it significant in research?

EGFR (Epidermal Growth Factor Receptor), also known as ERBB, ERBB1, or HER1, is a type I transmembrane protein belonging to the tyrosine protein kinase family. It belongs to a family of receptors including HER2, HER3, and HER4 that regulate cell growth and differentiation . EGFR signaling plays crucial roles in cell proliferation, differentiation, motility, and apoptosis. This receptor is frequently dysregulated in numerous cancers, particularly epithelial solid tumors, making it an excellent pharmacological target for cancer treatment .

The receptor binds ligands including EGF, TGF-α, amphiregulin, betacellulin, epiregulin, heparin-binding EGF, and neuregulin-2 . When ligands bind, EGFR undergoes homo- or heterodimerization with other ErbB family members, triggering autophosphorylation and downstream signaling pathways through adapter proteins like GRB2 .

What is the molecular structure of human EGFR?

Human EGFR is a 1210 amino acid residue precursor with a complex multi-domain structure consisting of:

  • A 24-amino acid signal peptide

  • A 621-amino acid extracellular domain containing two cysteine-rich domains separated by a spacer region involved in ligand binding

  • A 23-amino acid transmembrane domain

  • A 542-amino acid cytoplasmic domain containing a membrane-proximal tyrosine kinase domain and a C-terminal tail with multiple tyrosine autophosphorylation sites

This complex architecture allows EGFR to receive extracellular cues through ligand binding and transmit signals into the cell through its kinase activity, culminating in the activation of various signaling cascades that regulate cellular processes .

Why are Sf9 cells used for EGFR expression in research?

Sf9 (Spodoptera frugiperda) insect cells are commonly used for recombinant EGFR expression because they provide several advantages over other expression systems:

  • They can produce high yields of properly folded, active proteins with post-translational modifications

  • The baculovirus expression system allows for efficient expression of large mammalian proteins

  • EGFR kinase domains expressed in Sf9 cells retain their catalytic activity, making them suitable for functional studies

  • The system is scalable for producing larger quantities needed for structural and biochemical analyses

Most recombinant EGFR proteins produced in Sf9 cells focus on the intracellular kinase domain (typically amino acids 668-1210), which is the region most relevant for studying enzyme activity, inhibitor binding, and oncogenic mutations .

What are the optimal strategies for expressing EGFR kinase domains in Sf9 cells?

For optimal expression of EGFR kinase domains in Sf9 cells, researchers should consider the following strategies:

  • Construct design: Most successful EGFR constructs include amino acids 668-1210 or 672-1210, which encompass the complete intracellular domain with the tyrosine kinase region . Include an appropriate fusion tag (GST or His) for purification, with a cleavage site if tag removal is desired.

  • Baculovirus infection conditions: Optimize the multiplicity of infection (MOI) and harvest timing (typically 48-72 hours post-infection) to maximize protein yield while maintaining quality.

  • Cell culture parameters: Maintain Sf9 cells in exponential growth phase prior to infection and use high-quality culture media to ensure optimal expression.

  • Temperature considerations: Expression at lower temperatures (24-27°C) can enhance proper folding of kinase domains compared to higher temperatures.

  • Supplementation: Adding protease inhibitors during cell harvest and lysis can prevent degradation of the expressed protein.

For expression of EGFR mutants (such as T790M, L858R, or C797S), the same basic approaches apply, but careful optimization may be required for each specific variant to account for potential differences in stability or expression efficiency .

How can I assess EGFR expression success before proceeding to full purification?

Before investing time and resources in large-scale purification, researchers can assess EGFR expression success using several complementary approaches:

  • Small-scale expression analysis:

    • Collect a small sample (1-5 ml) of infected cells

    • Lyse cells and analyze total protein content by SDS-PAGE

    • Look for a prominent band at the expected molecular weight (approximately 60-80 kDa for the kinase domain plus fusion tag)

    • Compare with uninfected cells to identify overexpressed protein

  • Western blotting:

    • Use antibodies against EGFR or the fusion tag to confirm identity

    • Anti-phosphotyrosine antibodies can indicate autophosphorylation status, suggesting functional activity

  • Small-scale affinity purification:

    • Perform a miniature version of the purification protocol using a small volume of cell lysate

    • Analyze bound and eluted fractions to assess binding efficiency and purity

    • Estimate potential yield based on this small-scale test

  • Activity assays:

    • Crude lysates can be tested for kinase activity using simple ATP consumption assays

    • Compare with positive controls to estimate relative activity levels

These approaches provide valuable information about expression levels, protein solubility, and preliminary activity, allowing researchers to optimize conditions before scaling up .

What purification approach yields the highest quality EGFR kinase domain?

