Phospho-HDAC8 (Ser39) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

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) .

Functional Role of HDAC8 Phosphorylation at Ser39

Phosphorylation of HDAC8 at Ser39 has been linked to critical cellular mechanisms:

Functional ImpactMechanismReferences
Enzyme InhibitionPKA-mediated phosphorylation reduces HDAC8’s deacetylase activity, leading to histone H3/H4 hyperacetylation .
Substrate InteractionPhosphorylated HDAC8 binds hEST1B (human ever-shorter telomeres 1B), stabilizing it against ubiquitin-mediated degradation .
Cancer RelevanceHDAC8 phosphorylation influences tumor cell proliferation, metastasis, and drug resistance .

Cancer Studies

  • 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 .

Cell Signaling Pathways

  • 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 .

Key Validation Studies

  • 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 .

Product Specs

Form
Supplied at a concentration of 1.0mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, containing 150mM NaCl, 0.02% sodium azide, and 50% glycerol.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on your location and shipping method. Please contact your local distributor for specific delivery details.
Synonyms
CDA07 antibody; CDLS5 antibody; HD 8 antibody; HD8 antibody; HDAC 8 antibody; Hdac8 antibody; HDAC8_HUMAN antibody; HDACL 1 antibody; HDACL1 antibody; Histone deacetylase 8 antibody; Histone deacetylase like 1 antibody; MRXS6 antibody; RPD 3 antibody; RPD3 antibody; WTS antibody
Target Names
Uniprot No.

