Phospho-ELK1 (Serine 389) Antibody is a specialized immunological tool designed to detect endogenous ELK1 protein phosphorylated at residue Ser389. ELK1 is a transcription factor critical for regulating cellular proliferation, apoptosis, and neuronal differentiation via its interaction with the serum response element (SRE) and serum response factor (SRF) . Phosphorylation at Ser389, mediated by mitogen-activated protein kinases (MAPKs), is essential for ELK1’s transcriptional activation and nuclear translocation . This antibody enables researchers to study dynamic post-translational modifications of ELK1 in response to extracellular signals such as growth factors, stress, or neurotransmitters .
The Phospho-ELK1 (Ser389) Antibody is typically a rabbit polyclonal antibody developed using synthetic peptides corresponding to phosphorylated Ser389 residues in human ELK1 . Key attributes include:
The antibody’s specificity is validated through phospho-peptide competition assays, ensuring it recognizes ELK1 only when phosphorylated at Ser389 . The immunogen is a synthesized peptide derived from residues surrounding phosphorylated Ser389 in human ELK1 . Cross-reactivity studies confirm minimal off-target binding, with positive signals observed in UV-treated Jurkat cell lysates .
Phospho-ELK1 (Ser389) Antibody is widely used to investigate:
MAPK/ERK Signaling Dynamics: Phosphorylation at Ser389 correlates with ERK activation, enabling studies of growth factor-induced signaling .
Neuronal Differentiation: ELK1 nuclear translocation, driven by Ser389 phosphorylation, regulates dendritic elongation and cytoskeleton dynamics in neurons .
Transcriptional Regulation: The antibody identifies phosphorylated ELK1 bound to SRE-containing promoters (e.g., c-Fos, Egr-1) during mitogen stimulation .
Dual Role of Ser389 Phosphorylation:
Pathophysiological Relevance:
ELK1 functions as a critical component of the ternary complex that binds to the serum response element (SRE) and mediates gene activity in response to serum and growth factors. The protein undergoes phosphorylation by MAP kinase pathways at multiple S/T motifs located at its C-terminus, with phosphorylation at Ser389 being particularly significant for its activity . This post-translational modification is crucial for transcriptional activation of ELK1 and represents a key regulatory step in the signaling cascade .
ELK1 serves as a nuclear target for the ras-raf-MAPK signaling cascade, with phosphorylation events at sites like Ser389 directly linking cytoplasmic signaling to nuclear gene regulation . While Ser383 phosphorylation has received significant attention in earlier studies, research indicates that phosphorylation at Ser389 works in concert with Ser383 modification to achieve maximal activation of ELK1-mediated transcription .
Phosphorylation at Ser389 induces significant conformational changes in both the secondary and tertiary structure of ELK1. Spectroscopic studies using circular dichroism (CD) and fluorescence emission have demonstrated that phosphorylation events, including at Ser389, increase α-helicity in the protein structure and alter the tertiary conformation . These structural changes directly influence:
DNA binding capacity - Phosphorylation increases ELK1's affinity for target DNA sequences
Protein-protein interactions - Although phosphorylation does not appear to significantly alter binding efficiency with SRF as shown by GST pull-down assays
Transcriptional activation potential - The conformational shift exposes functional domains necessary for recruiting transcriptional machinery
The structural change appears to involve interactions between the phosphorylated C-terminal activation domain and the ETS domain, as evidenced by fluorescence emission spectroscopy showing altered environments around tryptophan residues following phosphorylation .
Phospho-specific antibodies against ELK1 (S389) provide powerful tools for studying signaling kinetics in real-time. When designing experiments to investigate signal transduction dynamics, researchers should consider:
Time-course experiments: Monitoring phosphorylation at multiple time points (0-60 minutes) following stimulation can reveal the temporal dynamics of ELK1 activation. Evidence suggests maximum phosphorylation occurs after approximately 60 minutes of stimulation, coinciding with maximal DNA binding capacity .
Spatial distribution analysis: Combining phospho-ELK1 (S389) antibodies with subcellular fractionation or immunofluorescence microscopy enables tracking of the activated transcription factor's translocation between cytoplasmic and nuclear compartments.
Multiplexed pathway analysis: Using phospho-ELK1 (S389) antibodies alongside markers for upstream kinases (like ERK, JNK, or p38) allows for comprehensive pathway mapping and identification of rate-limiting steps in the signaling cascade.
