This antibody specifically binds to phosphorylated EGFR at tyrosine 1092 (pY1092), a key site involved in receptor activation and downstream signaling cascades. EGFR phosphorylation at this residue is associated with receptor dimerization, autophosphorylation, and the recruitment of adaptor proteins like GRB2, which drive pathways such as RAS-RAF-MEK-ERK and PI3K-AKT .
The antibody is produced via recombinant DNA technology:
Immunization: Animals are immunized with a synthetic phospho-peptide corresponding to human pY1092-EGFR .
B Cell Cloning: Positive B cells are isolated, and single clones are identified .
Recombinant Expression: Light and heavy chains are amplified via PCR, inserted into a plasmid vector, and transfected into host cells (e.g., HEK293F) for antibody production .
Purification: Affinity chromatography is used to isolate the antibody from cell culture supernatant .
The antibody is validated for:
T790M and T854A Mutations: The antibody has been used to study resistance to EGFR tyrosine kinase inhibitors (TKIs). For example, the T790M mutation (common in lung cancer) reduces drug binding, while T854A (a secondary resistance mutation) decreases inhibitor efficacy .
Combination Therapy: Dual targeting with cetuximab (anti-EGFR antibody) and BIBW-2992 (irreversible TKI) depletes both phosphorylated and total EGFR, overcoming resistance in T790M-positive tumors .
The Phospho-EGFR (Y1092) Recombinant Monoclonal Antibody is produced using protein technology and DNA recombinant techniques. The process involves immunizing animals with a synthesized peptide derived from human phospho-EGFR (Y1092), followed by the isolation of B cells. Positive B cells are then selected and undergo single clone identification. The light and heavy chains of the phospho-EGFR (Y1092) antibody are amplified through PCR and inserted into a plasmid vector to create a recombinant vector. This recombinant vector is transfected into host cells for antibody expression. The phospho-EGFR (Y1092) recombinant monoclonal antibody is purified from the cell culture supernatant using affinity chromatography. This antibody serves as a valuable tool for the detection of human phospho-EGFR (Y1092) protein in ELISA and WB applications.
The Epidermal Growth Factor Receptor (EGFR) is a receptor tyrosine kinase that binds ligands of the EGF family, activating multiple signaling cascades to translate extracellular cues into appropriate cellular responses. These ligands include EGF, TGFA/TGF-alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG, and HBEGF/heparin-binding EGF. Ligand binding triggers receptor homo- and/or heterodimerization and autophosphorylation on key cytoplasmic residues. The phosphorylated receptor recruits adapter proteins such as GRB2, which activates complex downstream signaling cascades. EGFR activates at least four major downstream signaling cascades, including the RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC, and STATs modules. It may also activate the NF-kappa-B signaling cascade. Additionally, EGFR directly phosphorylates other proteins, such as RGS16, activating its GTPase activity and potentially coupling the EGF receptor signaling to the G protein-coupled receptor signaling. It also phosphorylates MUC1, increasing its interaction with SRC and CTNNB1/beta-catenin. EGFR positively regulates cell migration through interaction with CCDC88A/GIV, which retains EGFR at the cell membrane following ligand stimulation, promoting EGFR signaling that triggers cell migration. EGFR plays a role in enhancing learning and memory performance. Isoform 2 may act as an antagonist of EGF action. As a receptor for hepatitis C virus (HCV) in hepatocytes, EGFR facilitates its cell entry. It mediates HCV entry by promoting the formation of the CD81-CLDN1 receptor complexes, which are essential for HCV entry, and by enhancing membrane fusion of cells expressing HCV envelope glycoproteins.
EGFR Y1092 phosphorylation serves as a critical regulatory site in EGFR-mediated signal transduction. Upon ligand binding, EGFR undergoes autophosphorylation at multiple tyrosine residues, including Y1092, which creates binding sites for adaptor molecules such as Grb2. This interaction initiates the mitogen-activated protein kinase/extracellular signal-related kinase (MAPK/ERK) cascade, playing a crucial role in cellular proliferation and survival pathways. Y1092 phosphorylation specifically mediates the activation of downstream signaling cascades including RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCγ-PKC, and STATs modules, converting extracellular stimuli into appropriate cellular responses .
EGFR has two distinct numbering systems that researchers must consider when selecting antibodies and interpreting published data. The first system designates the initiating methionine in the signal sequence as amino acid -24, while the second system (used in some research contexts) denotes this methionine as amino acid +1. Commercial antibodies, including Y1068-specific anti-phospho-EGFR, typically use the first nomenclature. Therefore, Y1068 in commercial antibody nomenclature corresponds to Y1092 in the alternative numbering system .
The Phospho-EGFR (Y1092) Recombinant Monoclonal Antibody has been validated primarily for Western blot (WB) applications using human samples. The antibody demonstrates high specificity for EGFR phosphorylated at the Y1092 residue, making it suitable for measuring EGFR activation status in response to various stimuli or interventions .
