Phospho-EIF4EBP1 (S65) antibodies (e.g., ab259329, A00968S65-1, #9451) are typically rabbit-derived monoclonal or polyclonal reagents validated for techniques like Western blot (WB), immunohistochemistry (IHC), and ELISA . Key features include:
These antibodies exhibit specificity for the S65-phosphorylated form, though some cross-reactivity with phosphorylated serine 101 has been reported under specific conditions .
EIF4EBP1 regulates cap-dependent translation by binding to eIF4E, a component of the eIF4F complex. Phosphorylation at S65—often coordinated with Thr37, Thr46, and Thr70—releases EIF4EBP1 from eIF4E, enabling translation initiation . Key regulatory insights:
Hierarchical Phosphorylation: S65 phosphorylation requires priming at Thr37/Thr46, which are mTORC1 targets .
Functional Impact: Hypophosphorylated EIF4EBP1 inhibits translation, while hyperphosphorylation promotes oncogenic protein synthesis .
EMT and Metastasis: Loss of EIF4EBP1 function elevates Snail translation, driving epithelial-mesenchymal transition (EMT) and cancer cell invasion .
Therapeutic Targeting: Hypoxia-induced dephosphorylation (e.g., in HCT116 colon cancer cells) correlates with reduced eIF4F complex formation and tumor growth inhibition .
Mitotic Translation: During mitosis, 4E-BP1 remains bound to eIF4E despite phosphorylation at S65 and Thr70, suggesting translation initiation remains active .
Rapamycin Sensitivity: Treatment with rapamycin (an mTOR inhibitor) reduces S65 phosphorylation, reactivating EIF4EBP1’s translation-repressive function .
Band Patterns: Western blots often show a primary band at 15–20 kDa and a secondary unknown band at ~35 kDa, likely due to phosphorylation-dependent aggregation .
Buffer Compatibility: Optimal results require blocking buffers like 5% NFDM/TBST to reduce background .
Phosphatase Controls: Lambda phosphatase treatment abolishes signal, confirming phosphorylation specificity .
Hierarchy in Phosphorylation: S65 phosphorylation is contingent on prior modifications at Thr37/Thr46, as shown in HEK293 cells .
Antibody Cross-Reactivity: The "anti-S65" antibody may recognize S101-phosphorylated EIF4EBP1 in mutants lacking S65, necessitating careful experimental design .
Mitotic Resistance: Mitotic 5′-TOP mRNA translation remains active despite mTOR inhibition, linked to unique 4E-BP1 phosphorylation patterns .
Phospho-EIF4EBP1 (S65) antibodies are predominantly used in these research applications:
Western Blotting (WB): To detect and quantify the phosphorylation status of 4E-BP1 at Ser65 in cell and tissue lysates
Immunohistochemistry (IHC): For visualizing expression and localization patterns in tissue sections
ELISA: For quantitative assessment of phosphorylation levels
Signaling Pathway Analysis: For monitoring mTOR pathway activity and response to therapeutic interventions
Cell Cycle Studies: Particularly in examining mitosis-G1 transition regulation where CDK4 has been shown to influence 4E-BP1 phosphorylation
To effectively differentiate between total and phosphorylated 4E-BP1:
Sequential immunoblotting: Run duplicate gels or strip and reprobe membranes with both phospho-specific and total 4E-BP1 antibodies
Band shift analysis: Phosphorylated 4E-BP1 demonstrates characteristic migration patterns on SDS-PAGE, appearing as multiple slower-migrating bands compared to the hypophosphorylated form
Phosphatase treatment controls: Include samples treated with phosphatases to confirm band identity
Signal normalization: Always normalize phospho-specific signals to total protein levels
Phospho-mutant controls: When possible, utilize S65A mutants as negative controls to validate antibody specificity
Research has demonstrated that extensive analysis of the migration pattern of 4E-BP1 shows Ser65 phosphorylation substantially contributes to the characteristic SDS-PAGE migration pattern. The hyperphosphorylated form appears as slower-migrating bands compared to the hypophosphorylated form .
