EGFR Human, CHO

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

Description

Introduction to EGFR and CHO Expression Systems

The epidermal growth factor receptor (EGFR), a transmembrane receptor tyrosine kinase, plays a critical role in cell proliferation, survival, and differentiation . In cancer, EGFR overexpression or mutations drive tumorigenesis, making it a therapeutic target . Chinese Hamster Ovary (CHO) cells are widely used for recombinant protein production due to their ability to perform complex post-translational modifications, such as glycosylation, and high protein yield . Human EGFR expressed in CHO cells (EGFR Human, CHO) serves as a robust model for studying receptor structure, signaling, and therapeutic resistance mechanisms.

Domain Architecture

EGFR comprises four domains:

DomainFunction
Extracellular (Domains I-IV)Ligand binding (e.g., EGF, TGF-α)
TransmembraneAnchoring to the plasma membrane
Intracellular KinaseTyrosine phosphorylation and signal transduction
C-Terminal TailAutophosphorylation sites and regulatory interactions

CHO-expressed EGFR retains structural fidelity, including glycosylation patterns critical for ligand binding and receptor stability . Recombinant soluble EGFR (extracellular domain) migrates at ~97.5 kDa on SDS-PAGE , while full-length membrane-bound variants exhibit higher molecular weights due to post-translational modifications .

Therapeutic Target Validation

CHO-EGFR systems enable precise studies of:

  • Kinase Inhibitor Efficacy: Erlotinib and gefitinib inhibit EGFR phosphorylation but show reduced efficacy in resistant mutants (e.g., L858R/T790M) .

  • Protein Stability: Drug-sensitive mutants (e.g., L858R, delE746-A750) exhibit shorter half-lives compared to resistant variants, correlating with proteasomal degradation .

  • EGFRvIII, a glioblastoma-associated mutant with a 267-amino acid extracellular deletion, is constitutively active and resistant to ligand binding .

Ligand Binding and Clustering Dynamics

Single-molecule studies in CHO cells reveal:

  • Pre-formed EGFR clusters with stoichiometry ~6 molecules .

  • EGF binding induces larger clusters (~31 EGFR molecules) with a 4:1 EGFR-to-EGF ratio, favoring monomer binding .

  • HER2 heterodimerization occurs transiently, with co-diffusion observed before and after EGF exposure .

Mutant Stability and Drug Resistance

EGFR MutantHalf-Life (CHO Cells)Erlotinib Sensitivity
L858RShorterSensitive
delE746-A750ShorterSensitive
L858R/T790MLongerResistant
Data derived from erlotinib-induced degradation studies .

EGFR-HER2 Interactions

  • Dual-labeled CHO-K1 cells show transient EGFR-HER2 co-localization (dwell time: ~335 ms) .

  • Pertuzumab (anti-HER2/HER3) alters EGFR cluster dynamics, suggesting HER3 involvement in heteroclusters .

Table 1: EGFR Mutant Stability and Drug Response

MutantProtein StabilityDrug SensitivityMechanism
L858RLowErlotinib-sensitiveProteasomal degradation
L858R/T790MHighErlotinib-resistantReduced drug binding
EGFRvIIIHighConstitutively activeLigand-independent dimerization

Table 2: EGFR-EGF Cluster Dynamics

ConditionCluster Size (EGFR)EGFR:EGF Ratio
No EGF~11 moleculesN/A
EGF-bound~31 molecules4:1

