Phospho-HDAC8 (Ser39) antibody is a rabbit polyclonal antibody that selectively recognizes HDAC8 when phosphorylated at Ser39. This post-translational modification is catalyzed by protein kinase A (PKA) and negatively regulates HDAC8’s deacetylase activity . Key attributes include:
Specificity: Detects endogenous HDAC8 only when phosphorylated at Ser39 .
Immunogen: Derived from the peptide sequence around Ser39 (R-A-S(p)-M-V) in human HDAC8 .
Applications: Validated for immunohistochemistry (IHC) and immunofluorescence (IF) .
Phosphorylation of HDAC8 at Ser39 has been linked to critical cellular mechanisms:
Cervical Cancer: HDAC8 overexpression in HeLa cells correlates with cytoplasmic localization and interaction with α-tubulin, promoting cell migration and proliferation .
Leukemia: HDAC8 phosphorylation modulates interactions with fusion proteins in acute myeloid leukemia .
PKA Regulation: Forskolin (PKA activator) increases Ser39 phosphorylation, while H-89 (PKA inhibitor) blocks it, providing a tool to study HDAC8 activity dynamics .
Substrate Identification: The antibody has been used in co-immunoprecipitation assays to identify HDAC8-binding partners like hEST1B .
In Vitro Kinase Assays: Confirmed PKA-dependent phosphorylation of HDAC8 at Ser39 using recombinant proteins .
Co-Immunoprecipitation: Demonstrated enhanced HDAC8-hEST1B interaction upon forskolin treatment .
Pathological Relevance: Linked HDAC8 phosphorylation to Cornelia de Lange Syndrome (CdLS) and infectious diseases .
HDAC8 (Histone deacetylase 8) catalyzes the deacetylation of lysine residues on the N-terminal part of core histones (H2A, H2B, H3, and H4). This deacetylation process provides a tag for epigenetic repression and plays crucial roles in:
Transcriptional regulation
Cell cycle progression
Developmental events
Smooth muscle cell contractility
Phosphorylation of HDAC8 at Serine 39 (Ser39) by cyclic AMP-dependent protein kinase A (PKA) functions as a negative regulatory mechanism. When phosphorylated at this site, HDAC8's deacetylase activity is substantially reduced, particularly toward histones H3 and H4 . This post-translational modification represents a key mechanism by which cells can rapidly modulate epigenetic regulation in response to cellular signaling events.
Research has demonstrated that PKA phosphorylates HDAC8 exclusively on serine residues, with Ser39 being the principal phosphoacceptor site. Forskolin treatment, which activates adenyl cyclase and increases cAMP levels, enhances HDAC8 phosphorylation, while PKA inhibitors such as H-89 block this phosphorylation .
The optimal working concentration varies by application and must be empirically determined. Here are evidence-based recommendations based on manufacturer guidelines:
| Application | Recommended Dilution Range | Optimization Approach |
|---|---|---|
| Western Blot | 1:500-1:2000 | Begin with 1:1000 and adjust based on signal-to-noise ratio |
| Immunohistochemistry | 1:50-1:300 | Start with 1:100 for paraffin sections |
| Immunofluorescence | 1:50-1:500 | Begin with 1:100 for fixed cells |
| ELISA | 1:20000 | Follow this dilution for plate coating |
To optimize antibody concentration:
Perform a dilution series experiment
Include appropriate positive controls (cells treated with forskolin to activate PKA)
Include negative controls (cells treated with PKA inhibitors like H-89)
For Western blotting, include HDAC8(S39A) mutant samples as specificity controls if possible
The antibody demonstrates reactivity with human, mouse, and rat samples, making it versatile for cross-species research applications .
Proper experimental controls are essential for interpreting results obtained with phospho-specific antibodies:
Cells or tissues treated with forskolin (0.5-10 μM for 30-45 minutes), which activates PKA and increases HDAC8 Ser39 phosphorylation
Recombinant GST-HDAC8 phosphorylated in vitro by purified PKA
Cells pre-treated with PKA inhibitors (H-89 at 5-10 μM or PKI at 10-20 μM)
HDAC8 knockout or knockdown cells
Cells expressing HDAC8(S39A) mutant, which cannot be phosphorylated at position 39
Peptide competition assay using the immunizing phosphopeptide
Dual detection with total HDAC8 antibody on parallel blots or through stripping and reprobing
Phosphatase treatment of cell lysates to remove phosphorylation
These controls help verify that the observed signal truly represents phosphorylated HDAC8 rather than non-specific binding or cross-reactivity with other phosphoproteins.