Recommended controls for phospho-4E-BP1 (S65) experiments:
When designing phosphorylation studies, rapamycin and TOR-KI compound MLN0128 can be particularly useful as negative controls, as they have demonstrated differing effects on 4E-BP1 phosphorylation sites, with rapamycin specifically inhibiting phosphorylation at Ser65 .
Optimization protocol for Western blotting of phospho-4E-BP1 (S65):
Sample preparation:
Lyse cells in buffer containing phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate)
Maintain cold temperatures throughout processing
Use fresh samples when possible or snap-freeze in liquid nitrogen
Gel selection and separation:
Use 12-15% SDS-PAGE gels to optimize resolution of the low molecular weight 4E-BP1 (~12-15kDa)
Consider using Phos-tag™ gels for enhanced separation of phosphorylated species
Transfer conditions:
Use PVDF membrane (0.2μm) rather than nitrocellulose for better protein retention
Short transfer times with careful temperature monitoring
Blocking and antibody incubation:
Signal detection considerations:
Note that researchers should anticipate multiple bands representing different phosphorylation states, with the hyperphosphorylated forms migrating more slowly .
The phosphorylation of 4E-BP1 at Ser65 can be studied across various tissue and cell types, with certain systems showing particular relevance:
Highly suitable experimental systems:
Lymphocytes (B and T cells): Exhibit unique rapamycin sensitivity in the 4E-BP–eIF4E axis that promotes clonal expansion. Studies have shown differential phosphorylation patterns of 4E-BP1 and 4E-BP2 in activated B cells in response to rapamycin and MLN0128 .
Cancer cell lines: Various carcinoma cell lines show overexpression of 4E-BP1 and altered phosphorylation patterns, making them valuable for studying dysregulation in cancer contexts .
Fibroblasts: Have been used extensively for fundamental studies of 4E-BP1 phosphorylation mechanisms .
Epidermal cells: JB6 mouse epidermal cell lines have demonstrated UVB-induced phosphorylation of 4E-BP1 at multiple sites including Ser65, mediated by p38MAPK and MSK1, providing insight into UV-promoted tumorigenesis mechanisms .
HEK293 cells: Frequently used as model systems for studying rapamycin-sensitive phosphorylation of 4E-BP1 .
When selecting an experimental system, researchers should consider the specific signaling pathways of interest, as different cell types exhibit varying kinase activities and 4E-BP1 regulation patterns.
Multiple kinases can phosphorylate 4E-BP1 at Ser65, including mTORC1, ERK, p38MAPK, MSK1, PIM2, and CDK4. Distinguishing between these kinases experimentally requires a multi-faceted approach:
Experimental strategies for kinase identification:
Selective kinase inhibitors:
Kinase knockdown/knockout approaches:
In vitro kinase assays:
Cell cycle synchronization:
Signal pathway activation:
The experimental evidence indicates differential effects of kinases on 4E-BP1 phosphorylation sites. For instance, Erk efficiently phosphorylates S65 but acts less efficiently at other sites like S101 , while CDK4 has been newly identified as a 4E-BP1 kinase involved in mitosis-G1 transition .
Cross-reactivity is a significant concern with phospho-specific antibodies. For Phospho-EIF4EBP1 (S65) antibodies, several important issues have been documented:
Known cross-reactivity issues:
S101 recognition: Research has demonstrated that some anti-S65[P] antibodies also recognize 4E-BP1 phosphorylated at S101 due to sequence similarity (RNS65PV and RNS101PE contexts) .
4E-BP family cross-reactivity: Antibodies may recognize phosphorylated sites on other 4E-BP family members (4E-BP2, 4E-BP3) due to conserved phosphorylation motifs.
Unknown ~35kDa band: Some antibodies recognize an unknown phosphorylation-related band around 35kDa .
