eIF4E is a cap-binding protein essential for initiating mRNA translation. Phosphorylation at serine 209 (S209) enhances its ability to recruit oncogenic mRNAs (e.g., MYC, ATF4) and drive tumor growth . Elevated p-eIF4E (S209) correlates with poor prognosis in cancers such as colorectal carcinoma (CRC) and melanoma .
Western Blot: Detects a single 25 kDa band in HEK293, NIH/3T3, and CRC cell lysates. Specificity confirmed via peptide competition assays and phosphatase treatment .
Dot Blot: Distinguishes phosphorylated S209 peptide from non-phosphorylated controls at 1:1,000 dilution .
Immunoprecipitation: Efficiently enriches p-eIF4E from whole-cell lysates .
Knockin Models: Heterozygous EIF4E S209A/+ CRC cells (HCT 116) showed reduced tumor growth in mice and organoids, linked to suppressed MYC and ATF4 translation .
Stress Response: p-eIF4E (S209) is required for integrated stress response (ISR)-driven glutamine metabolism and AKT activation in CRC .
MYC Regulation: p-eIF4E (S209) licenses MYC mRNA translation, promoting cell proliferation in CRC and prostate cancer .
Stress Adaptation: Drives ATF4-dependent metabolic reprogramming under glutamine deprivation .
Preclinical Models: Inhibition of eIF4E phosphorylation (via S209A mutation or MNK1/2 kinase inhibitors) reduces tumor burden in Apc/KRAS mutant mice .
Biomarker Potential: Overexpression in CRC precursor lesions and hormone-refractory prostate cancer highlights clinical relevance .
Mechanistic Role: Phosphorylated eIF4E recruits eIF4G to form the eIF4F complex, enabling translation of malignancy-associated mRNAs .
Therapeutic Vulnerability: Targeting p-eIF4E (S209) sensitizes cancer cells to metabolic stress and reduces chemoresistance .
Clinical Correlation: High p-eIF4E levels predict poor survival in melanoma and prostate cancer .
CUSABIO has developed a phospho-EIF4E (S209) recombinant monoclonal antibody. This antibody was generated using DNA sequence obtained from splenocytes isolated from animals immunized with human EIF4E phosphopeptide (S209). The DNA sequence was cloned into a plasmid and subsequently transfected into cell lines for in vitro expression. The resulting product is an affinity-chromatography purified rabbit IgG antibody. This phospho-EIF4E (S209) antibody effectively detects the phospho-EIF4E (S209) protein from human sources in ELISA, WB, and IHC applications.
EIF4E plays a crucial role in the efficient translation of a vast majority of capped cellular mRNAs. It acts as a nucleation point for the assembly of the 48S preinitiation complex by binding to the 5'-methylated guanosine cap of mRNA. EIF4E is frequently overexpressed in numerous epithelial tumors and regulates the translation of multiple malignancy-associated mRNAs. Research by Julia H Carter et al. suggests that phosphorylation of EIF4E at serine 209 is associated with tumor growth and decreased survival in malignant melanoma.
Phospho-EIF4E (S209) refers to the eukaryotic translation initiation factor 4E (eIF4E) protein when phosphorylated at serine residue 209. eIF4E is a critical component of the translation initiation complex that binds to the 5' cap structure of mRNAs, facilitating their translation into proteins. The phosphorylation at S209 is a key regulatory event in the mTOR signaling pathway . This post-translational modification enhances eIF4E's affinity for the cap structure and promotes translation of specific mRNAs involved in cell proliferation, survival, and oncogenesis.
eIF4E is also known by several alternative names including EIF4EL1, EIF4F, eIF-4F 25 kDa subunit, and mRNA cap-binding protein . The importance of this phosphorylation lies in its role as a convergence point for multiple signaling pathways that regulate protein synthesis in response to various stimuli. Dysregulation of eIF4E phosphorylation has been implicated in numerous pathological conditions, particularly cancer, making it both a valuable research target and potential therapeutic biomarker.
Phospho-EIF4E (S209) recombinant monoclonal antibodies, such as EP2151Y , are engineered to specifically recognize and bind to eIF4E only when it is phosphorylated at serine 209. These antibodies are developed using recombinant technology, which ensures batch-to-batch consistency and high specificity compared to traditional monoclonal or polyclonal antibodies.