Product Specs

Introduction
The epidermal growth factor receptor (EGFR) family, a subset of receptor tyrosine kinases, includes four members: EGFR (also called HER1, ErbB1, or ErbB), ErbB2 (Neu, HER-2), ErbB3 (HER-3), and ErbB4 (HER-4). These type I transmembrane glycoproteins share a common structure: an extracellular domain with two cysteine-rich ligand-binding domains 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) with 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 EGF family ligands, including EGF, amphiregulin, TGFα, betacellulin, epiregulin, heparin-binding EGF, and neuregulin-2, without requiring a co-receptor. Ligand binding triggers EGFR homodimerization or heterodimerization with ErbB2, leading to kinase activation, tyrosine phosphorylation, and signal transduction. EGFR can also form heterodimers with ligand-activated ErbB3 or ErbB4. EGFR signaling is involved in various cellular processes, including proliferation, differentiation, motility, and apoptosis. Additionally, EGFR signaling plays a role in carcinogenesis.
Description
Produced in CHO cells, EGFR is a single, glycosylated polypeptide chain with a molecular weight of 95.5 kDa. It consists of 860 amino acids (residues 25-645) and migrates at 100-150 kDa on SDS-PAGE under reducing conditions. This EGFR protein is expressed with a C-terminal 239 amino acid hIgG-His tag and purified using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The EGFR protein solution is provided at a concentration of 0.25 mg/ml and is formulated in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.

Stability
For optimal storage, the EGFR protein should be kept at 4°C if the entire vial will be used within 2-4 weeks. For longer-term storage, it is recommended to freeze the protein at -20°C.
To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advisable. 
Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the EGFR protein is determined by SDS-PAGE analysis and is found to be greater than 90%.
Synonyms
Epidermal Growth Factor Receptor, Receptor Tyrosine-Protein Kinase ErbB-1, Erb-B2 Receptor Tyrosine Kinase, Proto-Oncogene C-ErbB-1, EC 2.7.10.1, ERBB1, ERBB, HER1, Epidermal Growth Factor Receptor (Avian Erythroblastic Leukemia Viral (V-Erb-B) Oncogene Homolog), Erythroblastic Leukemia Viral (V-Erb-B) Oncogene Homolog (Avian), Avian Erythroblastic Leukemia Viral (V-Erb-B) Oncogene Homolog, Cell Proliferation-Inducing Protein 61, Cell Growth Inhibiting Protein 40, EC 2.7.10, NISBD2, PIG61, MENA.
Source
Chinese Hamster Ovary cells.
Amino Acid Sequence
LEEKKVCQGT SNKLTQLGTF EDHFLSLQRM FNNCEVVLGN LEITYVQRNY DLSFLKTIQE VAGYVLIALN TVERIPLENL QIIRGNMYYE NSYALAVLSN YDANKTGLKE LPMRNLQEIL HGAVRFSNNP ALCNVESIQW RDIVSSDFLS NMSMDFQNHL GSCQKCDPSC PNGSCWGAGE ENCQKLTKII CAQQCSGRCR GKSPSDCCHN QCAAGCTGPR ESDCLVCRKF RDEATCKDTC PPLMLYNPTT YQMDVNPEGK YSFGATCVKK CPRNYVVTDH GSCVRACGAD SYEMEEDGVR KCKKCEGPCR KVCNGIGIGE FKDSLSINAT NIKHFKNCTS ISGDLHILPV AFRGDSFTHT PPLDPQELDI LKTVKEITGF LLIQAWPENR TDLHAFENLE IIRGRTKQHG QFSLAVVSLN ITSLGLRSLK EISDGDVIIS GNKNLCYANT INWKKLFGTS GQKTKIISNR GENSCKATGQ VCHALCSPEG CWGPEPRDCV SCRNVSRGRE CVDKCNLLEG EPREFVENSE CIQCHPECLP QAMNITCTGR GPDNCIQCAH YIDGPHCVKT CPAGVMGENN TLVWKYADAG HVCHLCHPNC TYGCTGPGLE GCPTNGPKIP SRSPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPGKHHHHHH.

Q&A

What is EGFR and its role in human cell signaling?

