Used to detect phosphorylated EGFR (S1071) in lysates from human, mouse, or rat tissues/cell lines .
EGFR phosphorylation at S1071 is linked to receptor activation and downstream signaling cascades (e.g., RAS-RAF-MEK-ERK, PI3K-AKT) .
Post-translational modifications at S1071 may regulate EGFR internalization or interaction with adaptor proteins like GRB2 .
Specificity: The antibody shows no cross-reactivity with non-phosphorylated EGFR or other phosphorylated tyrosine residues (e.g., Tyr1068, Tyr1173) .
Validation: Tested in WB using HEK293T cells treated with EGF to induce EGFR phosphorylation .
EGFR Function: A receptor tyrosine kinase critical for cell growth, survival, and migration. Dysregulation is implicated in cancers (e.g., lung, colorectal) .
Phosphorylation Role: S1071 phosphorylation may modulate receptor dimerization, kinase activity, or interaction with downstream effectors .
Cancer Studies: Enables tracking of EGFR activation status in tumor samples, aiding in therapeutic response studies (e.g., tyrosine kinase inhibitors) .
Mechanistic Insights: Useful for dissecting phosphorylation-dependent EGFR interactions in signal transduction pathways .
Phospho-EGFR (S1071) Antibody is a polyclonal antibody specifically designed to recognize the Epidermal Growth Factor Receptor (EGFR) protein only when phosphorylated at serine residue 1071. The antibody is typically generated using synthesized peptides derived from human EGFR encompassing the phosphorylation site of S1071 as immunogens . The specificity for the phosphorylated form allows researchers to distinguish the activated state of EGFR at this particular residue from the non-phosphorylated form, enabling detailed signaling studies.
Phospho-EGFR (S1071) antibody specifically targets serine phosphorylation, while many commonly used phospho-EGFR antibodies target tyrosine residues such as Y1068, Y1086/Y1110, or Y1172 . This distinction is important because:
Serine/threonine phosphorylation often regulates receptor desensitization and internalization, whereas tyrosine phosphorylation typically activates downstream signaling cascades .
Different kinases are responsible for these modifications - serine/threonine kinases versus tyrosine kinases - representing distinct regulatory mechanisms.
The functional outcomes of S1071 phosphorylation may differ significantly from those of tyrosine phosphorylation sites.
This table summarizes key differences between common phospho-EGFR antibodies:
Based on validation data, Phospho-EGFR (S1071) Antibody is primarily recommended for:
Western Blotting (WB): Optimal dilution range 1:500-1:2000
Can detect bands at approximately 134-180 kDa depending on post-translational modifications
ELISA: Recommended dilution of 1:5000
Useful for quantitative assessment of phosphorylation levels
While some vendors may suggest other applications, these two have the most consistent validation data across manufacturers. For specialized applications like immunohistochemistry (IHC) or immunofluorescence (IF), additional validation may be necessary in your specific experimental system.
For effective detection of Phospho-EGFR (S1071), consider these critical sample preparation guidelines:
Phosphatase inhibitors: Always include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride, β-glycerophosphate) in lysis buffers to preserve phosphorylation status .
Lysis conditions: Use RIPA or similar buffers supplemented with protease inhibitors. For membrane proteins like EGFR, addition of 0.1% SDS can improve extraction efficiency .
Sample handling: Process samples rapidly at 4°C to minimize dephosphorylation. Flash freezing in liquid nitrogen is recommended if immediate processing is not possible.
Positive controls: EGF-stimulated A431 cells (human epithelial carcinoma) serve as excellent positive controls, showing robust phosphorylation at S1071 after treatment with 100 ng/mL recombinant human EGF for 5-10 minutes .
Reduction and denaturation: Complete denaturation is critical; use reducing conditions (DTT or β-mercaptoethanol) and heat samples at 95°C for 5 minutes.
To effectively study the kinetics of EGFR S1071 phosphorylation:
Time course experiment design:
Establish baseline (0 min) phosphorylation levels
Stimulate cells with appropriate ligand (typically EGF at 100 ng/mL)
Collect samples at multiple timepoints (30 sec, 2 min, 5 min, 10 min, 30 min, 1 hr, 2 hr)
Process all samples simultaneously to minimize technical variation
Quantification approach:
Always normalize phospho-signal to total EGFR expression levels using a non-phospho-specific EGFR antibody on stripped membranes
Use densitometry software for accurate quantification
Present data as fold-change in phosphorylation relative to baseline
Controls to include:
Unstimulated controls at each timepoint to account for basal changes
Phosphatase treatment controls to confirm signal specificity
EGFR inhibitor controls (e.g., erlotinib) to demonstrate signal regulation
Research has shown that different phosphorylation sites on EGFR demonstrate distinct temporal patterns, with S1071 potentially showing different kinetics compared to tyrosine sites like Y1068 or Y1086 .