To investigate the temporal dynamics of HDAC8 phosphorylation:
Time-course experiments:
Treat cells with forskolin (5-10 μM) for varying durations (5, 15, 30, 45, 60, 120 minutes)
Harvest cells and analyze by Western blotting with Phospho-HDAC8 (Ser39) antibody
Plot the phosphorylation intensity normalized to total HDAC8 versus time
Pulse-chase experiments with radiolabeled phosphate:
Phospho-HDAC8 (Ser39) cell-based ELISA:
Live-cell imaging:
Express HDAC8 fused to a fluorescent protein
Use phospho-specific antibodies conjugated to different fluorophores in fixed cells at different timepoints
Analyze subcellular localization changes upon phosphorylation
This multifaceted approach enables robust characterization of the kinetics and cellular context of HDAC8 phosphorylation events.
Phosphorylation of HDAC8 at Ser39 significantly impacts its enzymatic function:
Research indicates that phosphorylation alters HDAC8 substrate specificity:
| Substrate | Effect of Ser39 Phosphorylation |
|---|---|
| Histone H3 | Significantly reduced deacetylation |
| Histone H4 | Significantly reduced deacetylation |
| SMC3 (cohesin complex) | Less pronounced effect on deacetylation |
| Non-histone substrates | Variable effects requiring further investigation |
The molecular basis for this selectivity involves:
Altered enzyme conformation affecting the substrate binding pocket
Potential changes in protein-protein interactions with cofactors
Modified subcellular localization (nuclear vs. cytoplasmic distribution)
Researchers can leverage phospho-mimetic mutants (S39E) and phospho-resistant mutants (S39A) to further dissect these mechanisms in experimental systems .
Optimizing immunohistochemistry (IHC) with phospho-specific antibodies requires careful attention to several parameters:
Use freshly prepared tissue sections when possible
For paraffin-embedded tissues, ensure rapid fixation (<24 hours in 10% neutral buffered formalin)
Consider using phosphatase inhibitors (sodium fluoride, sodium orthovanadate) in fixatives to preserve phosphorylation status
Heat-induced epitope retrieval in citrate buffer (pH 6.0) is recommended
Alternative: Tris-EDTA buffer (pH 9.0) if signal is weak
Optimize retrieval time (15-30 minutes) and temperature
Block with 5-10% normal serum from the species of the secondary antibody
Include 1% BSA to reduce non-specific binding
Incubate at 4°C overnight for optimal sensitivity
For DAB detection systems, optimize development time to prevent overdevelopment
For fluorescent detection, select fluorophores with minimal spectral overlap if performing multiplex staining
Consider tyramide signal amplification for detecting low-abundance phosphoproteins
Adjacent tissue sections treated with lambda phosphatase
Tissue from animals/cells treated with PKA activators and inhibitors
Peptide competition controls to verify specificity
This methodical approach ensures reliable detection of phosphorylated HDAC8 within the complex environment of tissue specimens.
The phosphorylation status of HDAC8 at Ser39 influences its subcellular distribution, with important implications for its function:
HDAC8 is typically found in both nuclear and cytoplasmic compartments, with exclusion from nucleoli. In cells showing smooth muscle differentiation, HDAC8 exhibits pronounced cytoplasmic localization .
Research indicates that PKA-mediated phosphorylation at Ser39 can alter this distribution pattern:
Phosphorylated HDAC8 may show increased cytoplasmic retention
The nuclear-cytoplasmic shuttling rate may be affected
Association with specific subcellular structures (chromatin, cytoskeleton) can change
To investigate these dynamics, researchers can employ:
Immunofluorescence with phospho-specific antibodies (recommended dilution 1:50-200)
Subcellular fractionation followed by Western blotting
Live-cell imaging of fluorescently tagged HDAC8 variants (wild-type, S39A, S39E)
Co-localization studies with nuclear envelope markers, chromatin markers, or cytoskeletal components
The altered localization upon phosphorylation contributes to HDAC8's reduced activity on nuclear histone substrates and may redirect its activity toward cytoplasmic substrates, representing a sophisticated regulatory mechanism beyond simple inhibition of catalytic activity.
When encountering weak or absent signals with Phospho-HDAC8 (Ser39) antibodies, systematically address these potential issues:
Phosphorylation loss during sample processing:
Add phosphatase inhibitors (50 mM NaF, 1 mM Na3VO4, 10 mM β-glycerophosphate) to all buffers
Keep samples cold throughout processing
Use fresh samples; avoid repeated freeze-thaw cycles
Insufficient PKA activation:
Verify forskolin activity with a known PKA substrate
Optimize forskolin concentration (5-10 μM) and treatment time (30-45 minutes)
Consider alternative PKA activators (8-Br-cAMP, dibutyryl-cAMP)
Antibody concentration:
Detection system sensitivity:
Switch to more sensitive detection (e.g., from colorimetric to chemiluminescent)
Try signal amplification systems (e.g., biotin-streptavidin)
Use higher-sensitivity substrates for Western blot detection
Cross-reactivity concerns:
Verify antibody lot performance with positive control lysates
Perform peptide competition assays to confirm specificity
Western blot transfer issues:
Optimize transfer conditions for proteins in the HDAC8 size range (~42 kDa)
Consider semi-dry vs. wet transfer methods
Verify transfer with reversible protein staining
Antigen retrieval for tissue sections:
Test different antigen retrieval methods (heat, enzymatic)
Extend antigen retrieval time for heavily fixed samples
This systematic troubleshooting approach addresses the most common reasons for weak phospho-specific antibody signals.
