Acetyl-HIST1H3A (K64) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Characteristics of Acetyl-HIST1H3A (K64) Antibody

The Acetyl-HIST1H3A (K64) Antibody is a rabbit recombinant monoclonal antibody (clone EPR20713) designed to specifically recognize histone H3 acetylated at lysine 64 (H3K64ac). It is a critical tool for studying chromatin dynamics, transcriptional regulation, and epigenetic mechanisms. Key features include:

PropertyDetails
ClonalityMonoclonal (rabbit)
ImmunogenSynthetic peptide acetylated at lysine 64 of histone H3
ReactivityHuman, Mouse, Rat, Synthetic peptides
ApplicationsChIP, Western blot (WB), Immunocytochemistry (ICC), Peptide array, ChIP-seq
IsotypeIgG
ConjugationCarrier-free (unconjugated) or conjugation-ready formats available

This antibody demonstrates high specificity for H3K64ac, as validated through peptide competition assays and limited tryptic digestion experiments, which confirmed its ability to detect core histone acetylation without cross-reactivity to other histone modifications (e.g., H3K9ac, H3K18ac) or methylated states (e.g., H3K64me3) .

Key Research Applications and Findings

The Acetyl-HIST1H3A (K64) Antibody has been instrumental in elucidating the functional role of H3K64ac in chromatin biology. Below are its major applications and associated research insights:

Chromatin Immunoprecipitation (ChIP) and ChIP-seq

  • Nucleosome Stability and Transcriptional Activation: H3K64ac is enriched at transcriptional start sites (TSS) of active genes, where it facilitates nucleosome eviction and chromatin remodeling. This modification opposes the repressive H3K64me3 mark, creating a dynamic equilibrium between active and silent chromatin states .

  • Enrichment Patterns:

    Gene TypeH3K64ac EnrichmentExample Genes
    Pluripotency genesHigh in undifferentiated cellsNanog, Pou5f1, Dppa3
    Differentiation genesIncreased after retinoic acid treatmentHoxb3, Hoxd3, Pax6

Data derived from ChIP-seq analyses in embryonic stem cells .

Western Blot (WB)

  • Detection of H3K64ac:

    Cell LineTreatmentDilutionObserved Band
    HeLaUntreated1:100015 kDa
    HeLaTrichostatin A (400 ng/ml)1:1000Enhanced 15 kDa
    NIH/3T3Trichostatin A (500 ng/ml)1:500015 kDa

Note: Trichostatin A, an HDAC inhibitor, increases H3K64ac levels .

Immunofluorescence (ICC)

  • Nuclear Localization: H3K64ac exhibits a distinct nuclear staining pattern, contrasting with heterochromatin-poor regions. This localization is disrupted in chromatin remodeling experiments (e.g., Chd1-mediated nucleosome repositioning) .

Peptide Array Validation

The antibody was tested against 501 histone peptides, demonstrating high specificity for K64-acetylated peptides. Key results include:

Peptide TypeBinding AffinityCompetitors Tested
H3K64ac (full-length)HighUnmodified H3, H3K9ac, H3K64me3
H3K64ac (core region)RetainedN-terminal acetylation sites

Data from Abcam peptide array assays .

Functional Insights from H3K64ac Research

The Acetyl-HIST1H3A (K64) Antibody has enabled critical discoveries about H3K64ac’s role in chromatin function:

Nucleosome Dynamics

  • Stability Reduction: H3K64ac decreases nucleosome stability, as shown by salt-disruption assays and single-molecule FRET. This contrasts with H3K56ac, which has no measurable effect on stability .

  • Chromatin Remodeling: H3K64ac-acetylated nucleosomes are repositioned faster by Chd1 but not RSC remodelers, suggesting modification-dependent enzyme specificity .

Transcriptional Regulation

  • p300/CBP-Dependent Acetylation: The p300 co-activator directly acetylates H3K64, linking this modification to transcriptional activation. Knockdown of p300 reduces H3K64ac levels and disrupts its genomic distribution .

  • Opposition to H3K64me3: H3K64ac and H3K64me3 are mutually exclusive, with H3K64ac marking active alleles in imprinted loci .

Association with H3 Variants

H3K64ac is preferentially enriched on H3.3, a histone variant linked to active chromatin. Mutation of K64 to arginine (K64R) abolishes antibody recognition, confirming specificity .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. For specific delivery times, please contact your local distributors.
Synonyms
Histone H3.1 (Histone H3/a) (Histone H3/b) (Histone H3/c) (Histone H3/d) (Histone H3/f) (Histone H3/h) (Histone H3/i) (Histone H3/j) (Histone H3/k) (Histone H3/l), HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, H3FA, H3FL, H3FC, H3FB, H3FD, H3FI, H3FH, H3FK, H3FF, H3FJ
Target Names
Uniprot No.