Discrepancies between phosphorylation levels at different EGFR sites are common and biologically significant. When analyzing such differences:
Different regulatory mechanisms: Serine phosphorylation (including S1071) is often mediated by downstream kinases rather than receptor autophosphorylation. For example, studies have shown that while EGF stimulation induces rapid Y1068 phosphorylation, S1071 phosphorylation may follow different kinetics and might be regulated by distinct pathways .
Functional implications: Tyrosine phosphorylation sites (e.g., Y1068, Y1086) primarily mediate downstream signaling through recruitment of adaptor proteins like GRB2, while serine phosphorylation often regulates receptor trafficking, desensitization, or cross-talk with other pathways .
Cell type-specific patterns: Different cell types may exhibit distinct phosphorylation patterns. For example, research on EGFR phosphorylation in lung cancer cells (A549) versus breast cancer cells (MDA-MB-231) showed distinct patterns even with the same stimulus .
Interpretative approach: When observing discrepancies, consider:
Is the difference stimulus-specific?
Does inhibition of specific pathways differentially affect these sites?
Are these differences consistent across cell types or unique to your model?
Several factors can contribute to false results when using phospho-specific antibodies:
Potential sources of false positives:
Cross-reactivity with similar phosphorylation motifs on other proteins
Insufficient blocking during Western blot procedures
Sample overloading causing non-specific binding
Inadequate washing steps in immunoassays
Potential sources of false negatives:
Rapid dephosphorylation during sample preparation (insufficient phosphatase inhibitors)
Epitope masking due to protein-protein interactions
Suboptimal antibody concentration
Incomplete protein transfer during Western blotting for large proteins like EGFR
Verification strategies:
Phosphatase treatment controls - sample treatment with lambda phosphatase should abolish signal
Peptide competition assays - pre-incubation with phosphopeptide should block specific binding
siRNA knockdown of EGFR - should reduce or eliminate specific signal
Comparison with other well-characterized phospho-specific antibodies using the same samples
Phosphorylation at serine residues like S1071 has been implicated in EGFR internalization and trafficking. To study these processes:
Subcellular fractionation approach:
Separate cellular compartments (membrane, cytosolic, endosomal fractions)
Analyze the distribution of phospho-EGFR (S1071) versus total EGFR
Compare with markers for different cellular compartments (e.g., Na+/K+ ATPase for plasma membrane, EEA1 for early endosomes)
Imaging techniques:
Combined immunofluorescence using phospho-EGFR (S1071) antibody with compartment markers
Live-cell imaging using fluorescently tagged EGFR combined with phospho-specific antibody staining after fixation
Super-resolution microscopy for detailed localization studies
Receptor trafficking inhibitors:
Use specific inhibitors of different trafficking pathways (e.g., dynamin inhibitors, clathrin inhibitors)
Measure effects on S1071 phosphorylation compared to other EGFR phosphorylation sites
Compare with receptor downregulation measurement using surface biotinylation assays
Research has shown that antibody combinations targeting different EGFR domains can reduce surface receptor levels by up to 80%, suggesting complex regulation of receptor internalization . The relationship between S1071 phosphorylation and this process remains an active area of investigation.
EGFR-targeted therapies, including tyrosine kinase inhibitors like erlotinib, are important cancer treatments, but resistance frequently develops. Investigating the role of S1071 phosphorylation in this context:
Resistance model systems:
Compare S1071 phosphorylation levels between sensitive and resistant cell lines
Develop acquired resistance models through long-term drug exposure
Use patient-derived xenografts from responders versus non-responders
Signaling bypass mechanisms:
Investigate whether S1071 phosphorylation persists despite tyrosine kinase inhibition
Determine which upstream kinases maintain S1071 phosphorylation during drug treatment
Assess whether combined inhibition of these kinases restores drug sensitivity
Methodological approaches:
Phosphoproteomics to comprehensively map changes in EGFR phosphorylation patterns during resistance development
Site-directed mutagenesis (S1071A) to determine functional significance
In vivo models to validate findings from cell culture systems
Research has shown that while EGFR tyrosine phosphorylation sites like Y1092, Y1110, and Y1172 correlate with sensitivity to erlotinib , the role of serine phosphorylation sites including S1071 in modulating drug response represents an understudied area that could reveal novel resistance mechanisms.