Target Background

Function
Histone Deacetylase 8 (HDAC8) is responsible for removing acetyl groups from lysine residues on the N-terminal tails of core histones (H2A, H2B, H3, and H4). This deacetylation process is a key regulatory mechanism in epigenetic repression, influencing gene expression, cell cycle progression, and developmental processes. HDAC8 functions within large multiprotein complexes. Additionally, it plays a role in the deacetylation of SMC3, a protein component of the cohesin complex, regulating the release of cohesin complexes from chromatin. HDAC8 is also implicated in smooth muscle cell contractility.
Gene References Into Functions
  1. A de novo missense mutation in HDAC8 has been identified that leads to overlapping features of Rett-related disorder and Cornelia de Lange syndrome. This mutation potentially affects MECP recruitment, which may contribute to the development of Rett-related disorder. PMID: 29519750
  2. This study identified a somatic HDAC8 mutation in an individual with Cornelia de Lange Syndrome (CdLS) features, expanding our understanding of the genetic basis of CdLS. PMID: 29279609
  3. A novel missense mutation (c.806T>G, p.I269R) in the HDAC8 gene has been associated with CdLS, further expanding the range of known pathogenic mutations associated with this condition. PMID: 29991052
  4. A chemical crosslinking approach has identified novel HDAC8-specific substrates with high catalytic efficiency, providing a valuable strategy for discovering additional deacetylase substrates. PMID: 29035536
  5. Co2+ and Zn2+ are the most potent metal ions for HDAC8 biocatalysis, followed by Fe2+. Manganese and magnesium ions exhibit the lowest activity. PMID: 26996235
  6. Mesenchymal stem cells (MSCs) from patients with myeloproliferative neoplasms (MPNs) exhibit higher HDAC8 expression than normal MSCs. Inhibition of HDAC8 expression using specific inhibitors reduces the ability of the stroma to support hematopoietic cells from MPN patients, suggesting HDAC8 as a potential therapeutic target in MPN. PMID: 28390197
  7. HDAC8 activity regulates susceptibility to cell cycle arrest induced by Anthrax Lethal Toxin by modulating the PI3K-PTEN-AKT signaling axis. PMID: 28509866
  8. The aryl hydrocarbon receptor (AHR) directly targets and activates HDAC8 expression in hepatocellular carcinoma cells, influencing cell proliferation and tumorigenesis. PMID: 27283490
  9. The Epac-Rap1-Akt pathway mediates the inhibition of JNK-dependent HDAC8 degradation induced by cAMP signaling. This increase in HDAC8 levels enhances cisplatin-induced apoptosis in H1299 lung cancer cells by suppressing TIPRL expression. PMID: 28232663
  10. The X-ray crystal structure of HDAC8 in complex with an inhibitor reveals the formation of a unique subpocket within HDAC8. PMID: 27374062
  11. Evidence suggests that HDAC8 may undergo metal switching in vivo, potentially regulating its activity. Further studies are needed to investigate the identity of the metal ion bound to HDAC8 under various cellular conditions. PMID: 28937750
  12. The crystal structure of the HDAC8-trapoxin A complex at 1.24 A resolution reveals that the ketone moiety of l-Aoe undergoes nucleophilic attack to form a zinc-bound tetrahedral gem-diolate, mimicking the tetrahedral intermediate and transition states involved in catalysis. PMID: 28846375
  13. MicroRNA-455-3p plays a crucial role in chondrogenesis by directly targeting HDAC2/8 and promoting histone H3 acetylation. PMID: 27638301
  14. 7-amino-4-methylcoumarin (AMC) significantly enhanced KDAC6 substrate affinity and reduced Sirt1 activity, while eliminating the substrate sequence specificity of KDAC4. AMC had no consistent effect on KDAC8 substrates. PMID: 28749131
  15. Enzymological, biophysical, and structural studies of HDAC8 mutants associated with Cornelia de Lange Syndrome have provided insights into compromised catalysis in vitro. Most CdLS HDAC8 mutations induce structural changes that directly or indirectly impact substrate binding and catalysis. PMID: 27576763
  16. Class I HDACs (HDAC1, 2, 3, and 8) play a significant role in regulating the extracellular matrix and epithelial-mesenchymal transition, while class IIa HDACs (HDAC4 and 5) are less effective. PMID: 27420561
  17. Mutations G304A and G305A in histone deacetylase 8 (HDAC8) result in decreased catalytic efficiency. PMID: 27933794
  18. Sequencing of HDAC8 is recommended as an essential part of routine molecular diagnostics for patients with CdLS or CdLS-overlapping features. PMID: 26671848
  19. HDAC8 is overexpressed in oral squamous cell carcinoma tissues, primarily localized in the cytoplasm. PMID: 28004115
  20. HDAC8 can bind and deacetylate a variety of acetylated peptides derived from cellular, non-histone proteins, expanding our understanding of its functional role in cells. PMID: 26933971
  21. HDAC8 inhibition holds therapeutic potential to suppress Notch1 signaling in breast cancer. PMID: 26625202
  22. HDAC8 inhibition leads to the accumulation of acetylated-SMC3 protein without affecting the transcription of estrogen-responsive genes. PMID: 27072133
  23. Direct evidence indicates that potential KDAC8 substrates can be deacetylated by KDAC8. PMID: 26402585
  24. A study elucidates an HDAC8-mediated mechanism that inactivates p53, promoting leukemia stem cell activity. PMID: 26387755
  25. Aberrant deacetylation of p53 by HDAC8 contributes to the transformation and maintenance of leukemia stem cells (LSCs). PMID: 26923266
  26. HDAC8 plays a significant role in promoting gastric cancer tumorigenesis, highlighting its potential as a therapeutic target for this malignancy. PMID: 26412386
  27. The H143A and H142A/H143A mutants exhibit activity that is >80000-fold lower than wild-type HDAC8. The buried D176N and D176A mutants have significant catalytic effects, with more subtle effects caused by D183N and D183A. PMID: 26806311
  28. cAMP signaling increases HDAC8 protein levels by reducing JNK-mediated autophagy and ubiquitin-proteasome-dependent degradation of HDAC8 in H1299 lung cancer cells. PMID: 26792731
  29. HDAC8 inhibition induced cell growth inhibition and marked S-phase cell cycle arrest in human and murine-derived MPNST cells. PMID: 26200462
  30. Five HDAC8 mutants (P91L, G117E, H180R, D233G, and G304R) and double mutants (D233G-Y306F and P91L-Y306F) have been prepared and biophysically evaluated to enable cocrystallization of intact enzyme-substrate complexes. PMID: 26463496
  31. HDAC8 is elevated in BRAF-mutated melanoma. Increased cytoplasmic HDAC8 immunoreactivity is independently associated with improved survival from both diagnosis of primary melanoma and from first detection of stage IV disease to melanoma death. PMID: 25836739
  32. Findings highlight how HDAC8 drives nonalcoholic fatty liver disease-associated hepatocarcinogenesis. PMID: 26383163
  33. MicroRNA-21-3p regulates HDAC8 expression and the Akt/Gsk3beta pathway in cardiac hypertrophy. PMID: 25504627
  34. A study investigated the kinetics, thermodynamics, and selectivity of Zn(II) and Fe(II) binding to HDAC8. PMID: 25516458
  35. HDAC8 X-ray crystal structures reveal that each Cornelia de Lange syndrome mutation causes local structural changes that compromise catalysis and/or thermostability. PMID: 25075551
  36. The X-ray crystal structures of HDAC8 complexed with three synthetic analogues of Largazole, where the depsipeptide ester is replaced with a rigid amide linkage, have been reported. PMID: 25793284
  37. On the BMF gene promoter, loss of HDAC8 is associated with signal transducer and activator of transcription 3 (STAT3)/specificity protein 3 (Sp3) transcription factor exchange and recruitment of p300. PMID: 25321483
  38. Mechanistic and cellular studies on HDAC8 activation provide insights into the development of novel anticancer drugs. PMID: 25605725
  39. HDAC8 deregulation is frequent in urothelial cancer, but specific pharmacological inhibition or siRNA-mediated knockdown of HDAC8 did not significantly impair viability of urothelial cancer cell lines in a therapeutically useful manner. PMID: 25011684
  40. HDAC8 inhibition reduces gene expression and production of proinflammatory cytokines in vitro and in vivo. PMID: 25451941
  41. Detailed thermodynamic studies have been conducted on the binding of structurally similar ligands, varying in their "cap", "linker", and "metal-binding" regions of the suberoylanilide hydroxamic acid pharmacophore, to HDAC8. PMID: 25407689
  42. Loss-of-function mutations in HDAC8 cause a range of overlapping phenotypic features resembling Cornelia de Lange syndrome. PMID: 24403048
  43. In response to contractile stimulation, HDAC8 may mediate cortactin deacetylation, promoting actin filament polymerization and smooth muscle contraction. PMID: 24920679
  44. DEC1 coordinates with HDAC8 to differentially regulate TAp73 and DeltaNp73 expression. PMID: 24404147
  45. Urothelial cancer cell lines (UCCs) often exhibit up-regulation of HDAC2 and/or HDAC8. PMID: 22944197
  46. Molecular dynamics simulations have revealed a mechanism by which interactions and dynamics of two loops influence the configuration of functionally important residues of HDAC8, potentially affecting its activity. PMID: 24171457
  47. HDAC8 is overexpressed in hepatocellular carcinoma. Knockdown of HDAC8 suppresses tumor growth and enhances apoptosis. PMID: 24077923
  48. HDAC8 plays a crucial role in the modulation of SOCS1 and SOCS3 by curcumin. PMID: 23430957
  49. SOX4 is a direct target gene of FRA-2 and induces expression of HDAC8 in adult T-cell leukemia/lymphoma. PMID: 23482931
  50. Knockdown of HDAC8 leads to increased expression of SOCS1 and SOCS3. Overexpression of SOCS1 and SOCS3 significantly inhibits cell growth and suppresses JAK2/STAT signaling. PMID: 23111066