Target Background

Function
Histone H3A is a core component of nucleosomes, playing a crucial role in DNA packaging and regulation. Nucleosomes are responsible for wrapping and compacting DNA into chromatin, limiting DNA accessibility to cellular machinery that utilizes DNA as a template. This regulation of DNA accessibility is critical for various cellular processes, including transcription, DNA repair, DNA replication, and chromosomal stability. Histones are regulated through a complex system of post-translational modifications, known as the histone code, which involves the modification of histone tails and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests a mechanism for epigenetic regulation in cancer involving the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Increased expression of H3K27me3 during a patient's clinical course may indicate the presence of heterochronous tumors. PMID: 29482987
  3. JMJD5, a Jumonji C (JmjC) domain-containing protein, has been identified as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that cause a DNA damage response. PMID: 28982940
  4. Data suggests that the Ki-67 antigen proliferative index has limitations, and phosphohistone H3 (PHH3) serves as an alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. In the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This series of 47 diffuse midline gliomas reveals that histone H3-K27M mutation is mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurs with BRAF-V600E mutation and is frequently associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research demonstrates that the histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral, and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments showed that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl) where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This research describes, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG) including GBM and DIPG harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not rare. PMID: 28547652
  14. Data show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest whereas the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate the formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. In contrast, histone 3 mutations do not seem to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis and repression of APAK. PMID: 25961932
  27. The authors verified that histone H3 is a real substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. Circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. MCM2 binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. De novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is the biological significance of H3K64 acetylation in chromatin regulation?

H3K64 acetylation represents a critical epigenetic modification located on the lateral surface of the histone octamer, in close proximity to the inner gyre of DNA. Unlike histone tail modifications, H3K64ac directly impacts nucleosome stability and dynamics. Research has demonstrated that this modification regulates chromatin accessibility by decreasing nucleosome stability, which facilitates nucleosome eviction and consequently promotes gene expression .

H3K64ac is particularly enriched at:

  • Transcriptional start sites (TSS) of active genes

  • Active enhancers, where it co-localizes with established enhancer marks such as H3K27ac, H3K4me1, and p300-binding

The functional significance of H3K64ac is highlighted by its mutually exclusive distribution with H3K64me3, a repressive mark associated with pericentromeric heterochromatin. At imprinted loci, the transcriptionally active alleles are specifically enriched in H3K64ac, whereas the inactive alleles are enriched in H3K64me3, suggesting these modifications define functionally opposing chromatin states .

How does H3K64ac differ from other histone H3 acetylation marks?

H3K64ac is distinct from commonly studied histone H3 acetylation marks in several key aspects:

FeatureH3K64acH3K9ac/H3K14ac/H3K27ac
LocationLateral surface of histone octamer (globular domain)N-terminal histone tail
Proximity to DNADirect contact with inner gyre of DNAExtends away from nucleosome core
Effect on nucleosomeDirectly affects nucleosome stability and assembly/disassembly dynamicsPrimarily affects recruitment of reader proteins
Impact on salt stabilityDecreases nucleosome stability by ~60mM NaCl compared to unmodified nucleosomesVariable effects, generally less direct impact on nucleosome structure
Enrichment patternTSS of active genes, active enhancersH3K9ac/K14ac at active promoters; H3K27ac at active enhancers
Associated histone variantHighest enrichment on H3.3 variantPresent on multiple H3 variants

The strategic location of H3K64ac on the lateral surface allows it to directly influence nucleosome biophysical properties, distinguishing it from tail modifications that primarily function through recruitment of effector proteins .

What are the recommended applications for Acetyl-HIST1H3A (K64) antibodies?

Based on the validation data, Acetyl-HIST1H3A (K64) antibodies have been successfully employed in the following applications:

ApplicationRecommended DilutionNotes
ELISAOptimal dilutions determined by end userHigh specificity for K64-acetylated peptide
IF/ICC1:50-1:500Distinct nuclear localization with relative depletion from heterochromatin
ChIP/ChIP-seq1:500-1:1000Successfully used for genome-wide profiling of H3K64ac
Western Blot1:500-1:2000Detects endogenous acetylated histone H3
Dot Blot1:1000For peptide specificity testing

When using these antibodies, it is critical to include appropriate controls to confirm specificity, particularly when investigating subtle changes in H3K64ac levels in different experimental conditions. The antibody's recognition of H3 is efficiently competed by K64ac immunizing peptide but not by other peptides containing acetylated, methylated, or unmodified histone regions .