Cell-based ELISAs: These provide quantitative measurement of phosphorylated ELK1 levels in response to different stimuli, allowing for high-throughput screening of pathway modulators .
For optimal results, normalize phospho-ELK1 (S389) levels to total ELK1 protein expression to account for variations in baseline protein abundance across experimental conditions.
Threshold effects: High levels of phosphorylation are required to achieve maximal DNA binding capacity, suggesting a cooperative mechanism between phosphorylation sites .
Temporal coordination: Different phosphorylation sites may be modified in a specific sequence, with early phosphorylation events priming the protein for subsequent modifications.
Conformational heterogeneity: Mutational studies involving Elk-1(S383A/S389A) demonstrate that these mutants can still undergo phosphorylation-induced conformational changes, albeit defective ones, indicating that multiple determinants influence the precise structural changes induced by phosphorylation .
Functional specialization: Different phosphorylation patterns may direct ELK1 toward specific gene targets or protein interaction partners.
When investigating multi-site phosphorylation, researchers should employ phospho-specific antibodies against various sites in combination with mass spectrometry to map the complete phosphorylation landscape and correlate specific patterns with downstream outcomes.
For successful Western blot detection of phosphorylated ELK1 at Ser389, researchers should follow these methodological guidelines:
Sample preparation:
Rapidly harvest and lyse cells to prevent dephosphorylation by endogenous phosphatases
Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) in lysis buffers
Maintain samples at 4°C throughout processing
Gel electrophoresis and transfer:
Antibody incubation:
Detection and validation:
Include both phosphorylated and non-phosphorylated control samples
Consider using lambda phosphatase-treated samples as negative controls
Compare results with total ELK1 antibody staining on parallel blots or after stripping
Expected results include detection of a band at approximately 45 kDa that increases in intensity following stimulation with growth factors or activators of the MAPK pathway .
Optimizing immunohistochemistry (IHC) protocols for phospho-ELK1 (S389) detection requires careful attention to tissue preservation, antigen retrieval, and signal amplification:
Tissue preparation:
Use freshly fixed tissues or properly stored paraffin-embedded specimens
Perfusion fixation is preferable for animal tissues to preserve phospho-epitopes
Limit fixation time to prevent masking of phospho-epitopes
Antigen retrieval:
Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 8.0)
Optimize retrieval time (10-20 minutes) to balance epitope exposure and tissue integrity
Allow slides to cool slowly to room temperature after retrieval
Antibody application:
Controls and validation:
Successful IHC staining typically shows nuclear localization of phospho-ELK1 (S389), with intensity varying based on tissue type and activation state of the MAPK pathway.
Designing experiments to study ELK1 phosphorylation kinetics requires careful consideration of temporal resolution, stimulus parameters, and detection methods:
Stimulus selection and optimization:
Choose physiologically relevant stimuli (e.g., growth factors, serum, stress inducers)
Determine optimal stimulus concentration through dose-response experiments
Consider using specific pathway activators (e.g., phorbol esters for PKC/MAPK) alongside broader stimuli
Time-course design:
Detection methods:
Data analysis approach:
Plot phosphorylation intensity versus time
Calculate rate constants for phosphorylation and dephosphorylation phases
Correlate phosphorylation kinetics with downstream functional outcomes (e.g., gene expression)
| Time Point | Expected Phospho-ELK1 (S389) Response |
|---|---|
| 0-5 min | Initial phosphorylation detectable |
| 10-30 min | Rapid increase in phosphorylation |
| 60 min | Maximum phosphorylation achieved |
| 120+ min | Potential decrease due to feedback |
For meaningful interpretation, always normalize phospho-ELK1 (S389) signal to total ELK1 levels to account for variations in protein expression.
Researchers frequently encounter several challenges when working with phospho-ELK1 (S389) antibodies that can be addressed through methodological refinements:
Low signal intensity:
Cause: Rapid dephosphorylation during sample preparation
Solution: Immediately lyse samples in buffers containing phosphatase inhibitors; maintain samples at 4°C throughout processing
High background:
Cause: Non-specific antibody binding or inadequate blocking
Solution: Use 5% BSA instead of milk for blocking; optimize antibody dilutions; increase washing times and volumes
Inconsistent results between replicates:
Cause: Variable baseline phosphorylation or technical variability
Solution: Synchronize cells prior to experiments; standardize cell densities and lysis conditions; include internal control samples across experiments
Cross-reactivity with other phospho-proteins:
Cause: Antibody recognizing similar phospho-epitopes on other proteins
Solution: Validate specificity using knockout/knockdown samples or peptide competition assays; compare results with alternative phospho-ELK1 antibodies
Inability to detect phosphorylation changes:
Cause: Suboptimal stimulation conditions or timing
Solution: Verify pathway activation using established markers (e.g., phospho-ERK); optimize stimulus concentration and exposure time; consider cell type-specific response kinetics
When troubleshooting, systematically modify one variable at a time while maintaining appropriate controls to identify the specific source of the issue.