Strategies to address cross-reactivity:
Multiple mutation controls:
Kinase-specific phosphorylation:
Peptide competition assays:
Pre-incubate antibody with phospho-S65 peptide vs. phospho-S101 peptide
Differential blocking indicates cross-reactivity
Antibody validation across sources:
Mass spectrometry confirmation:
For critical experiments, validate phosphorylation site assignments using phospho-proteomics
The ability of antibodies raised against S65[P] to also recognize S101[P] varies between sources and batches, making validation crucial for each experimental context .
The phosphorylation of 4E-BP1 occurs in a hierarchical and coordinated manner across multiple sites, with Ser65 playing a key role in the complete inactivation of 4E-BP1's repressor function. Understanding these relationships is crucial for interpreting experimental data:
Phosphorylation site relationships:
Hierarchical phosphorylation model:
Thr37/46 phosphorylation typically precedes Ser65 phosphorylation
Ser65 phosphorylation often depends on prior phosphorylation at Thr37/46
Complete hyperphosphorylation at multiple sites (including Thr70) is required for full dissociation from eIF4E
Differential sensitivity to inhibitors:
Cell-type specific patterns:
Kinase-specific patterns:
Migration pattern significance:
Understanding these complex relationships helps researchers interpret the biological significance of observed phosphorylation patterns and design appropriate experimental controls.
Researchers should be aware of several common artifacts and misinterpretations when working with Phospho-EIF4EBP1 (S65) antibodies:
Common issues and solutions:
Misidentification of phosphorylation sites:
Misinterpretation of band patterns:
Issue: 4E-BP1 appears as multiple bands on Western blots representing different phosphorylation states
Solution: Include controls with kinase inhibitors or phosphatase treatment to identify specific forms
Unknown bands:
Inconsistent results between experimental repeats:
Issue: Phosphorylation states are highly dynamic and sensitive to cell conditions
Solution: Standardize cell culture conditions, harvesting methods, and lysis buffer composition; include positive controls in each experiment
Rapamycin resistance misinterpretation:
Cell cycle-dependent variations:
Stimulus-dependent phosphorylation kinetics:
Issue: Different stimuli lead to different phosphorylation patterns and kinetics
Solution: Include appropriate time courses and document stimulation conditions precisely
Degradation during sample preparation:
Issue: Phosphorylation signals can be lost due to phosphatase activity during processing
Solution: Use strong phosphatase inhibitor cocktails in all buffers; maintain samples at cold temperatures
Understanding these potential artifacts and implementing appropriate controls allows for more accurate interpretation of experimental results and avoids common pitfalls in 4E-BP1 phosphorylation analysis.
Accurate quantification of 4E-BP1 Ser65 phosphorylation requires careful consideration of normalization approaches and quantitative techniques:
Quantification methods and considerations:
Western blot quantification:
Normalization strategy: Express phospho-signal as ratio to total 4E-BP1
Dynamic range issues: Use appropriate exposure times to avoid signal saturation
Multiple band consideration: Decide whether to quantify all hyperphosphorylated bands or only specific forms
Software tools: Use appropriate image analysis software with background subtraction
ELISA-based approaches:
Sandwich ELISA: Capture with total 4E-BP1 antibody, detect with phospho-specific antibody
Sensitivity enhancement: Consider amplification systems for low abundance detection
Standard curves: Generate using recombinant phosphorylated proteins
Flow cytometry:
Single-cell resolution: Allows assessment of cell-to-cell variability in signaling
Multiparameter analysis: Combine with cell cycle or phenotypic markers
Fixation protocol: Optimize to preserve phospho-epitopes
Mass spectrometry-based quantification:
Absolute quantification: Use isotope-labeled synthetic phosphopeptides as standards
Phosphosite occupancy: Calculate percentage of 4E-BP1 phosphorylated at Ser65
Multi-site analysis: Simultaneously quantify all phosphorylation sites
In-cell western/cytoblotting:
High-throughput capability: Allows screening of multiple conditions
Direct cell-based measurement: Minimizes processing artifacts
Normalization considerations:
Loading controls: Include both total 4E-BP1 and housekeeping proteins
Phosphorylation site ratios: Compare Ser65 to other sites (e.g., Thr37/46)
Treatment controls: Include maximally stimulated samples (e.g., insulin) and maximally inhibited samples (e.g., mTOR inhibitors)
Statistical analysis:
Biological replicates: Minimum of three independent experiments
Technical replicates: Triplicate measurements when possible
Appropriate statistical tests: Paired analyses for treatment comparisons
Implementing these quantification strategies will provide more reliable and reproducible measurements of 4E-BP1 Ser65 phosphorylation across experimental conditions.