How should I design ChIP-seq experiments to accurately profile H3K64ac genome-wide distribution?

When designing ChIP-seq experiments to profile H3K64ac, consider these critical methodological approaches:

Experimental Design Considerations:

  • Crosslinking optimization: Since H3K64ac is located within the nucleosome core, optimize formaldehyde crosslinking time (8-10 minutes typically works well) to ensure adequate fixation without overfixation.

  • Sonication parameters: Aim for fragments of 150-300bp to enhance resolution. Monitor fragmentation efficiency using a Bioanalyzer.

  • Controls to include:

    • Input DNA control

    • IgG control for background binding

    • H3 ChIP for normalization (to distinguish changes in H3K64ac from changes in nucleosome occupancy)

    • Spike-in normalization with foreign chromatin (e.g., Drosophila) for quantitative comparisons

  • Antibody validation: Perform peptide competition assays with K64-acetylated peptides versus other acetylated lysines to confirm specificity before ChIP-seq.

Data Analysis Guidelines:

  • Correlate H3K64ac enrichment with:

    • RNA Polymerase II occupancy

    • Active histone marks (H3K9ac, H3K4me2)

    • Repressive marks (H3K27me3, H3K64me3) for anti-correlation validation

  • Create meta-gene plots showing total H3 enrichment around TSSs grouped by expression level

  • Analyze enhancer regions separately, with special focus on correlations with H3K27ac, H3K4me1, and p300-binding

From published studies, H3K64ac shows strong enrichment at TSSs of active genes, with a correlation coefficient of >0.8 with RNA Pol II occupancy. This enrichment is indicative of the steady-state mRNA level of the respective gene .

What experimental approaches can determine the functional consequences of H3K64ac on chromatin dynamics?

To investigate the functional impact of H3K64ac on chromatin dynamics, researchers have employed several sophisticated experimental approaches:

Biophysical and Biochemical Approaches:

  • FRET measurements: Attach fluorescent dyes to specific sites on nucleosomal DNA (35bp from each end) and measure FRET signal changes during salt-dependent disruption. H3K64ac nucleosomes show decreased stability between ~0.5M and 1.0M NaCl concentrations.

  • Single-molecule FRET: For more precise measurement of nucleosome stability at the single-molecule level, confirming lower stability of H3K64ac nucleosomes.

  • ATP-dependent chromatin remodeling assays: Compare remodeling rates of H3K64ac versus unmodified nucleosomes with different remodeling enzymes (e.g., Chd1 shows faster repositioning of H3K64ac nucleosomes than unmodified ones, while RSC shows no difference).

  • Site-specific incorporation of acetylated lysine: Use genetic approaches to incorporate acetyl-lysine directly at position 64 in recombinant histone H3 for in vitro studies.

Cellular and In Vivo Approaches:

  • Expression of histone mutants: Compare wildtype H3.3, H3.3K64Q (acetylated lysine mimetic), and H3.3K64R (retains positive charge but is non-acetylatable) in cells.

  • Gene expression analysis: Measure the effect of these mutants on gene expression, particularly for genes involved in specific pathways (e.g., TPA-dependent early-response genes).

  • Chromatin accessibility assays: Use ATAC-seq or DNase-seq to determine if H3K64ac increases chromatin accessibility.

Data from such studies have shown that H3K64Q mutants can promote expression of certain genes (e.g., c-fos, Egr1, and c-myc) above levels obtained with wildtype H3.3 or acetylation-deficient H3.3K64R, demonstrating H3K64ac's intrinsic ability to impact transcription mechanisms in vivo .

How can I determine if p300/CBP is responsible for H3K64 acetylation in my experimental system?

To establish p300/CBP as H3K64 acetyltransferases in your experimental system, implement a multi-layered approach:

In Cellulo Approaches:

  • Knockdown experiments: Systematically deplete candidate HATs from different families and assess changes in H3K64ac levels by Western blot. In published studies, knockdown of p300 and CBP, but not other HATs, decreased the steady-state levels of H3K64ac.

  • ChIP analysis after knockdown: Perform ChIP with H3K64ac antibody after p300/CBP knockdown, focusing on p300/CBP-specific genomic target regions where the decrease should be most pronounced.