When performing Western blot analysis, the recommended dilution is 1.35 μg/mL, though optimization may be necessary depending on sample type and experimental conditions. While the antibody has been extensively validated for WB applications with human samples, applications in other species may require additional validation due to potential differences in epitope conservation across species .
Proper experimental controls are essential for accurate interpretation of phospho-EGFR (Y1092) data:
Positive controls:
EGF-stimulated cell lysates (100 ng/ml EGF) from cells expressing wild-type EGFR
Transiently transfected cells overexpressing wild-type EGFR that has been serum-starved and then EGF-stimulated
Negative controls:
Unstimulated/serum-starved cell lysates
Phosphatase-treated samples
Y1092F mutant EGFR (tyrosine replaced with phenylalanine)
Kinase inhibitor-treated samples (gefitinib or erlotinib at appropriate concentrations)
Loading and normalization controls:
The ratio of phospho-Y1092 signal to total EGFR provides a more accurate quantification of the phosphorylation state than phospho-signal alone, accounting for variations in total EGFR expression levels across samples.
Optimizing Western blot protocols for phospho-EGFR (Y1092) detection requires careful attention to multiple factors:
Sample preparation:
Rapid sample collection and immediate lysis in ice-cold buffer containing phosphatase inhibitors
Inclusion of sodium orthovanadate (1-2 mM) to inhibit tyrosine phosphatases
Maintenance of samples at 4°C throughout processing to minimize dephosphorylation
Gel electrophoresis and transfer:
Use of 7-8% gels for optimal resolution of high molecular weight EGFR (~170 kDa)
Low-methanol transfer buffers for efficient transfer of large proteins
Wet transfer systems at controlled temperature for consistent results
Antibody incubation:
Blocking in BSA rather than milk (milk contains phosphatases)
Primary antibody dilution at 1.35 μg/mL in 5% BSA/TBST
Overnight incubation at 4°C for optimal sensitivity
Detection and stripping:
The table below summarizes relative phosphorylation levels by different kinases at various EGFR tyrosine sites:
Kinase | Y1016 | Y1069 | Y1092 | Y1138 | Y1172 | Y1197 |
---|---|---|---|---|---|---|
ABL (in vitro) | High | Low | High | High | Low | Low |
ABL (co-expression) | High | Low | High | High | Medium | Medium |
EPHB1 (in vitro) | Low | Low | Low | Low | Low | Low |
EPHB1 (serial induction) | High | High | High | High | High | High |
LYN | Medium | Medium | High | Medium | Medium | High |
This differential phosphorylation pattern highlights the importance of selecting appropriate experimental systems when studying specific phosphorylation events .
Several advanced techniques enable researchers to analyze multiple EGFR phosphorylation sites simultaneously:
Phos-Tag Analysis:
This technique allows for the separation of proteins based on their phosphorylation status. Studies have demonstrated that Phos-Tag analysis can reveal the complex degree of phosphorylation across multiple EGFR sites, distinguishing between singly phosphorylated forms and various multi-site phosphorylated species. This approach is particularly valuable for estimating the relative stoichiometry of phosphorylation at different sites and understanding how targeting sequences affect phosphorylation patterns .
LC-MS/MS with SILAC:
Liquid chromatography-tandem mass spectrometry (LC-MS/MS) combined with stable isotope labeling with amino acids in cell culture (SILAC) provides quantitative insights into site-specific phosphorylation. By producing a SILAC standard spike-in and comparing non-phosphorylated peptide forms of experimental phosphoprotein to this standard, researchers can estimate the degree of phosphorylation at specific sites. This approach attributes the loss of non-phosphorylated peptide fragments to the gain of phosphorylated forms .
Multiplexed Phospho-Specific Antibody Arrays:
Using multiple phospho-specific antibodies in array formats allows for the simultaneous detection of numerous phosphorylation sites. This approach can reveal how different kinases, inhibitors, or experimental conditions affect the global phosphorylation landscape of EGFR.
Y1092 phosphorylation plays a significant role in determining sensitivity to EGFR tyrosine kinase inhibitors (TKIs) such as gefitinib and erlotinib, particularly in the context of EGFR mutations. Research comparing wild-type EGFR with mutant forms has revealed important differences in phosphorylation patterns and drug responses.
For the L858R point mutant in exon 21, phosphorylation at Y1092 is inhibited at 10-fold lower concentrations of TKIs compared to wild-type EGFR. This heightened sensitivity to inhibition correlates with the clinical observation that tumors harboring this mutation often respond well to TKI therapy. In contrast, the exon 19 deletion mutant (del L747-S752) demonstrates diminished levels of basal phosphorylation at Y1092 compared to wild-type EGFR, with approximately 16-fold lower ability to autophosphorylate this residue even after EGF stimulation .
These findings suggest that monitoring Y1092 phosphorylation status could serve as a biomarker for predicting TKI response, particularly in the context of specific EGFR mutations. The differential patterns of phosphorylation and inhibitor sensitivity provide valuable insights into the molecular mechanisms underlying TKI efficacy and resistance.