When faced with contradictory results between different detection methods, researchers should follow a systematic approach to troubleshooting and interpretation:
Resolving contradictory results:
Method-specific biases assessment:
Western blotting: May be affected by transfer efficiency, antibody specificity, and protein extraction methods
ELISA: Can be influenced by matrix effects and epitope accessibility in native proteins
Immunohistochemistry: Fixation methods significantly impact phospho-epitope detection
Mass spectrometry: Sample preparation and ionization efficiency can bias detection
Antibody-related considerations:
Biological context evaluation:
Technical validation approaches:
Genetic controls: Use S65A mutants to confirm signal specificity
Pharmacological controls: Apply kinase inhibitors and phosphatase treatments
Method comparison: Systematically compare detection across platforms using identical samples
Contextual interpretation framework:
| Scenario | Possible Interpretation | Recommended Investigation |
|---|---|---|
| Positive WB, negative ELISA | Epitope masking in native form | Use denatured protein in ELISA |
| Positive IHC, negative WB | Fixation-induced artifact | Test multiple fixation methods |
| Different results with different antibodies | Epitope-specific recognition | Map epitope regions; try multiple antibodies |
| Different results in different cell types | Cell-type specific regulation | Check kinase expression profiles |
| Signal in WT but also in S65A mutant | Antibody cross-reactivity | Test S65A/S101A double mutant |
Resolution strategies:
Orthogonal validation: Confirm with an antibody-independent method (e.g., mass spectrometry)
Functional correlation: Assess whether observed phosphorylation correlates with expected biological outcomes (e.g., eIF4E binding)
Mechanistic testing: Manipulate upstream pathways to determine which accurately reflects expected biology
When interpreting contradictory results, consider that different methods may reveal complementary aspects of 4E-BP1 biology rather than simply representing technique failure.
The phosphorylation of 4E-BP1 at Ser65 plays a critical role in cancer development through its effects on cap-dependent translation of oncogenic mRNAs:
Cancer relevance and therapeutic targeting:
Oncogenic mechanisms:
Cancer-specific regulation:
UVB exposure induces phosphorylation of 4E-BP1 at multiple sites (including Ser65) via p38MAPK and MSK1, contributing to UV-promoted tumorigenesis
Ionizing radiation stimulates protein synthesis via ATM-dependent ERK phosphorylation of 4E-BP1 at Ser65
PIM kinases can directly phosphorylate 4E-BP1 at Ser65, creating rapamycin resistance in some cancers
Therapeutic targeting strategies:
mTOR inhibitors: First-generation (rapamycin/rapalogs) and second-generation (TOR-KI compounds like MLN0128) targeting the mTORC1 pathway
Dual pathway inhibition: Combining mTOR and MEK/ERK pathway inhibitors to block multiple kinases that phosphorylate Ser65
CDK4/6 inhibitors: May influence 4E-BP1 phosphorylation during cell cycle progression
eIF4E-eIF4G interface inhibitors: Compounds like EGPI-1 target downstream of 4E-BP1 phosphorylation
Combination therapy approaches:
Biomarker development:
Future research directions:
The role of 4E-BP1 in cancer continues to be an active area of research, with particular emphasis on understanding the differential sensitivities of tumors to translation-targeting therapies and developing more effective combination approaches.