  • Overexpression studies: Overexpress p300 in your system and measure H3K64ac levels. Published data show increased levels of H3K64ac upon p300 overexpression.

  • Correlation analysis: Compare the genomic distribution of p300 binding sites with H3K64ac enrichment. Strong correlation would support a functional relationship.

In Vitro Biochemical Validation:

  • HAT assays with recombinant proteins: Purify recombinant p300/CBP and incubate with:

    • Free histone H3

    • Recombinant nucleosomes

    • Native chromatin isolated from cells

  • Mass spectrometry analysis: Confirm acetylation specifically at K64 rather than other residues by analyzing the products of the HAT reaction.

  • Inhibitor studies: Use specific p300/CBP inhibitors (e.g., C646, A-485) to confirm the specificity of the acetylation reaction both in vitro and in cells.

Previous research has confirmed that p300 and CBP can acetylate H3K64 in vitro on both free H3 and within chromatin, and that p300 and H3K64ac distributions show a strong correlation genome-wide .

How should I address potential cross-reactivity when using Acetyl-HIST1H3A (K64) antibodies?

Cross-reactivity is a significant concern when using histone modification antibodies due to sequence similarities around different lysine residues. To address this issue:

Antibody Validation Approaches:

  • Peptide competition assays: Test whether the antibody's recognition of H3 is competed by:

    • The immunizing K64ac peptide (should compete)

    • Other acetylated histone peptides (should not compete)

    • Methylated or unmodified histone regions (should not compete)

  • Limited tryptic digestion: Perform limited tryptic digestion of native nucleosomes, which removes the H3 tails while leaving the DNA-protected H3 core region largely intact. A true H3K64ac antibody will still recognize the truncated H3 core (unlike antibodies to tail modifications like H3K9ac, H3K18ac, and H3K27ac).

  • Knockout/mutation controls: Test the antibody on samples where:

    • H3K64 is mutated to arginine (K64R)

    • HATs responsible for H3K64ac (p300/CBP) are depleted or inhibited

Signs of Potential Cross-Reactivity:

  • Detection of bands at unexpected molecular weights in Western blots

  • Immunofluorescence patterns that don't match known H3K64ac distribution (nuclear with depletion from heterochromatin)

  • ChIP-seq profiles that correlate better with other histone marks than expected

In published studies, properly validated H3K64ac antibodies specifically detect endogenous acetylated histone H3 and recognize a K64-acetylated peptide with high specificity compared to other H3 acetylated lysines .

How can I interpret changes in H3K64ac levels in relation to gene expression and other histone modifications?

Interpreting H3K64ac changes requires consideration of its functional context and relationship with other epigenetic marks:

Integration with Gene Expression Data:

  • H3K64ac enrichment at TSSs positively correlates with gene expression levels.

  • When analyzing changes in H3K64ac, group genes by expression level to assess if H3K64ac changes correspond to transcriptional changes.

  • For genes showing discordant patterns (H3K64ac changes without expression changes), investigate compensatory mechanisms or additional regulatory factors.

Correlation with Other Histone Marks:
H3K64ac should be analyzed in relation to other marks:

CorrelationHistone ModificationBiological Interpretation
PositiveH3K9ac, H3K4me2Active transcription signatures
PositiveH3K27ac, H3K4me1, p300 bindingEnhancer activity
NegativeH3K27me3Polycomb-mediated repression
NegativeH3K64me3Heterochromatin formation

Imprinted Loci Analysis:

  • At imprinted control regions (ICRs), active alleles should be enriched in H3K64ac while inactive alleles should show H3K64me3 enrichment.

  • Disruption of this pattern may indicate compromised imprinting regulation.

Developmental/Differentiation Context:

  • During cellular differentiation, H3K64ac enrichment should shift from pluripotency genes to lineage-specific genes.

  • In mouse embryonic stem (ES) cells, H3K64ac is enriched at pluripotency genes (e.g., Nanog, Pou5f1, Dppa3).

  • After retinoic acid-induced differentiation, enrichment shifts towards differentiation-associated genes (e.g., Hoxb3, Hoxd3, Pax6).

When interpreting changes, consider that H3K64ac functions by directly affecting nucleosome stability rather than primarily through reader protein recruitment, distinguishing it from many other histone modifications .

What methodological considerations are important when analyzing H3K64ac in different cell types or disease models?