EGFR (Epidermal Growth Factor Receptor), also known as ERBB, ERBB1, and HER1, is a type I transmembrane protein belonging to the tyrosine protein kinase family. It belongs to a family of tyrosine kinase receptors including Human EGF Receptors (HER) 2, 3, and 4, which all play important roles in cell growth and differentiation. The primary ligands for EGFR include EGF, Heparin-Binding EGF, and Transforming Growth Factor α. Upon ligand binding, EGFR undergoes asymmetric dimerization, composed of an "activator" and a "receiver," which initiates downstream signaling cascades .

For studying EGFR signaling, the A-431 cell line (a human epidermoid carcinoma cell type rich with EGFR) has been instrumental. Stanley Cohen's pioneering work demonstrated that EGF addition caused the phosphorylation of EGFR, triggering various signaling cascades . EGF stimulation causes significant differences in expression of over 3,000 genes and nearly 600 proteins in human epithelial cells, demonstrating the receptor's broad impact on cellular function . These pathways ultimately regulate critical processes including proliferation, differentiation, and inhibition of apoptosis.

What are the common EGFR isoforms expressed in human cells?

Human EGFR is encoded by multiple transcripts resulting in different isoforms. The full-length EGFR (isoform A) arises from two transcripts of 10.5 kb and 5.8 kb from a single promoter region on chromosome 7, with identical protein products . Additionally, alternative transcripts of 1.8, 2.4, and 3.0 kb encode soluble EGFR (sEGFR) isoforms . The 1.8 kb isoform C transcript codes for a secreted 60/80 kDa soluble EGFR protein containing only subdomains I, II, and half of subdomain III of the EGFR extracellular region followed by a unique carboxy-terminal Leu-Ser and 3' UTR .

To identify and characterize these isoforms, researchers typically employ RT-PCR for transcript detection and Western blotting for protein expression. In experimental analyses, CHO/EGFR and CHO/sEGFR lysates serve as positive controls for EGFR and sEGFR expression, respectively . Samples are typically loaded onto 7.5% acrylamide gels for optimal separation of these proteins . These soluble receptor isoforms can modulate EGFR/HER tyrosine kinase activity, adding an additional layer of regulation to EGFR signaling.

How do CHO cell expression systems contribute to EGFR research?

CHO (Chinese Hamster Ovary) cells serve as an excellent expression system for studying human EGFR due to their ability to perform complex post-translational modifications and their relatively low endogenous expression of similar receptors. Researchers typically transfect CHO cells with human EGFR constructs (wild-type or mutant) to study receptor function, ligand binding, dimerization, and downstream signaling in a controlled environment.

The Human EGFR Stable Cell Line in CHO cells enables researchers to study specific aspects of receptor biology isolated from the confounding factors present in cancer cell lines . CHO/EGFR and CHO/sEGFR lysates are frequently used as positive controls in experimental analyses . These systems can be particularly valuable when studying the impact of specific mutations or comparing wild-type and variant receptors, as they provide a consistent cellular background against which differences can be clearly observed.

What methodologies are most effective for detecting EGFR expression?

Researchers employ various techniques to detect and quantify EGFR expression in experimental systems, each offering specific advantages depending on the research question.

MethodApplicationSensitivityKey Considerations
Western BlottingProtein expression and phosphorylationModerateUse 7.5% acrylamide gels for optimal separation of EGFR (~170 kDa)
qRT-PCRmRNA expression of specific isoformsHighDesign primers to distinguish between full-length and soluble isoforms
Flow CytometrySurface expression quantificationHighUse non-permeabilized cells to specifically detect membrane EGFR
ImmunofluorescenceReceptor localization and traffickingModerateCan distinguish between membrane, cytoplasmic, and nuclear EGFR
PCR + Gel ElectrophoresisVerification of EGFR domainsModerateEGFR tyrosine kinase domain PCR products are typically 540 bp
SequencingMutation identificationVery HighCritical for identifying variants in exons 18-22 of tyrosine kinase domain

For comprehensive characterization, combining multiple methods is recommended. For instance, PCR products can be detected on a DNA agarose gel and subsequently sequenced to identify potential mutations or polymorphisms in the tyrosine kinase domain .