EGFR mutations, particularly in lung cancer, significantly impact receptor function and drug sensitivity. Understanding their effect on S1071 phosphorylation:
Comparative analysis across mutation types:
Common activating mutations (exon 19 deletions, L858R)
Resistance mutations (T790M, C797S)
Uncommon mutations with variable clinical responses
Experimental approach:
Isogenic cell lines expressing wild-type versus mutant EGFR
Analysis of basal and ligand-induced phosphorylation patterns
Temporal dynamics of phosphorylation/dephosphorylation at different sites
Structural considerations:
How mutations affect the conformation of the kinase domain
Impact on accessibility of S1071 to kinases and phosphatases
Potential for altered protein-protein interactions affecting serine phosphorylation
Mass spectrometry (MS) provides powerful complementary approaches to antibody-based detection:
Unbiased phosphorylation site mapping:
MS can identify novel and known phosphorylation sites simultaneously
Enables discovery of previously uncharacterized sites that may function together with S1071
Can detect multiple modifications on the same peptide to reveal combinatorial regulation
Quantitative assessment advantages:
Label-free quantitation allows precise measurement of phosphorylation stoichiometry
Multiple reaction monitoring (MRM) provides highly sensitive targeted quantification
SILAC or TMT labeling enables multiplexed comparison across conditions
Methodological workflow:
EGFR immunoprecipitation followed by tryptic digestion
Phosphopeptide enrichment (TiO2, IMAC, or phospho-specific antibodies)
High-resolution LC-MS/MS analysis
Database searching with phosphorylation as variable modification
Studies utilizing mass spectrometry have identified 30 EGFR phosphorylation sites, including 12 serine phosphorylation sites, providing a comprehensive map of receptor modification that extends beyond what can be studied with available antibodies . These approaches have revealed that five sites (pT693, pY1092, pY1110, pY1172 and pY1197) are inhibited by erlotinib in a concentration-dependent manner .
EGFR undergoes multiple types of post-translational modifications that may interact with S1071 phosphorylation:
Cross-talk with other phosphorylation sites:
Hierarchical phosphorylation events where one site influences modification of another
Competitive phosphorylation where different kinases target overlapping regions
Combinatorial effects where specific patterns of multi-site phosphorylation create unique signaling outputs
Interaction with non-phosphorylation modifications:
Ubiquitination - potentially influenced by serine phosphorylation status
Glycosylation - may affect accessibility of kinases to specific residues
Acetylation - emerging modification with potential regulatory functions
Experimental approaches:
Site-directed mutagenesis to create phosphomimetic (S1071D/E) or phospho-dead (S1071A) mutants
Kinase inhibitor profiling to identify relevant upstream enzymes
Proteomics analysis of interactomes specific to phosphorylation status
Methodological considerations:
Use of appropriate phosphatase inhibitors to preserve physiological modification patterns
Analysis of intact proteins to maintain modification relationships
Consideration of spatio-temporal regulation of different modifications
Research has shown that AIB1 knockdown affects both tyrosine and serine/threonine phosphorylation of EGFR, suggesting complex regulatory networks governing receptor modification . Understanding these relationships is critical for developing more effective therapeutic strategies.
EGFR (ErbB1) belongs to a family of receptors including ErbB2, ErbB3, and ErbB4, with significant implications for comparing phosphorylation patterns:
Comparative analysis across ErbB family members:
Sequence alignment to identify equivalent or distinct phosphorylation sites
Conservation of S1071 or analogous sites across the family
Distinct regulatory mechanisms potentially affecting these sites
EGFR isoform considerations:
Experimental approaches:
Isoform-specific detection using selective antibodies
Recombinant expression of different family members for comparative analysis
Receptor co-expression studies to determine heterodimer effects on phosphorylation
Functional implications:
How phosphorylation patterns influence homo- versus heterodimerization
Differential sensitivity to therapeutic antibodies or small molecule inhibitors
Potential for isoform-specific targeting strategies
Research indicates that EGFR isoforms may have distinct functions, with isoform 2 potentially acting as an antagonist of EGF action . Understanding the phosphorylation patterns across these variants could reveal important regulatory mechanisms and therapeutic opportunities.
Recent technological advances have significantly improved detection of low-abundance phosphorylation events:
Enhanced sensitivity methods:
Proximity ligation assay (PLA) - allows visualization of specific phosphorylation events in situ with single-molecule sensitivity
Single-molecule pull-down (SiMPull) - combines immunoprecipitation with single-molecule fluorescence detection
Nanoscale antibody arrays - miniaturized immunoassays with femtomolar sensitivity
Signal amplification approaches:
Tyramide signal amplification (TSA) - enzymatic deposition of fluorescent tyramide for significantly enhanced signal
Rolling circle amplification (RCA) - DNA amplification linked to antibody binding events
Quantum dot-conjugated secondary antibodies - brighter and more stable than conventional fluorophores
Novel sample preparation strategies:
Laser capture microdissection to isolate specific cell populations from heterogeneous tissues
Digital protein analysis through single-cell western blotting
Selective phosphopeptide enrichment using titanium dioxide or immobilized metal affinity chromatography
Research on combination antibody treatments has demonstrated the ability to downregulate EGFR surface expression by up to 80% , highlighting the need for increasingly sensitive detection methods to capture the remaining receptor population and its phosphorylation status.