The recent identification of CDK4 as a 4E-BP1 kinase has expanded our understanding of how translation is regulated during cell cycle progression:
CDK4 and cell cycle-dependent regulation:
Novel kinase discovery:
Cell cycle phase specificity:
Integrated regulatory model:
Traditional view: mTORC1 as the primary regulator of 4E-BP1 phosphorylation
Expanded view: Multiple kinases including mTORC1, ERK, p38MAPK, and now CDK4 act in concert or sequentially
Temporal regulation: Different kinases may predominate at different cell cycle phases
Therapeutic implications:
CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib) may affect translation through 4E-BP1
Combined targeting of cell cycle and translational machinery may provide synergistic effects
Resistance to mTOR inhibitors might be addressed by co-targeting CDK4/6
Research opportunities:
Determining the specific 4E-BP1 phosphorylation sites targeted by CDK4
Investigating potential crosstalk between CDK4 and mTORC1 in regulating 4E-BP1
Exploring cell type-specific differences in CDK4-mediated regulation
Examining whether other CDKs also target 4E-BP1 at different cell cycle phases
Experimental approaches:
Cell synchronization studies to isolate mitosis-G1 transition effects
CDK4 inhibitor time course experiments with phospho-site specific antibodies
In vitro kinase assays with recombinant CDK4 and 4E-BP1 variants
Mass spectrometry to map CDK4-dependent phosphorylation sites
The discovery of CDK4 as a 4E-BP1 kinase introduces an important new dimension to our understanding of translational control, suggesting that cap-dependent translation is specifically regulated during cell cycle transitions to coordinate growth with division.
Several cutting-edge technologies are transforming our ability to study 4E-BP1 phosphorylation with unprecedented spatial and temporal resolution:
Emerging technologies and approaches:
Live-cell phosphorylation sensors:
FRET-based biosensors: Engineered constructs with 4E-BP1 sandwiched between fluorescent proteins that change energy transfer upon phosphorylation
Split luciferase complementation: Systems that report on 4E-BP1-eIF4E interaction in real-time
Phospho-specific nanobodies: Combined with fluorescent tags for real-time visualization
Advanced microscopy techniques:
Super-resolution microscopy: Techniques like STORM and PALM for nanoscale visualization of 4E-BP1 localization and complex formation
Light-sheet microscopy: For rapid 3D imaging of phosphorylation dynamics
Correlative light and electron microscopy (CLEM): Connecting phosphorylation status with ultrastructural features
Single-cell analysis technologies:
Single-cell phosphoproteomics: Measuring phosphorylation heterogeneity within populations
Mass cytometry (CyTOF): Simultaneous measurement of multiple phosphorylation sites in thousands of individual cells
Microfluidic approaches: Tracking individual cells over time while manipulating signaling pathways
Genome editing for endogenous tagging:
CRISPR-Cas9 knock-in: Introduction of fluorescent or epitope tags at endogenous 4E-BP1 loci
Base editing: Precise mutation of phosphorylation sites without double-strand breaks
Auxin-inducible degron (AID): Rapid protein depletion systems for studying 4E-BP1 dynamics
Optogenetic and chemogenetic tools:
Optogenetic kinase activation: Light-controlled activation of mTOR or other 4E-BP1 kinases
Chemically-induced dimerization: Rapid and reversible recruitment of 4E-BP1 to subcellular compartments
Photocaged phosphoamino acids: Spatiotemporal control of specific phosphorylation sites
Computational approaches:
Machine learning algorithms: For pattern recognition in phosphorylation dynamics
Network modeling: Integrating 4E-BP1 phosphorylation into broader signaling networks
Single-cell trajectory analysis: Mapping phosphorylation changes through cell state transitions
Spatial proteomics:
Proximity labeling: BioID or APEX2 fusion proteins to identify spatial regulators of 4E-BP1
Subcellular fractionation with phosphoproteomics: Compartment-specific phosphorylation analysis
These technologies are enabling researchers to move beyond static snapshots of 4E-BP1 phosphorylation to understand the dynamic regulation of translation initiation in living cells under physiological and pathological conditions.