The antibody contains a variable region that precisely recognizes the three-dimensional epitope created by the phosphorylated S209 residue and its surrounding amino acid sequence. The specificity is typically achieved through immunization with a synthetic phosphopeptide corresponding to the region surrounding Ser209 of human eIF4E. This design enables the antibody to discriminate between phosphorylated and non-phosphorylated forms of eIF4E with high selectivity.
In experimental applications, the antibody binds to phosphorylated eIF4E in samples, and this binding is then detected through various methods depending on the application (e.g., secondary antibodies in Western blots or immunofluorescence, direct detection in TR-FRET assays). The signal intensity correlates with the amount of phosphorylated eIF4E present, allowing both qualitative and quantitative assessments.
Phospho-EIF4E (S209) antibodies are versatile tools employed across multiple research applications:
Western Blot (WB): This is one of the most common applications, allowing quantitative analysis of eIF4E phosphorylation levels in cell or tissue lysates. The recombinant monoclonal antibody EP2151Y has been extensively validated for this application across multiple species .
Immunocytochemistry/Immunofluorescence (ICC/IF): These techniques enable visualization of the subcellular localization of phosphorylated eIF4E, providing insights into spatial regulation. Both EP2151Y and polyclonal antibodies have demonstrated utility in this application .
Immunoprecipitation (IP): This application allows isolation of phosphorylated eIF4E and its associated proteins, facilitating studies of protein-protein interactions influenced by this phosphorylation .
Dot Blot: A rapid screening method that can be used for quick assessment of phosphorylation status without the need for electrophoretic separation .
TR-FRET Assays: Homogeneous, high-throughput detection methods like THUNDER™ and HTRF allow quantitative measurement of eIF4E phosphorylation in cell-based experiments without washing steps, making them ideal for drug screening applications .
Each of these applications provides different insights into the regulation, function, and dynamics of eIF4E phosphorylation, allowing researchers to address diverse experimental questions from mechanistic studies to high-throughput screens.
Optimal Western blotting conditions for Phospho-EIF4E (S209) antibodies require careful attention to several parameters:
Critical inclusion of phosphatase inhibitors (sodium fluoride, sodium orthovanadate) in lysis buffers to preserve phosphorylation
Rapid processing of samples at cold temperatures to minimize phosphorylation loss
5% non-fat dry milk (NFDM) in TBST has been validated as effective
Maintain consistent blocking conditions during both blocking and antibody incubation steps
Primary antibody (EP2151Y/ab76256): Ranges from 1/1,000 to 1/100,000 depending on the sample and detection method
For standard applications, 1/1,000 dilution in 5% NFDM/TBST works well for most samples
For strong signals (e.g., stimulated samples), higher dilutions (1/50,000 or 1/100,000) may be necessary to prevent oversaturation
Secondary antibody (e.g., Goat Anti-Rabbit IgG H&L (HRP)): Typically used at 1/10,000 to 1/20,000 dilution
Exposure times vary from 15 seconds to 3 minutes depending on signal strength
Short exposures (15-30 seconds) are often sufficient for stimulated samples
Longer exposures (1-3 minutes) may be required for basal phosphorylation detection
Always include appropriate positive controls (e.g., EGF or serum-stimulated cell lysates)
Include negative controls such as phosphatase-treated lysates to confirm signal specificity
Consider running total eIF4E detection in parallel for normalization purposes
For optimal Immunocytochemistry/Immunofluorescence (ICC/IF) with Phospho-EIF4E (S209) antibodies, follow these sample preparation guidelines:
100% methanol fixation has been validated for preserving phospho-epitopes of eIF4E
Cell density should be optimized to allow clear visualization of individual cells