Show More

Hide All

Database Links

HGNC: 13315

OMIM: 300269

KEGG: hsa:55869

STRING: 9606.ENSP00000362674

UniGene: Hs.310536

Involvement In Disease
Cornelia de Lange syndrome 5 (CDLS5); Wilson-Turner X-linked mental retardation syndrome (WTS)
Protein Families
Histone deacetylase family, HD type 1 subfamily
Subcellular Location
Nucleus. Cytoplasm. Note=Excluded from the nucleoli. Found in the cytoplasm of cells showing smooth muscle differentiation.
Tissue Specificity
Weakly expressed in most tissues. Expressed at higher level in heart, brain, kidney and pancreas and also in liver, lung, placenta, prostate and kidney.

Q&A

What is HDAC8 and why is its phosphorylation at Serine 39 biologically significant?

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

  • Chromatin structure modification

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 .

How can I determine the optimal working concentration for Phospho-HDAC8 (Ser39) antibodies in different applications?

The optimal working concentration varies by application and must be empirically determined. Here are evidence-based recommendations based on manufacturer guidelines:

ApplicationRecommended Dilution RangeOptimization Approach
Western Blot1:500-1:2000Begin with 1:1000 and adjust based on signal-to-noise ratio
Immunohistochemistry1:50-1:300Start with 1:100 for paraffin sections
Immunofluorescence1:50-1:500Begin with 1:100 for fixed cells
ELISA1:20000Follow 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 .

What experimental controls should be included when using Phospho-HDAC8 (Ser39) antibodies?

Proper experimental controls are essential for interpreting results obtained with phospho-specific antibodies:

Positive Controls:

  • 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

Negative Controls:

  • 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

Specificity Controls:

  • 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.

What are the best methods for studying the dynamics of HDAC8 phosphorylation in living cells?