A multi-step purification strategy typically yields the highest quality EGFR kinase domain preparations:

  • Affinity chromatography (first step):

    • For GST-tagged EGFR: Use glutathione Sepharose resin

    • For His-tagged EGFR: Use nickel or cobalt affinity resin

    • Include extensive washing steps to remove non-specifically bound proteins

    • Elute with appropriate buffer (glutathione for GST-tag, imidazole for His-tag)

  • Ion exchange chromatography (second step):

    • Separates proteins based on charge differences

    • Removes contaminating proteins and nucleic acids

  • Size exclusion chromatography (final step):

    • Resolves based on molecular size

    • Provides buffer exchange and final polishing

    • Separates monomeric protein from aggregates

  • Tag removal (optional):

    • If the fusion tag might interfere with downstream applications, remove it using the appropriate protease (e.g., thrombin)

    • Perform a second affinity step to separate cleaved protein from the tag

This approach can yield EGFR preparations with ≥70% purity as mentioned in the product specifications, which is sufficient for most biochemical and structural studies .

How do I troubleshoot common issues in EGFR purification from Sf9 cells?

IssuePossible CausesSolutions
Low expression yieldPoor viral titer; Suboptimal infection conditions; Cell health issuesRe-titer virus stock; Optimize MOI (multiplicity of infection); Use healthy cells in log phase; Determine optimal harvest time through time-course experiments
Poor protein solubilityImproper folding; Aggregation; Membrane associationLower expression temperature; Optimize lysis buffer composition (add glycerol, detergents); Include reducing agents; Use mild detergents like CHAPS or NP-40
Low purity after affinity stepNon-specific binding; Insufficient washing; Protein degradationIncrease wash stringency; Add low imidazole concentration in wash buffer for His-tagged proteins; Include protease inhibitors; Perform purification at 4°C
Loss of kinase activityImproper folding; Loss of cofactors; Oxidation; AutoinhibitionAdd Mg²⁺ to all buffers (5-10 mM); Include reducing agents (DTT or β-mercaptoethanol); Minimize purification time; Store with ATP or stabilizing agents
Protein instability during storageFreeze-thaw damage; Aggregation; Proteolytic degradationAdd 10-20% glycerol to storage buffer; Make small aliquots to avoid repeated freeze-thaw; Store at -80°C; Add protease inhibitors before storage

Systematic troubleshooting using this approach can help resolve most common issues encountered during EGFR expression and purification from Sf9 cells .

What approaches can accurately measure EGFR kinase activity in vitro?

Several complementary approaches can be used to accurately measure EGFR kinase activity in vitro:

  • Radiometric assays:

    • Use [γ-³²P]ATP to measure phosphate incorporation into peptide substrates

    • Quantify activity by scintillation counting or phosphorimaging

    • Provides highly sensitive, direct measurement of kinase activity

  • Non-radiometric assays:

    • ELISA-based methods using phospho-specific antibodies

    • Fluorescence-based assays using phospho-sensing dyes

    • ADP-Glo™ or similar assays that measure ATP consumption or ADP production

  • Autophosphorylation analysis:

    • Western blotting with phospho-specific antibodies targeting key EGFR autophosphorylation sites

    • Time-course experiments to determine rates of autophosphorylation

    • Mass spectrometry to identify all phosphorylation sites

  • Real-time monitoring:

    • Fluorescence resonance energy transfer (FRET)-based sensors

    • Surface plasmon resonance (SPR) to monitor substrate binding and modification

For accurate kinetic analysis, researchers should determine key parameters including Km and kcat for ATP and peptide substrates, and compare these values between wild-type and mutant EGFR variants to understand how mutations impact catalytic efficiency .

How do cancer-associated mutations affect EGFR activation mechanisms?

Cancer-associated mutations dramatically alter EGFR activation mechanisms through several mechanisms:

  • "Superacceptor" activity: Mutated EGFRs found in lung cancer (like L858R and T790M) preferentially assume the "acceptor" or "receiver" position in asymmetric dimers when coexpressed with wild-type EGFR. This leads to enhanced association with wild-type EGFR and hyperphosphorylation of the wild-type counterpart .

  • Altered dimerization dynamics: The crystal structure of the L834R/T766M (L858R/T790M in alternate numbering) mutant reveals an asymmetric dimer interface similar to wild-type EGFR, but with altered preferences for the acceptor role that changes signaling dynamics .

  • Cross-activation of other ErbB receptors: Mutated EGFRs can hyperphosphorylate related family members, such as ErbB-2 (HER2), expanding their oncogenic signaling network .

  • Drug resistance mechanisms: Mutations like T790M confer resistance to first-generation EGFR tyrosine kinase inhibitors, while the C797S mutation creates resistance to third-generation inhibitors, necessitating ongoing drug development efforts .

These findings have significant therapeutic implications, especially considering that wild-type EGFR alleles are typically preserved within EGFR-mutant tumor cells, creating heterogeneous receptor populations with complex interactions .