HDAC8 phosphorylation at Ser39 modulates its ability to participate in multi-protein complexes, with significant functional consequences:
Phosphorylation introduces a negatively charged phosphate group that can:
Disrupt existing protein-protein interactions
Create new binding sites for phospho-binding domain-containing proteins
Induce conformational changes affecting interaction surfaces
Research indicates several impacts on HDAC8's participation in protein complexes:
To investigate these dynamics:
Co-immunoprecipitation:
Immunoprecipitate with anti-HDAC8 or anti-phospho-HDAC8 antibodies
Compare binding partners between phosphorylated and non-phosphorylated states
Use phospho-mimetic (S39E) and phospho-resistant (S39A) mutants
Proximity labeling:
Fuse HDAC8 variants to BioID or APEX2
Identify differential interactors when HDAC8 is phosphorylated vs. non-phosphorylated
Crosslinking mass spectrometry:
Compare crosslinked complexes from cells with activated or inhibited PKA
Identify structural changes in complexes containing phosphorylated HDAC8
These methodological approaches reveal how phosphorylation serves as a molecular switch regulating HDAC8's participation in diverse cellular processes.
Accurate quantification of HDAC8 phosphorylation requires careful selection of methods and rigorous normalization:
Dual detection approach:
Probe replicate blots with phospho-specific and total HDAC8 antibodies
Calculate phospho-HDAC8/total HDAC8 ratio
Use appropriate loading controls (GAPDH, β-actin)
Quantification parameters:
Use chemiluminescence detection within the linear range
Capture multiple exposure times to ensure linearity
Use densitometry software (ImageJ, Image Studio Lite) for analysis
Phospho-HDAC8 (Ser39) colorimetric cell-based ELISA kits provide quantitative data:
Plate cells directly in 96-well format
Use dual detection of phospho-HDAC8 and total HDAC8
For absolute quantification:
Selected reaction monitoring (SRM):
Use isotopically labeled phosphopeptide standards
Monitor specific transitions for phosphorylated and non-phosphorylated HDAC8 peptides
Calculate stoichiometry of phosphorylation
Parallel reaction monitoring (PRM):
Targeted MS method with higher specificity
Identify and quantify phosphopeptides containing Ser39
Compare peak areas to synthetic standards
For spatial information:
Quantitative immunofluorescence with phospho-specific antibodies
Normalize signal to total HDAC8 staining
Analyze nuclear/cytoplasmic distribution ratio
These complementary approaches provide robust quantification of HDAC8 phosphorylation status under diverse experimental conditions.
The phosphorylation of HDAC8 at Ser39 has profound effects on chromatin structure and gene expression patterns:
When HDAC8 is phosphorylated at Ser39 by PKA, its deacetylase activity toward histone substrates is significantly reduced . This leads to:
Increased histone acetylation at HDAC8 target sites
More open chromatin structure at affected genomic regions
Enhanced accessibility for transcription factors
Studies indicate that HDAC8 phosphorylation affects specific gene sets:
To investigate these effects comprehensively:
ChIP-seq analysis:
Compare histone acetylation patterns in cells with wild-type vs. phosphomimetic HDAC8
Identify genomic regions with differential HDAC8 occupancy upon PKA activation
Correlate with gene expression changes
RNA-seq:
Profile transcriptome changes upon forskolin treatment
Compare with HDAC8 inhibitor treatment
Identify genes specifically regulated by HDAC8 phosphorylation
ATAC-seq:
Analyze chromatin accessibility changes linked to HDAC8 phosphorylation
Map open chromatin regions in PKA-activated vs. control cells
This multilayered approach provides insight into how HDAC8 phosphorylation contributes to signal-responsive gene regulation through chromatin-based mechanisms.
Thorough validation of phospho-specific antibodies is critical for research integrity and involves several complementary approaches:
Peptide competition assays:
Pre-incubate antibody with phosphorylated and non-phosphorylated peptides
A specific signal should be blocked only by the phospho-peptide
Use graduated concentrations to determine blocking efficiency
Phosphatase treatment controls:
Treat one portion of positive control lysate with lambda phosphatase
Compare Western blot signals between treated and untreated samples
Phospho-specific signal should disappear after phosphatase treatment
Genetic validation:
PKA modulation:
Mass spectrometry correlation:
Quantify Ser39 phosphorylation by MS
Compare with antibody-based quantification
Establish correlation between methods
Orthogonal antibody comparison:
These rigorous validation steps ensure that experimental observations genuinely reflect HDAC8 phosphorylation status rather than artifacts or cross-reactivity.