The relationship between ELK1 phosphorylation at Ser389 and its functional properties has been characterized through multiple experimental approaches that reveal a mechanistic connection:
Conformational dynamics:
Phosphorylation at Ser389, along with other sites, induces measurable changes in both secondary and tertiary structure of ELK1
Circular dichroism studies show increased α-helicity following phosphorylation
Fluorescence emission spectroscopy demonstrates altered environments around tryptophan residues, indicating tertiary structural rearrangements
Structure-function relationship:
The B-box domain plays a critical role in mediating phosphorylation-induced conformational changes
Mutational analysis (e.g., L158P mutation) reveals that while secondary structural changes still occur, tertiary structural alterations are compromised in this mutant
This structure-function relationship directly impacts DNA binding capacity
DNA binding capacity correlation:
Maximum DNA binding is achieved at high levels of phosphorylation (around 60 minutes post-stimulation)
The stoichiometry of phosphorylation is crucial, suggesting cooperative effects between multiple phosphorylation sites
Phosphorylation releases an inhibitory intramolecular interaction, allowing the ETS domain to effectively engage with DNA
Protein-protein interactions:
Interestingly, while phosphorylation enhances DNA binding, GST pull-down assays indicate that interaction with SRF is not significantly affected by phosphorylation status
This suggests that phosphorylation primarily regulates ELK1 function through conformational effects rather than by directly modulating protein partner selection
These insights provide a molecular framework for understanding how phosphorylation events translate into functional outputs in transcriptional regulation mediated by ELK1.
Cell-based ELISA methodologies offer unique advantages for studying phospho-ELK1 (S389) in intact cellular contexts:
Quantitative capabilities:
Cell-based ELISAs provide precise quantification of phosphorylation levels in response to different stimuli
Results can be normalized to total cell number using crystal violet staining, enabling adjustment for plating differences
This approach allows for direct comparison of phosphorylation levels across different experimental conditions
High-throughput applications:
Experimental workflow:
Cells are cultured directly in 96-well plates
Following stimulation, cells are fixed and permeabilized
Detection uses target-specific primary antibodies against phospho-ELK1 (S389) followed by HRP-conjugated secondary antibodies
Colorimetric measurement of HRP activity provides quantitative readout of phosphorylation levels
Multiplexed analysis potential:
Parallel wells can be probed for multiple signaling components
This allows for comprehensive pathway mapping by simultaneously monitoring phosphorylation of ELK1 alongside upstream kinases and downstream effectors
Relative activation levels of different pathway components can reveal bottlenecks or critical nodes in signal transduction
When implementing cell-based ELISAs, researchers should optimize cell density, fixation conditions, and antibody concentrations to ensure maximum sensitivity and specificity for phospho-ELK1 (S389) detection.
Integrating phospho-ELK1 (S389) analysis with transcriptomic approaches provides a comprehensive view of signal-dependent gene regulation:
Temporal correlation analyses:
Monitor phospho-ELK1 (S389) levels alongside expression of known ELK1 target genes
Establish time-lag relationships between peak phosphorylation and maximum transcriptional output
This temporal mapping can reveal the kinetic relationship between signaling events and gene expression changes
ChIP-seq integration:
Combine phospho-ELK1 (S389) quantification with chromatin immunoprecipitation sequencing
Determine whether phosphorylation status correlates with genomic occupancy patterns
Identify consensus binding motifs for phosphorylated versus non-phosphorylated ELK1
Multi-omics experimental design:
Collect matched samples for phospho-protein analysis and RNA-seq from the same experimental conditions
Use statistical approaches to correlate phosphorylation dynamics with transcriptional profiles
Employ network analysis to identify phospho-ELK1 (S389)-dependent gene modules
Functional validation strategies:
Express phosphomimetic (S389D/E) or phospho-deficient (S389A) ELK1 mutants
Compare resulting gene expression profiles to identify phosphorylation-dependent transcriptional programs
Validate key regulatory relationships using reporter assays or CRISPR-based approaches
This integrated approach allows researchers to move beyond correlative observations to establish causal relationships between ELK1 phosphorylation states and specific transcriptional outcomes.