For adherent cells like HEK293, coating plates with poly-L-lysine may improve attachment
Primary phospho-eIF4E (S209) antibody: Typically used at 1/500 dilution
Secondary antibody (e.g., Alexa Fluor® 488-conjugated anti-rabbit IgG): Used at 1/1,000 dilution
Incubate antibodies in appropriate blocking buffer to minimize background
Optimal incubation times are typically 1-2 hours at room temperature or overnight at 4°C for primary antibodies
DAPI is recommended for nuclear visualization
Additional markers such as alpha-tubulin can provide cytoskeletal reference
Include a pan-eIF4E antibody (e.g., Y448/ab33766) in parallel samples to assess total protein localization versus phosphorylated form
Serum starvation followed by serum stimulation (20%) can be used to modulate eIF4E phosphorylation for positive controls
Phosphatase inhibitor treatment can enhance phospho-specific signals
The search results demonstrate that 20% serum treatment increases cytoplasmic staining of phospho-eIF4E in NIH/3T3 cells
The TR-FRET (Time-Resolved Förster Resonance Energy Transfer) assay for Phospho-EIF4E (S209) detection is a homogeneous, no-wash immunoassay designed for high-throughput screening and quantitative analysis. This sophisticated technique operates through the following mechanism:
The assay employs two specialized antibodies: one labeled with a donor fluorophore (Europium chelate; Eu-Ab1) that recognizes phosphorylated S209 on eIF4E, and another labeled with a far-red acceptor fluorophore (FR-Ab2) that binds to an invariant epitope of eIF4E
When both antibodies bind to the same eIF4E molecule, their proximity enables energy transfer from the donor to the acceptor molecule
Excitation of the Europium chelate (320-340 nm) triggers FRET to the acceptor, which emits at 665 nm
Residual energy from the Eu chelate generates light at 615 nm
The ratio of 665 nm/615 nm signal is used for quantification, which normalizes for well-to-well variations
Cell treatment: Cells are cultured in a microplate and treated with compounds of interest
Cell lysis: Media is removed and cells are lysed with a specialized lysis buffer containing phosphatase inhibitors (sodium fluoride at 1 mM and sodium orthovanadate at 2 mM), followed by a 30-minute incubation
Protein detection: Cell lysate (15 μL) is transferred to a detection plate, the antibody mix (5 μL) is added, followed by a 4-hour incubation before TR-FRET signal reading
Homogeneous format eliminates wash steps, reducing variability and simplifying automation
Time-resolved detection minimizes background fluorescence by measuring long-lived emission signals
Ratiometric readout (665/615 nm) provides internal normalization
High sensitivity due to signal amplification through the FRET process
The validated protocol demonstrates robust performance with Z'-factor values >0.6, indicating excellent assay quality for screening applications
Several cell treatments can effectively modulate eIF4E S209 phosphorylation, allowing researchers to establish experimental models with controlled phosphorylation states:
Treatment | Mechanism | Effect | Reference |
---|---|---|---|
CGP 57380 | MNK1/2 kinase inhibitor | Dose-dependent inhibition | |
Alkaline phosphatase | Direct dephosphorylation | Complete signal elimination |
Optimization of dose and time is essential for each cell type
For HEK293 cells, EGF at 3 nM for 30 minutes provides robust phosphorylation, sufficient for assay development and inhibitor screening
Pre-treatment with inhibitors (e.g., CGP 57380) for 30 minutes before stimulation allows assessment of inhibitory effects
The baseline phosphorylation state (affected by serum starvation protocols) influences the magnitude of response
Phosphatase inhibitors should be included in lysis buffers (typically sodium fluoride at 1 mM and sodium orthovanadate at 2 mM)
These validated treatments provide researchers with reliable tools for establishing positive and negative controls, developing assays, and investigating the regulation of eIF4E phosphorylation across different experimental contexts.