Phospho-flow cytometry offers unique advantages for analyzing phosphorylation events in mixed cell populations:
Sample preparation optimization:
Critical fixation timing to capture transient phosphorylation events
Permeabilization protocol selection based on epitope accessibility
Buffer compositions that preserve phosphorylation while enabling antibody penetration
Antibody validation for flow cytometry:
Confirmation of specificity in positive control systems (e.g., EGF-stimulated A431 cells)
Titration to determine optimal concentration for signal-to-noise ratio
Testing with phosphatase-treated controls to confirm phospho-specificity
Multi-parameter analysis strategies:
Combined surface marker and phospho-epitope detection
Inclusion of total EGFR measurement for normalization
Integration with cell cycle analysis or viability markers
Data analysis approaches:
Phosphorylation-specific gating strategies
Population-level versus single-cell analysis
Visualization tools for complex phosphorylation relationships
While Phospho-EGFR (S1071) antibodies have not been extensively validated for flow cytometry, approaches similar to those used for phospho-EGFR (Y1068) detection in flow cytometry could be adapted, with appropriate optimization and validation steps.
Phosphorylation status of EGFR has significant potential as a biomarker in cancer:
Diagnostic applications:
Differential phosphorylation patterns between normal and malignant tissues
Correlation with specific cancer subtypes or stages
Potential for non-invasive detection in liquid biopsies
Predictive biomarker potential:
Association with response to EGFR-targeted therapies
Indication of specific resistance mechanisms
Guidance for combination therapy approaches
Methodological considerations for clinical implementation:
Sample collection and preservation protocols to maintain phosphorylation status
Standardization of detection methods across clinical laboratories
Development of quantitative cutoff values for clinical decision-making
Evidence from existing research:
The relationship between S1071 phosphorylation and clinical outcomes remains an important area for investigation, with potential significance for patient stratification and treatment selection strategies.
Understanding the pattern of S1071 phosphorylation across cancer types could provide valuable insights:
Comparative analysis across cancer types:
Epithelial cancers with known EGFR involvement (lung, colorectal, head and neck)
Comparison with cancers where EGFR plays a variable role (breast, pancreatic)
Correlation with EGFR expression levels and mutation status
Progression analysis:
Changes in phosphorylation patterns during tumor development
Association with invasive or metastatic phenotypes
Comparison between primary tumors and metastatic lesions
Methodological approaches:
Tissue microarray analysis with phospho-specific immunohistochemistry
Quantitative image analysis for standardized scoring
Complementary proteomic profiling of selected samples
Research has demonstrated that EGFR phosphorylation patterns differ between cancer types, with studies showing distinct patterns between lung cancer cells (A549), breast cancer cells (MDA-MB-231), and pancreatic cancer cells (PANC-1) . These differences may reflect tissue-specific signaling networks and could influence therapeutic vulnerabilities.
Systems biology approaches can provide valuable insights into complex EGFR signaling networks:
Kinetic modeling approaches:
Ordinary differential equation (ODE) models of EGFR phosphorylation/dephosphorylation
Parameter estimation using time-course experimental data
Sensitivity analysis to identify critical regulatory nodes
Network-based analysis:
Integration of phospho-EGFR (S1071) data with larger signaling networks
Identification of feedback and feedforward loops involving this site
Prediction of system-level responses to perturbations
Multi-scale modeling considerations:
Linking molecular events (phosphorylation) to cellular phenotypes
Integration of spatial aspects of receptor trafficking
Incorporation of tissue-level heterogeneity
Data requirements and experimental design:
Time-resolved measurements across multiple conditions
Quantitative data for model training and validation
Targeted perturbation experiments to test model predictions
Research has shown that EGFR activates at least four major downstream signaling cascades: RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC and STATs modules . Integrating S1071 phosphorylation data into these signaling networks could reveal novel regulatory mechanisms and therapeutic opportunities.
EGFR signaling involves complex feedback mechanisms that influence phosphorylation dynamics:
Temporal sampling strategies:
High-density early timepoints (seconds to minutes) to capture rapid phosphorylation events
Extended timepoints (hours to days) to observe adaptation and feedback effects
Synchronized cell populations to reduce heterogeneity
Perturbation approaches:
Dose-response studies with varying EGF concentrations
Pulses versus sustained stimulation paradigms
Inhibitor time-course studies with pathway-specific inhibitors
Readout technologies:
Multiplexed detection of multiple phosphorylation sites simultaneously
Live-cell reporters for real-time monitoring where feasible
Single-cell approaches to capture population heterogeneity
Experimental considerations for feedback analysis:
Combined inhibition of receptor and downstream pathways
Chemical-genetic approaches for rapid and specific kinase inhibition
Protein synthesis inhibitors to distinguish between immediate and delayed feedback mechanisms