What is the relationship between EGFR and cancer development?

EGFR's link to cancer was first recognized when the transforming v-ErbB oncogene of the avian erythroblatosis virus was found to be a mutant homolog of human EGFR . The v-erbB oncogene contained recombinations of the transmembrane and cytoplasmic domains of the EGFR, implicating EGFR aberrations in cancer development . Beyond mutations, overexpression of EGFR promotes cancer progression in carcinomas, sarcomas, non-small cell lung cancer (NSCLC), and malignant gliomas .

EGFR-positive lung cancer occurs in approximately 15% of lung cancer cases, most commonly in adenocarcinoma, a type of non-small cell lung cancer (NSCLC) . This cancer type is more prevalent in women, people of Asian descent, younger individuals, and those without a history of smoking . The levels of EGFR expression predict tumor grade, patient prognosis, and relapse in cancer . Due to its high level of expression and prominent role in tumor growth and metastasis, EGFR is considered an excellent target for pharmacologic intervention in NSCLC .

How do specific mutations in the EGFR tyrosine kinase domain affect therapeutic responses?

EGFR mutations, particularly in exons 18-22 of the tyrosine kinase domain, significantly impact binding affinity and efficacy of targeted inhibitors like gefitinib. Two of the most common mutations found in EGFR-positive NSCLC are Exon 19 deletions and L858R mutations, which show different responses to treatment . Not all changes in EGFR respond equally to treatment, making precise characterization of specific mutations critical for developing effective therapeutic strategies .

To identify these mutations, researchers sequence the tyrosine kinase domain (exons 18-22) of EGFR . PCR products of approximately 540 bp are visualized on DNA agarose gels before sequencing . Interestingly, research has shown differences in how cell lines respond to EGFR modulation—the well-differentiated Ishikawa H cell line responds more robustly to EGFR activation and is more sensitive to receptor inhibition compared to Hec50co cells, which exhibit relative resistance . These differences manifest in both signaling pathway activation and cell cycle regulatory responses to EGFR tyrosine kinase inhibitors like gefitinib .

What mechanisms govern EGFR dimerization and higher-order oligomerization?

Recent studies have identified distinct interactions that stabilize EGFR dimers versus higher-order oligomers . These different states may regulate distinct aspects of EGFR signaling with important implications for normal physiology and disease states. Advanced methodologies for studying these complex structures include super-resolution microscopy techniques that can visualize receptor clusters below the diffraction limit, single-particle tracking to monitor receptor dynamics in live cells, and quantitative FRET approaches to measure multimerization states .

Understanding the structural basis of these oligomeric states is essential for developing next-generation therapeutics that could potentially target specific receptor configurations rather than just inhibiting kinase activity. The three-dimensional architecture of these complexes and the dynamic equilibrium between different oligomeric states remain active areas of investigation.

How do EGFR signaling networks differ between cell types?

EGFR activation triggers multiple downstream signaling pathways, including MAPK/ERK, PI3K/AKT, and JAK/STAT, which vary in importance depending on cell type and context. Studies have demonstrated significant differences in signaling responses between cell lines, such as Ishikawa H cells compared to Hec50co cells .

Specific genes including early growth response 1, sphingosine kinase 2, dual specificity phosphatase 6, and glucocorticoid receptor DNA binding factor 1 form a cluster downstream of EGFR that is differentially regulated by treatment with EGF compared to gefitinib in Ishikawa H cells, but not in Hec50co cells . The well-differentiated Ishikawa H cell line responds more robustly to EGFR activation and shows greater sensitivity to receptor inhibition compared to Hec50co cells, which demonstrate relative resistance .

These differences highlight the importance of cellular context in EGFR signaling. EGF stimulation can cause global phosphorylation of 2,244 proteins at 6,600 sites and significant differences in expression of 3,172 genes and 596 proteins in human mammary epithelial cells . The signaling network is estimated to encompass 122 proteins and 211 interactions, underscoring the complexity of EGFR-mediated cell regulation .