EGFR mutations are frequently observed in non-small cell lung cancers (NSCLCs), particularly adenocarcinomas from patients classified as "never smokers" (those who have smoked fewer than 100 cigarettes in their lifetime). These mutations, primarily located in the tyrosine kinase domain, significantly alter EGFR signaling characteristics, including Y1092 phosphorylation patterns .
Studies have revealed that somatic mutations in the EGFR tyrosine kinase domain are strongly associated with sensitivity to gefitinib and erlotinib. Specifically, in-frame deletions in exon 19 (such as del L747-S752) and point mutations in exon 21 (such as L858R) are commonly found in tumors responsive to these inhibitors. Approximately 75% of mutation-positive tumors in never smokers contained these types of alterations .
Functional analyses of these mutant EGFRs have demonstrated distinct phosphorylation profiles at Y1092 compared to wild-type EGFR:
The exon 19 deletion mutant shows significantly reduced basal phosphorylation at Y1092, with approximately 16-fold lower ability to autophosphorylate this residue even after EGF stimulation. This altered phosphorylation pattern results in markedly low levels of tyrosine-phosphorylated downstream proteins.
The L858R mutant exhibits phosphorylation levels at Y1092 similar to wild-type EGFR but demonstrates enhanced sensitivity to inhibition by TKIs. The phosphorylation at Y1092 in this mutant is inhibited at 10-fold lower concentrations of drug compared to wild-type EGFR .
These findings suggest that monitoring Y1092 phosphorylation might serve as a biomarker for identifying patients likely to respond to EGFR-targeted therapies, potentially guiding treatment decisions in clinical settings.
Non-specific binding and high background are common challenges when working with phospho-specific antibodies. To minimize these issues:
Optimize blocking conditions:
Use 5% BSA in TBST rather than milk-based blockers (milk contains phosphatases)
Consider alternative blockers like casein or commercial blocking solutions if BSA yields high background
Antibody dilution and incubation:
Test multiple antibody dilutions to determine optimal concentration
Extend primary antibody incubation to overnight at 4°C for improved specificity
Include 0.05-0.1% Tween-20 in antibody diluent to reduce non-specific binding
Washing protocols:
Increase washing duration and number of washes (e.g., 5-6 washes for 5-10 minutes each)
Use TBST with higher Tween-20 concentration (0.1-0.2%) for more stringent washing
Sample preparation:
Ensure complete lysis and denaturation of samples
Remove cellular debris by centrifugation at 14,000 × g for 10 minutes
Consider phosphatase inhibitor cocktails containing both serine/threonine and tyrosine phosphatase inhibitors
Validation controls:
Differentiating Y1092 phosphorylation between wild-type and mutant EGFR requires careful experimental design and analysis:
Expression system considerations:
Transiently transfect 293T cells with plasmids encoding wild-type or mutant EGFR constructs
Ensure equal expression levels by normalizing to total EGFR protein
Include untransfected cells as negative controls
Stimulation protocols:
Compare basal (serum-starved) versus EGF-stimulated (100 ng/ml) conditions
Include time course analysis (5, 15, 30, 60 minutes post-stimulation)
Document differential responses to stimulation between wild-type and mutant forms
Inhibitor studies:
Treat with various concentrations of gefitinib or erlotinib (0.001-10 μM)
Generate dose-response curves for inhibition of Y1092 phosphorylation
Calculate IC50 values to quantify differential sensitivity
Quantitative analysis:
Research has demonstrated that exon 19 deletion mutants show dramatically reduced Y1092 phosphorylation compared to wild-type EGFR, while L858R mutants exhibit comparable phosphorylation levels but enhanced sensitivity to TKI inhibition. These distinctive phosphorylation profiles can serve as biochemical signatures for different EGFR mutant forms .
Accurate quantification of Y1092 phosphorylation requires robust methodological approaches:
Western blot densitometry:
Capture images using a digital imaging system within the linear dynamic range
Use software like ImageQuant to perform densitometry analysis
Calculate the ratio of phospho-Y1092 to total EGFR for each sample
Normalize to appropriate controls for comparison across experiments
Phos-Tag analysis for multi-site phosphorylation:
Use Phos-Tag SDS-PAGE to separate proteins based on phosphorylation status
Quantify the ratio of unphosphorylated to phosphorylated bands
Estimate phosphoprotein yield by comparing MYC signal of non-phosphorylated bands to areas covering various phosphoforms
Mass spectrometry-based quantification:
Use SILAC labeling to create heavy-labeled standards (>97% incorporation efficiency)
Mix light phosphoproteins with SILAC standard protein at a 5:1 ratio
Measure light:heavy protein ratios using tryptic fragments lacking tyrosines as "quantitative barcodes"
Calculate site-specific phosphorylation by comparing unmodified peptide abundance between experimental and standard samples
Phosphospecific antibody arrays:
Utilize arrays containing multiple anti-EGFR antibodies recognizing different phosphorylation sites
Compare relative signal intensities across sites and experimental conditions
Include appropriate normalization controls
Each of these methods offers distinct advantages, and combining multiple approaches provides the most comprehensive assessment of EGFR phosphorylation status.