Investigating interactions between phospho-ELK1 (S389) and chromatin regulators requires specialized approaches:
Co-immunoprecipitation strategies:
Immunoprecipitate phospho-ELK1 (S389) from nuclear extracts using phospho-specific antibodies
Identify associated chromatin modifiers through immunoblotting or mass spectrometry
Compare interaction profiles between phosphorylated and non-phosphorylated states
Consider crosslinking approaches to capture transient interactions
Proximity ligation assays:
Visualize in situ interactions between phospho-ELK1 (S389) and chromatin regulators
Quantify interaction dynamics following pathway activation
This technique is particularly valuable for detecting interactions in their native chromatin context
ChIP-sequential approaches:
Perform sequential ChIP first with phospho-ELK1 (S389) antibodies followed by antibodies against chromatin modifiers
Identify genomic loci where both proteins co-occupy
This technique can reveal the subset of ELK1 target genes that undergo specific chromatin modifications
FRET-based interaction analysis:
Generate fluorescently tagged ELK1 with phosphomimetic mutations
Measure energy transfer between tagged ELK1 and chromatin regulators
This approach can provide kinetic information about complex formation and dissociation
The phosphorylation-induced conformational change in ELK1 likely creates binding surfaces for specific chromatin regulators, representing a direct mechanism linking signaling events to epigenetic modulation at target genes.
Cutting-edge approaches for real-time monitoring of phospho-ELK1 (S389) in living systems include:
Phospho-specific biosensors:
Engineered FRET-based sensors incorporating ELK1 domains and phospho-binding modules
These sensors undergo conformational changes upon phosphorylation, generating measurable signals
Enable real-time visualization of phosphorylation events in single cells with high temporal resolution
Proximity-based labeling approaches:
TurboID or APEX2 fusion proteins to identify the dynamic interactome of phosphorylated ELK1
Temporal control allows mapping of interaction partners at different stages of phosphorylation
This technique can reveal how phosphorylation at Ser389 reconfigures the molecular environment of ELK1
Live-cell phospho-protein imaging:
Antibody-based detection using cell-permeable nanobodies against phospho-ELK1 (S389)
Genetically encoded phospho-specific intrabodies that selectively recognize phosphorylated epitopes
These approaches enable tracking of endogenous phospho-ELK1 (S389) in living cells without overexpression artifacts
Optogenetic control of ELK1 phosphorylation:
Light-activatable kinases or phosphatases targeting ELK1
Enables precise spatiotemporal control of phosphorylation status
Allows direct testing of the functional consequences of Ser389 phosphorylation with minimal pathway cross-activation
These emerging technologies promise to overcome limitations of traditional biochemical approaches by providing dynamic, spatially resolved information about phosphorylation events within their native cellular context.
Single-cell approaches to studying phospho-ELK1 (S389) can reveal important insights about signaling heterogeneity:
Mass cytometry (CyTOF) applications:
Metal-conjugated antibodies against phospho-ELK1 (S389) enable quantification at the single-cell level
Simultaneous measurement of multiple phospho-proteins reveals correlations between different pathway components
Clustering approaches can identify distinct cell states based on phosphorylation profiles
This technique is particularly valuable for analyzing heterogeneous tissues or mixed cell populations
Single-cell phospho-proteomics:
Emerging techniques for phospho-protein analysis at the single-cell level
Can reveal cell-to-cell variability in ELK1 phosphorylation patterns
Potential to identify rare cell populations with unique phosphorylation signatures
Spatial considerations:
Imaging mass cytometry or multiplexed immunofluorescence to map phospho-ELK1 (S389) distribution
Analysis of spatial relationships between phospho-ELK1 positive cells and tissue microenvironments
This approach can reveal how local signaling niches influence ELK1 phosphorylation states
Integrated multi-modal analysis:
Combine single-cell transcriptomics with phospho-protein detection
Establish direct links between phosphorylation status and gene expression programs at the single-cell level
This integrated view can elucidate how signaling heterogeneity translates to transcriptional diversity
Understanding cellular heterogeneity in phospho-ELK1 (S389) responses could reveal new insights into differential cellular responses to the same stimulus and identify subpopulations with unique signaling behaviors relevant to development or disease.