Designing experiments to link HDAC8 phosphorylation to cellular functions requires a multifaceted approach:
HDAC8 phospho-mutant expression:
CRISPR-Cas9 genome editing:
Create cell lines with endogenous HDAC8-S39A knock-in
Compare with wild-type cells following PKA activation
PKA pathway modulation:
Combined HDAC and PKA inhibition:
Compare HDAC8-specific inhibitors alone vs. with PKA modulators
Assess additive or synergistic effects
Multi-omics approach:
Integrate phosphoproteomics, acetylomics, and transcriptomics
Map signaling networks connecting HDAC8 phosphorylation to cellular responses
Identify pathway crosstalk mechanisms
Temporal dynamics analysis:
Time-resolved measurements following PKA activation
Pathway reconstruction based on kinetic parameters
Mathematical modeling of HDAC8 phosphorylation/dephosphorylation cycle
This comprehensive experimental design strategy enables robust investigation of HDAC8 phosphorylation's role in cellular physiology across multiple scales.
Mass spectrometry-based phosphoproteomic analysis of HDAC8 requires specific technical considerations to ensure accurate identification and quantification:
Enrichment strategies:
Immunoprecipitation with HDAC8-specific antibodies
Phosphopeptide enrichment methods (TiO2, IMAC, phospho-tyrosine antibodies)
Consider fractionation to reduce sample complexity
Preservation of phosphorylation:
Add phosphatase inhibitors to all buffers:
Sodium fluoride (50 mM)
Sodium orthovanadate (1 mM)
β-glycerophosphate (10 mM)
Maintain cold temperatures throughout processing
Rapid protein extraction and denaturation
Peptide coverage considerations:
Fragmentation methods:
HCD provides good coverage of phosphopeptides
ECD/ETD preserves labile phosphorylation during fragmentation
Consider combined fragmentation approaches
Detection strategies:
Targeted methods (PRM/SRM) for known phosphosites
Data-dependent acquisition for discovery
Data-independent acquisition for comprehensive coverage
Label-based quantification:
SILAC for cell culture experiments
TMT/iTRAQ for multiplexed analysis
Heavy-labeled synthetic phosphopeptide standards
Label-free quantification:
Spectral counting
MS1 intensity-based methods
MS2 fragment-based methods
Database search parameters:
Include phosphorylation (+79.9663 Da) as variable modification
Consider other potential PTMs (acetylation, ubiquitination)
Implement site localization algorithms (Ascore, ptmRS)
Validation criteria:
Manual verification of MS/MS spectra for Ser39 phosphopeptides
Require diagnostic ions confirming phosphorylation at Ser39 vs. nearby sites
Apply stringent FDR thresholds for phosphosite identification
These technical considerations ensure reliable phosphoproteomic analysis of HDAC8 phosphorylation at Ser39 and other potentially relevant sites.
Integrating HDAC8 phosphorylation research into comprehensive epigenetic studies requires connecting this specific modification to broader regulatory networks:
Signaling pathway integration:
Map PKA-HDAC8 axis within cAMP signaling networks
Identify crosstalk with other pathways affecting histone modifications
Analyze temporal coordination between phosphorylation and acetylation changes
Epigenetic crosstalk mapping:
Investigate how HDAC8 phosphorylation affects other epigenetic marks:
Histone acetylation at specific lysines (H3K9ac, H3K27ac)
DNA methylation patterns
Chromatin remodeling complex activity
Integrated multi-omics:
Combine data from:
Phosphoproteomics (HDAC8 and other components)
Acetylomics (histone and non-histone targets)
Transcriptomics (gene expression changes)
Chromatin accessibility (ATAC-seq, DNase-seq)
Network modeling:
Construct directed graphs connecting:
PKA activation → HDAC8 phosphorylation
HDAC8 phosphorylation → histone acetylation changes
Histone acetylation → transcription factor binding
Transcription factor binding → gene expression
| Biological Context | Integration Approach |
|---|---|
| Development | Study HDAC8 phosphorylation changes during differentiation processes |
| Cell cycle | Analyze phosphorylation dynamics across cell cycle phases |
| Stress response | Examine PKA-HDAC8 axis activation under various cellular stresses |
| Disease models | Investigate dysregulation in cancer, neurodegenerative disorders |
Therapeutic targeting:
Investigate compounds that modulate HDAC8 phosphorylation
Compare with direct HDAC8 inhibitors
Analyze combination approaches targeting both activity and phosphorylation
Biomarker development:
This integrated approach positions HDAC8 phosphorylation research within the broader context of epigenetic regulation and signaling networks, enhancing its biological significance and translational potential.