How should researchers design experiments to compare wild-type and mutant EGFR activity?

For robust comparative analysis of wild-type and mutant EGFR activity, researchers should implement a multi-faceted experimental design:

  • Standardized expression and purification:

    • Use identical expression constructs (except for the specific mutations)

    • Apply consistent purification protocols for all variants

    • Verify comparable purity (≥70%) by SDS-PAGE

    • Normalize protein concentrations precisely

  • Comprehensive kinetic characterization:

    • Determine Km and kcat values for ATP and peptide substrates

    • Calculate catalytic efficiency (kcat/Km) for each variant

    • Generate Michaelis-Menten plots for visual comparison

  • Inhibitor profiling:

    • Test sensitivity to a panel of EGFR inhibitors

    • Determine IC50 values for first, second, and third-generation inhibitors

    • Calculate resistance factors (RF = IC50mutant/IC50wild-type)

  • Structural analysis:

    • When possible, obtain crystal structures of both wild-type and mutant proteins

    • Compare key structural features, especially around the active site and dimer interface

    • Use molecular dynamics simulations to predict structural differences

  • Dimerization assays:

    • Study the "superacceptor" activity by mixing wild-type and mutant proteins

    • Use differentially tagged constructs to track directional phosphorylation

    • Analyze preferences for "donor" versus "acceptor" roles in asymmetric dimers

This comprehensive approach enables researchers to fully characterize how mutations affect EGFR function at the molecular level, providing insights for therapeutic development .

How can EGFR asymmetric dimerization be studied using Sf9-expressed proteins?

The asymmetric dimerization mechanism of EGFR can be studied using Sf9-expressed proteins through several sophisticated approaches:

  • Biochemical reconstitution experiments:

    • Express and purify wild-type and mutant EGFR kinase domains separately

    • Mix proteins in controlled ratios to assess dimerization preferences

    • Measure kinase activity to detect activation patterns

    • Use crosslinking methods to capture transient dimeric structures

  • Structural analysis:

    • X-ray crystallography of kinase domain dimers (as reported for the L834R/T766M mutant at 4-Å resolution)

    • Small-angle X-ray scattering (SAXS) for solution-state dimer characterization

    • Cryo-electron microscopy for visualizing larger assemblies

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

  • Mutational analysis of interface residues:

    • Create "donor-impaired" and "acceptor-impaired" mutants by targeting key interface residues

    • Test combinations of these mutants with wild-type or cancer-associated mutants

    • Analyze how interface mutations affect the "superacceptor" phenomenon observed with cancer mutants

  • Biophysical characterization:

    • Determine binding affinities between different EGFR variants

    • Measure the thermodynamics and kinetics of dimer formation

    • Use analytical ultracentrifugation to quantify monomer-dimer equilibrium

These approaches collectively provide insights into how EGFR dimerization contributes to normal signaling and how cancer-associated mutations alter this process, potentially revealing new therapeutic opportunities .

What insights do recent structural studies provide about EGFR mutant activation?

Recent structural studies, particularly the crystal structure of the L834R/T766M (L858R/T790M in alternate numbering) mutant EGFR, have provided several key insights:

  • The structure reveals an asymmetric dimer interface that is essentially the same as in wild-type EGFR, indicating that the basic dimerization mechanism is preserved in mutant forms .

  • Despite similar dimer structures, mutant EGFRs show a strong preference for assuming the "acceptor" position in heterodimers with wild-type EGFR, creating a directional "superacceptor" effect .

  • This superacceptor activity results in enhanced association between mutant and wild-type EGFR, leading to hyperphosphorylation of the wild-type counterpart .

  • The findings suggest that heterodimers between mutant and wild-type EGFR molecules likely play a significant role in oncogenic signaling, a factor previously underappreciated in EGFR biology .

  • The structural insights explain why certain mutations confer resistance to specific inhibitors while maintaining sensitivity to others, based on subtle changes in the ATP-binding pocket structure .

These structural insights are particularly important considering that in almost all cases, wild-type EGFR alleles are preserved within EGFR-mutant tumor cells, creating mixed populations where these interactions can occur .

What are the best approaches for studying EGFR inhibitor resistance mechanisms?