Validating the specificity of Phospho-EIF4E (S209) antibody signals is crucial for ensuring reliable and reproducible results. Multiple complementary approaches can be employed:
Treat a portion of your lysate with alkaline phosphatase to remove phosphorylation
The search results demonstrate that phosphatase-treated 293 cell lysates show significant reduction in signal with phospho-eIF4E (S209) antibody, confirming phospho-specificity
This serves as a definitive negative control for phospho-specific detection
Compare unstimulated cells with those treated with known inducers of eIF4E phosphorylation (e.g., EGF, serum, dexamethasone)
Include samples treated with specific inhibitors like CGP 57380 that block the responsible kinases (MNK1/2)
The dose-dependent response to inhibitors provides further validation of signal specificity
Perform parallel detection with a phospho-specific and a pan-eIF4E antibody
The search results recommend using Anti-eIF4E antibody [Y448] (ab33766) as a pan control for the phospho-specific antibody
Changes in phosphorylation should be detectable with the phospho-specific antibody while total protein levels (detected by the pan antibody) remain constant
This approach confirms that signal changes reflect phosphorylation state rather than protein expression levels
Confirm findings using multiple detection techniques
For example, validate Western blot results with immunofluorescence or TR-FRET assays
The search results show consistency of results across Western blotting and immunofluorescence methods
For TR-FRET assays, Z'-factor determination between untreated and treated conditions provides statistical validation of assay performance
The search results report a Z'-factor of 0.68 for EGF-stimulated versus inhibitor-treated HEK293 cells, indicating excellent assay specificity and reproducibility
Several factors can contribute to weak or absent signals when using Phospho-EIF4E (S209) antibodies. Understanding and addressing these issues is essential for successful experiments:
Insufficient baseline phosphorylation: Without stimulation, some cell types may have very low levels of eIF4E S209 phosphorylation
Loss of phosphorylation during sample preparation: Inadequate phosphatase inhibitors in lysis buffers can result in rapid dephosphorylation
The search results emphasize the importance of phosphatase inhibitors like sodium fluoride (1 mM) and sodium orthovanadate (2 mM) in lysis buffers
Suboptimal antibody dilution: The search results show that antibody concentrations vary widely depending on the application (1/1,000 to 1/100,000 for Western blotting)
Inadequate incubation time: The TR-FRET assay specifies a 4-hour incubation for optimal detection
Antibody degradation due to improper storage or repeated freeze-thaw cycles
For Western Blotting:
Inefficient protein transfer, particularly for smaller proteins like eIF4E (25 kDa)
Improper blocking: The search results recommend 5% non-fat dry milk in TBST for blocking and antibody dilution
Suboptimal detection settings: Exposure times for Western blots range from 15 seconds to 3 minutes depending on signal strength
For Immunofluorescence:
Fixation method incompatible with phospho-epitope preservation
Excessive permeabilization leading to epitope loss
Inadequate antibody penetration into fixed cells
For TR-FRET Assays:
Suboptimal cell density: The protocols specify 50,000 cells/well for HEK293 cells
Insufficient lysis: Incomplete cell disruption reduces available epitopes
Plate reader settings not optimized for TR-FRET detection
Transient phosphorylation: eIF4E S209 phosphorylation may peak and decline rapidly; optimization of timepoints is essential
The search results indicate 30 minutes as an effective time point for EGF stimulation in HEK293 cells
Encountering unexpected band sizes in Western blots with Phospho-EIF4E (S209) antibodies requires systematic troubleshooting:
The predicted molecular weight of eIF4E is approximately 25 kDa
This should be the primary band observed in validated positive controls
Post-translational modifications: Additional modifications beyond phosphorylation (e.g., ubiquitination, SUMOylation) can increase apparent molecular weight
Protein complexes: Incomplete denaturation may preserve eIF4E interactions with binding partners
Solution: Ensure thorough sample denaturation with adequate SDS, heat (95-100°C for 5 minutes), and reducing agents
Proteolytic degradation: Include complete protease inhibitor cocktails in lysis buffers
Use freshly prepared samples or properly stored frozen samples to minimize degradation
Consider reducing sample heating time if fragmentation is suspected
Cross-reactivity: The antibody may recognize similar phospho-epitopes on related proteins
Validate specificity using the phosphatase treatment control shown in the search results
The alkaline phosphatase treatment should eliminate specific phospho-eIF4E bands
Compare patterns with total eIF4E antibodies to distinguish phospho-specific from general eIF4E bands
Use samples from cells treated with EGF or serum as positive controls
Compare with alternative phospho-eIF4E antibodies (the search results mention both EP2151Y monoclonal and a polyclonal antibody )
Gradient gels can improve resolution of closely migrating bands
Longer SDS-PAGE running times may better separate closely migrating species
Optimization of transfer conditions for small proteins (25 kDa range) by adjusting methanol concentration or using specialized transfer buffers
A methodical approach to troubleshooting, combined with appropriate controls, will help identify the source of unexpected bands and guide protocol adjustments to achieve clean, specific detection of phosphorylated eIF4E.