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:

    • Incubate cells with 32P-orthophosphate (0.5 mCi/ml for 4 hours)

    • Add PKA activators/inhibitors at different time points

    • Immunoprecipitate HDAC8 and analyze by SDS-PAGE and autoradiography

  • Phospho-HDAC8 (Ser39) cell-based ELISA:

    • Use colorimetric cell-based ELISA kits specific for Phospho-HDAC8 (Ser39)

    • Plate cells directly in 96-well format

    • Apply treatments and quantify phosphorylation in a high-throughput manner

  • 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.

How does HDAC8 phosphorylation at Ser39 affect its enzymatic activity and substrate specificity?

Phosphorylation of HDAC8 at Ser39 significantly impacts its enzymatic function:

Substrate Preference Shifts:

Research indicates that phosphorylation alters HDAC8 substrate specificity:

SubstrateEffect of Ser39 Phosphorylation
Histone H3Significantly reduced deacetylation
Histone H4Significantly reduced deacetylation
SMC3 (cohesin complex)Less pronounced effect on deacetylation
Non-histone substratesVariable 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 .

What are the critical parameters for optimizing immunohistochemistry protocols with Phospho-HDAC8 (Ser39) antibodies?

Optimizing immunohistochemistry (IHC) with phospho-specific antibodies requires careful attention to several parameters:

Tissue Preparation:

  • 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

Antigen Retrieval:

  • 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

Blocking and Antibody Incubation:

  • Block with 5-10% normal serum from the species of the secondary antibody

  • Include 1% BSA to reduce non-specific binding

  • Primary antibody dilution: Start at 1:100-1:300

  • Incubate at 4°C overnight for optimal sensitivity

Signal Detection:

  • 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

Critical Controls:

  • 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.

What is the relationship between HDAC8 phosphorylation and subcellular localization?

The phosphorylation status of HDAC8 at Ser39 influences its subcellular distribution, with important implications for its function:

Normal Localization Pattern:

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 .

Effects of Phosphorylation:

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

Experimental Approaches to Study Localization:

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

Functional Implications:

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.

How can I troubleshoot weak or absent signals when using Phospho-HDAC8 (Ser39) antibodies?

When encountering weak or absent signals with Phospho-HDAC8 (Ser39) antibodies, systematically address these potential issues:

Sample Preparation 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-Related Issues:

  • Antibody concentration:

    • Try different dilutions; for Western blot, test 1:500, 1:1000, and 1:2000

    • For immunostaining applications, use more concentrated antibody (1:50-1:100)

  • 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

Protocol Optimization:

  • 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.

How can phosphorylation of HDAC8 at Ser39 affect its interactions with protein complexes?

HDAC8 phosphorylation at Ser39 modulates its ability to participate in multi-protein complexes, with significant functional consequences:

Altered Protein-Protein Interactions:

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

Specific Complex Alterations:

Research indicates several impacts on HDAC8's participation in protein complexes:

Complex/Interaction PartnerEffect of Ser39 Phosphorylation
Transcriptional co-repressorsReduced association
SMC3 (cohesin complex)Altered interaction affecting cohesin release from chromatin
PP1 phosphatasePotential recruitment for dephosphorylation
14-3-3 proteinsPossible recruitment to phosphorylated HDAC8

Experimental Approaches to Study Complex Formation:

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.

What are the best approaches for quantifying changes in HDAC8 phosphorylation levels?

Accurate quantification of HDAC8 phosphorylation requires careful selection of methods and rigorous normalization:

Western Blot Quantification:

  • 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

ELISA-Based Quantification:

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

  • Calculate normalized phosphorylation ratio

Mass Spectrometry Approaches:

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

In-Cell Quantification:

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.

How does HDAC8 phosphorylation at Ser39 affect chromatin remodeling and gene expression?

The phosphorylation of HDAC8 at Ser39 has profound effects on chromatin structure and gene expression patterns:

Direct Effects on Histone Deacetylation:

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

Genome-Wide Effects:

Studies indicate that HDAC8 phosphorylation affects specific gene sets:

Gene CategoriesResponse to HDAC8 Phosphorylation
cAMP-responsive genesEnhanced expression due to reduced HDAC8 activity
Cell cycle regulatorsAltered expression patterns
Developmental genesContext-specific changes in expression
Smooth muscle-specific genesModified expression affecting contractility

Experimental Approaches to Study Transcriptional Effects:

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.

What considerations are important when validating the specificity of Phospho-HDAC8 (Ser39) antibodies?