To effectively study EGFR inhibitor resistance mechanisms, researchers should employ a comprehensive set of approaches:

  • Structural biology:

    • Obtain crystal structures of resistant mutants (e.g., T790M, C797S) bound to various inhibitors

    • Compare with structures of wild-type EGFR to identify critical binding differences

    • Use molecular dynamics simulations to understand inhibitor binding dynamics

  • Biochemical profiling:

    • Express and purify the full panel of clinically relevant EGFR mutants in Sf9 cells

    • Determine IC50 values for each mutant against all generations of inhibitors

    • Create comprehensive inhibition profiles across diverse mutation types

  • Resistance mutation modeling:

    • Introduce mutations into EGFR expression constructs corresponding to those observed in resistant tumors

    • Create double and triple mutants that emerge during sequential therapy

    • Test compound resistance mutations against combination therapies

  • Heterodimer-specific approaches:

    • Study how the "superacceptor" activity of mutant EGFR affects inhibitor sensitivity

    • Investigate whether heterodimers with wild-type EGFR or other ErbB family members contribute to resistance

    • Design assays that can distinguish between effects on monomers, homodimers, and heterodimers

  • Next-generation inhibitor development:

    • Use structural insights to design compounds that can overcome specific resistance mechanisms

    • Test allosteric inhibitors that target sites outside the ATP-binding pocket

    • Explore covalent inhibitors that form irreversible bonds with specific residues

This multi-pronged approach leverages the advantages of Sf9-expressed EGFR to systematically characterize resistance mechanisms and develop strategies to overcome them .

How can Sf9-expressed EGFR be used for drug discovery applications?

Sf9-expressed EGFR provides an excellent platform for drug discovery applications through several approaches:

  • High-throughput inhibitor screening:

    • Use purified EGFR kinase domains for biochemical screening assays

    • Screen compound libraries against both wild-type and mutant variants

    • Identify compounds with selective activity against specific mutations

  • Structure-based drug design:

    • Obtain co-crystal structures of EGFR kinase domains with lead compounds

    • Use structural insights to guide medicinal chemistry optimization

    • Design compounds targeting specific conformations or binding sites

  • Resistance profiling:

    • Test drug candidates against panels of clinically relevant EGFR mutants

    • Identify compounds effective against resistance mutations like T790M and C797S

    • Develop mutation-agnostic inhibition strategies

  • Allosteric modulator discovery:

    • Design assays to identify compounds that bind outside the ATP-binding site

    • Discover molecules that modulate dimerization or conformational dynamics

    • Develop compounds that specifically target the asymmetric dimer interface

  • Fragment-based approaches:

    • Screen fragment libraries against EGFR using biophysical methods

    • Identify novel chemical starting points for inhibitor development

    • Build towards larger compounds with optimized properties

Using these approaches, researchers can leverage the high quality and functional activity of Sf9-expressed EGFR to develop next-generation inhibitors that overcome resistance mechanisms and provide more effective targeted therapies .

What biophysical techniques are most informative for studying EGFR structural dynamics?

Several biophysical techniques provide valuable insights into EGFR structural dynamics:

These techniques collectively provide a comprehensive view of how EGFR structure and dynamics relate to function, particularly how cancer mutations and drug resistance mechanisms alter these properties at the molecular level .

Product Science Overview

Introduction

The Epidermal Growth Factor Receptor (EGFR) is a transmembrane protein that plays a crucial role in the regulation of cell growth, survival, proliferation, and differentiation. It is a member of the ErbB family of receptors, which includes four closely related receptors: EGFR (ErbB1), HER2 (ErbB2), HER3 (ErbB3), and HER4 (ErbB4). These receptors are involved in various cellular processes and are significant targets in cancer therapy due to their role in the development and progression of tumors.

Structure and Function

EGFR consists of an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain. Upon binding with its specific ligands, such as epidermal growth factor (EGF) or transforming growth factor-alpha (TGF-α), EGFR undergoes dimerization and autophosphorylation on specific tyrosine residues within the intracellular domain. This activation triggers a cascade of downstream signaling pathways, including the RAS-RAF-MEK-ERK, PI3K-AKT, PLCγ-PKC, and STAT pathways, which regulate various cellular functions .

Recombinant Expression in Sf9 Cells

The recombinant expression of human EGFR in Sf9 cells, a cell line derived from the fall armyworm Spodoptera frugiperda, is a common method used to produce large quantities of the protein for research and therapeutic purposes. Sf9 cells are widely used in the baculovirus expression system due to their high capacity for protein production and proper post-translational modifications.

Purification and Characterization

The purification of recombinant EGFR typically involves several chromatographic techniques to ensure high purity and activity of the protein. Common methods include affinity chromatography, ion-exchange chromatography, and size-exclusion chromatography. The characterization of the purified protein involves assessing its purity, molecular weight, and biological activity through various biochemical and biophysical assays .

Applications in Research and Medicine

Recombinant EGFR is extensively used in research to study its structure, function, and role in disease. It is also a critical component in the development of targeted cancer therapies. Inhibitors of EGFR, such as erlotinib and gefitinib, are used to treat various cancers, including non-small cell lung cancer and colorectal cancer. These inhibitors work by blocking the tyrosine kinase activity of EGFR, thereby preventing the activation of downstream signaling pathways that promote tumor growth and survival .

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.