Incorporating Phospho-EIF4E (S209) detection into high-throughput screening (HTS) assays requires optimization of scalable, reliable detection methods:
The THUNDER™ and HTRF TR-FRET assays described in the search results are specifically designed for HTS applications
These homogeneous assays require no wash steps, making them automation-friendly
The search results demonstrate robust performance with Z'-factor values of 0.68, indicating excellent assay quality for screening
Positive controls: EGF-treated cells (3-10 nM for 30 minutes) show strong phospho-eIF4E signals
Negative controls: Unstimulated cells or cells treated with CGP 57380 inhibitor
Include control wells on each plate for normalization of plate-to-plate variation
Primary screen: Use the TR-FRET assay to identify compounds that modulate eIF4E phosphorylation
Counter-screen: Test for effects on total eIF4E levels to distinguish specific phosphorylation modulators
Secondary validation: Confirm hits using orthogonal methods like Western blotting with the recombinant monoclonal antibody
Apply standard HTS statistical parameters (Z', signal/background ratio, coefficient of variation)
Implement dose-response testing of primary hits to establish potency
The search results demonstrate successful inhibition curves for CGP 57380 in both HEK293 and HeLa cells
This systematic approach enables efficient screening of compound libraries for modulators of eIF4E phosphorylation, with applications in both basic research and drug discovery.
Phospho-EIF4E (S209) antibodies can be powerful tools for investigating translation regulation in disease models, with applications spanning multiple research areas:
Monitor eIF4E phosphorylation as a biomarker of mTOR and MAPK pathway activation in tumor samples
Compare phosphorylation levels between normal and malignant tissues using immunohistochemistry (IHC-P)
Evaluate eIF4E phosphorylation as a predictive marker for response to targeted therapies (e.g., mTOR inhibitors, MNK inhibitors)
The search results indicate reactivity with rat brain lysates , suggesting utility in neurological research
Investigate translational control alterations in models of Alzheimer's, Parkinson's, or ALS
Study stress-induced translational reprogramming via eIF4E phosphorylation
Approach | Methodology | Research Insight |
---|---|---|
Tissue analysis | IHC-P or Western blotting of diseased vs. normal tissues | Disease-associated changes in translation regulation |
Cell-based models | Treatment of disease-relevant cell lines with stressors/therapeutics | Mechanism of translational dysregulation |
In vivo models | Analysis of phospho-eIF4E in animal models before/after treatment | Therapeutic response biomarkers |
Correlate phospho-eIF4E levels with disease progression or therapeutic response
Use TR-FRET assays to screen compound libraries for novel modulators of eIF4E phosphorylation
Develop combinatorial treatment approaches targeting both eIF4E phosphorylation and related pathways
Select appropriate models that reflect the disease pathophysiology
Include relevant stimuli or stressors that mirror disease conditions
Consider temporal dynamics of phosphorylation in relation to disease progression
Use multiple detection methods (Western blot, TR-FRET, immunostaining) for comprehensive analysis
By utilizing Phospho-EIF4E (S209) antibodies across these diverse applications, researchers can gain insights into the role of translational regulation in disease pathogenesis and identify potential therapeutic targets or biomarkers.
The study of Phospho-EIF4E (S209) continues to evolve with several promising future directions:
Integration with multi-omics approaches: Combining phospho-eIF4E detection with transcriptomics, proteomics, and ribosome profiling will provide comprehensive insights into how this phosphorylation event selectively influences translation of specific mRNA subsets.
Single-cell analysis: Adapting phospho-eIF4E detection methods for single-cell resolution will reveal cell-to-cell heterogeneity in translational regulation, particularly relevant in complex tissues and tumors.
In vivo dynamics: Development of methods to monitor eIF4E phosphorylation in real-time in living systems will advance our understanding of its temporal regulation in development and disease progression.
Therapeutic targeting: As understanding of eIF4E phosphorylation in disease contexts grows, development of targeted therapeutics modulating this phosphorylation may provide novel treatment approaches for cancers and other conditions with dysregulated translation.
Structural and mechanistic insights: Further investigation of how S209 phosphorylation alters eIF4E function at the molecular level will clarify the mechanistic basis for selective translational effects.