Thorough validation of phospho-specific antibodies is critical for research integrity and involves several complementary approaches:

Essential Validation Experiments:

  • 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:

    • Test antibody reactivity in HDAC8 knockout/knockdown cells

    • Compare wild-type HDAC8 with S39A mutant expression

    • Phospho-specific signal should be absent in S39A mutant samples

  • PKA modulation:

    • Treat cells with forskolin to enhance phosphorylation

    • Treat cells with H-89 or PKI to inhibit phosphorylation

    • Verify signal response mirrors expected phosphorylation changes

Advanced Validation Approaches:

  • Mass spectrometry correlation:

    • Quantify Ser39 phosphorylation by MS

    • Compare with antibody-based quantification

    • Establish correlation between methods

  • Orthogonal antibody comparison:

    • Test multiple phospho-specific antibodies from different vendors

    • Compare detection patterns across applications

    • Consistent results increase confidence in specificity

These rigorous validation steps ensure that experimental observations genuinely reflect HDAC8 phosphorylation status rather than artifacts or cross-reactivity.

How can I design experiments to investigate the relationship between HDAC8 phosphorylation and cellular physiology?

Designing experiments to link HDAC8 phosphorylation to cellular functions requires a multifaceted approach:

Genetic Models:

  • HDAC8 phospho-mutant expression:

    • Generate stable cell lines expressing:

      • Wild-type HDAC8

      • Phospho-resistant HDAC8 (S39A)

      • Phospho-mimetic HDAC8 (S39E)

    • Compare phenotypes under normal and stress conditions

  • CRISPR-Cas9 genome editing:

    • Create cell lines with endogenous HDAC8-S39A knock-in

    • Compare with wild-type cells following PKA activation

Pharmacological Approaches:

  • PKA pathway modulation:

    • Activators: Forskolin (5-10 μM), 8-Br-cAMP, dibutyryl-cAMP

    • Inhibitors: H-89 (5-10 μM), PKI (10-20 μM)

    • Time-course and dose-response experiments

  • Combined HDAC and PKA inhibition:

    • Compare HDAC8-specific inhibitors alone vs. with PKA modulators

    • Assess additive or synergistic effects

Functional Assays:

Cellular ProcessExperimental Approach
Cell cycle regulationFlow cytometry, BrdU incorporation, proliferation assays
Smooth muscle contractilityTraction force microscopy, contractility assays
Chromatin structureATAC-seq, MNase-seq, histone PTM analysis
Gene expressionRNA-seq, qRT-PCR of HDAC8-regulated genes
Protein complex formationCo-IP, proximity labeling, BiFC assays

Systems Biology Integration:

  • 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.

What are the technical considerations for phospho-proteomic analysis of HDAC8 phosphorylation?

Mass spectrometry-based phosphoproteomic analysis of HDAC8 requires specific technical considerations to ensure accurate identification and quantification:

Sample Preparation:

  • 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

MS Analysis Parameters:

  • Peptide coverage considerations:

    • Multiple proteases beyond trypsin (Lys-C, Glu-C) to generate diverse peptides

    • The Ser39-containing peptide is within residues 5-54 of human HDAC8

    • Ensure detection of peptides containing Ser39 site

  • 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

Quantification Approaches:

  • 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

Bioinformatic Analysis:

  • 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.

How can I integrate Phospho-HDAC8 (Ser39) research into broader epigenetic signaling studies?

Integrating HDAC8 phosphorylation research into comprehensive epigenetic studies requires connecting this specific modification to broader regulatory networks:

Multi-Level Regulatory Analysis:

  • 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

Systems Approach Implementation:

  • 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

Physiological Context Expansion:

Biological ContextIntegration Approach
DevelopmentStudy HDAC8 phosphorylation changes during differentiation processes
Cell cycleAnalyze phosphorylation dynamics across cell cycle phases
Stress responseExamine PKA-HDAC8 axis activation under various cellular stresses
Disease modelsInvestigate dysregulation in cancer, neurodegenerative disorders

Translational Research Connections:

  • Therapeutic targeting:

    • Investigate compounds that modulate HDAC8 phosphorylation

    • Compare with direct HDAC8 inhibitors

    • Analyze combination approaches targeting both activity and phosphorylation

  • Biomarker development:

    • Evaluate Phospho-HDAC8 (Ser39) as a potential biomarker using validated antibodies

    • Correlate with disease progression or treatment response

    • Develop tissue-specific detection protocols

This integrated approach positions HDAC8 phosphorylation research within the broader context of epigenetic regulation and signaling networks, enhancing its biological significance and translational potential.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.