What challenges exist in translating EGFR findings from model systems to clinical applications?

Translating findings from CHO cell systems and other models to human cancer applications requires careful consideration of multiple factors. The Human EGFR Stable Cell Line - MC38 can be used to create in vitro and in vivo models to study the tumor microenvironment, helping bridge the gap between artificial expression systems and clinical reality .

When transitioning from laboratory models to clinical applications, researchers must account for several key differences:

  • Tumor heterogeneity not present in cell lines

  • Influence of the tumor microenvironment on EGFR signaling

  • Differences in receptor density between overexpression models and patient tumors

  • Variations in post-translational modifications between model systems and human tissues

  • Species-specific differences in downstream signaling components

How can researchers optimize EGFR-targeted therapeutic approaches?

A comprehensive research approach for developing improved EGFR-targeted therapies includes:

  • Precise characterization of EGFR mutations in patient samples

  • Understanding resistance mechanisms to existing therapies

  • Exploring combination approaches targeting multiple nodes in EGFR signaling networks

  • Developing inhibitors specific to different EGFR conformational states

  • Investigating synergies between EGFR inhibitors and immunotherapies

Scientists have found many different changes in the EGFR gene in lung cancer, with Exon 19 deletions and L858R mutations being among the most common in EGFR-positive NSCLC . These different mutations can affect response to treatment, making personalized approaches necessary. Evidence suggests EGFR is an excellent target for pharmacologic intervention in NSCLC due to its high level of expression and prominent role in tumor growth and metastasis .

Advanced understanding of the distinct interactions that stabilize different EGFR oligomeric states may open new avenues for therapeutic development beyond traditional tyrosine kinase inhibitors . Such approaches could potentially overcome existing resistance mechanisms and provide more durable responses in EGFR-dependent cancers.

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 are receptor tyrosine kinases. EGFR is activated by binding to its specific ligands, such as epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α).

Structure and Function

EGFR is composed of an extracellular ligand-binding domain, a single transmembrane helix, an intracellular tyrosine kinase domain, and a C-terminal regulatory region. Upon ligand binding, EGFR undergoes dimerization, either as a homodimer or heterodimer with other ErbB family members. This dimerization activates the intrinsic kinase activity of the receptor, leading to autophosphorylation of specific tyrosine residues in the C-terminal domain. These phosphorylated tyrosines serve as docking sites for various signaling proteins, initiating multiple downstream signaling pathways, including the MAPK, PI3K/AKT, and JAK/STAT pathways.

Recombinant EGFR in CHO Cells

Chinese Hamster Ovary (CHO) cells are widely used in biotechnology for the production of recombinant proteins, including human EGFR. Recombinant human EGFR produced in CHO cells is a glycosylated polypeptide chain containing 860 amino acids and has a molecular mass of approximately 95.5 kDa . The recombinant protein is expressed with a 239 amino acid hIgG-His tag at the C-terminus and is purified using proprietary chromatographic techniques .

Applications

Recombinant human EGFR is used in various research and therapeutic applications. It is utilized in studies to understand the molecular mechanisms of EGFR signaling and its role in cancer and other diseases. Additionally, recombinant EGFR is employed in drug discovery and development, particularly in the screening and evaluation of potential EGFR inhibitors for cancer therapy.

Clinical Significance

EGFR is overexpressed or mutated in various cancers, including non-small cell lung cancer, colorectal cancer, and glioblastoma. These alterations lead to aberrant activation of EGFR signaling pathways, promoting uncontrolled cell proliferation and survival. Targeting EGFR with specific inhibitors, such as tyrosine kinase inhibitors (TKIs) and monoclonal antibodies, has become a key therapeutic strategy in the treatment of EGFR-driven cancers.

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.