When analyzing H3K64ac across different biological contexts, consider these critical methodological aspects:

Cell Type Considerations:

  • Cell cycle synchronization: H3K64ac levels may vary during cell cycle progression, particularly during S-phase when new nucleosomes are assembled. Consider synchronizing cells when making comparisons.

  • Histone variant distribution: Since H3K64ac shows highest enrichment on H3.3 variant, cell types with different H3 variant compositions may show different baseline levels of H3K64ac.

  • p300/CBP activity: Variations in p300/CBP activity or cofactor availability between cell types will affect H3K64ac levels. Measure these factors when comparing cell types.

Disease Model Considerations:

  • Cancer models: Many cancers show altered p300/CBP activity or mutations. When analyzing H3K64ac in cancer models:

    • Account for copy number changes of histone genes

    • Consider competing modifications (H3K64me3)

    • Evaluate global versus gene-specific changes

  • Developmental disorders: For disorders with known chromatin dysregulation:

    • Focus on loci with established developmental regulation

    • Consider imprinted regions where H3K64ac and H3K64me3 show allele-specific patterns

    • Analyze enhancer regions where H3K64ac may mark active enhancers

Technical Normalization Approaches:

  • Internal normalization: Always normalize H3K64ac signal to total H3 levels to account for differences in nucleosome density.

  • Spike-in controls: Use spike-in chromatin from a different species (e.g., Drosophila) for quantitative comparisons across samples.

  • Multi-antibody validation: For critical findings, confirm results using independent antibodies against H3K64ac or orthogonal approaches like mass spectrometry.

Published research has shown that H3K64ac is present in various mouse and human cell lines and tissues, suggesting a ubiquitous function. HDAC-inhibitor treatment increases H3K64ac levels, which may be relevant when studying diseases treated with such inhibitors .

How can I optimize ChIP-qPCR protocols specifically for H3K64ac detection?

Optimizing ChIP-qPCR for H3K64ac requires attention to several parameters specific to this modification:

Sample Preparation Optimization:

  • Crosslinking conditions: Test multiple formaldehyde concentrations (1-1.5%) and fixation times (8-12 minutes) to optimize crosslinking without affecting epitope accessibility.

  • Chromatin preparation: Since H3K64 is within the nucleosome core and close to DNA, ensure thorough sonication to expose the epitope (verify fragment size of 200-300bp).

  • Antibody concentration: Titrate antibody amounts (2-10μg per ChIP) to determine optimal concentration that maximizes signal-to-noise ratio.

Control Region Selection:

Region TypeExpected H3K64acExamplesPurpose
Active promotersHighKnown housekeeping genesPositive control
EnhancersModerate-HighCell type-specific enhancersPositive control
Repetitive elementsVery lowSatellite repeatsNegative control
Inactive genesLowTissue-specific genes inactive in your cell typeNegative control

Protocol Adjustments:

  • Washing stringency: Optimize salt concentration in wash buffers (150-500mM NaCl) to reduce background while maintaining specific binding.

  • Elution conditions: For efficient elution of H3K64ac-bound chromatin, consider using SDS concentrations of 1-2% at 65°C.

  • qPCR primer design: Design primers to amplify 80-150bp regions, avoiding regions with repetitive sequences.

Validation Strategy:

  • Perform ChIP for total H3 in parallel to normalize H3K64ac signal

  • Include IgG control to establish background levels

  • Test regions with known H3K64ac enrichment (based on published ChIP-seq data)

  • Compare results with other active marks (H3K9ac, H3K27ac) as correlation controls

In published studies, H3K64ac ChIP-qPCR has been successfully used to validate genome-wide data and confirm enrichment at active promoters and enhancers, with very low levels at repetitive elements .

What is the relationship between H3K64ac and nucleosome stability, and how can I study this experimentally?

H3K64ac has been shown to decrease nucleosome stability, facilitating DNA accessibility and nucleosome eviction. To investigate this relationship:

Biophysical Approaches to Measure Nucleosome Stability:

  • Salt-dependent nucleosome stability assays: Reconstitute nucleosomes with recombinant H3K64ac (using genetic incorporation of acetyl-lysine) and measure stability across a salt gradient (0.2-2.0M NaCl). H3K64ac nucleosomes display decreased stability particularly between 0.5-1.0M NaCl.

  • FRET-based nucleosome stability assays: Attach fluorescent dyes to nucleosomal DNA ends to monitor DNA unwrapping. Technical approach:

    • Label DNA at positions ~35bp from each end

    • Reconstitute nucleosomes with labeled DNA and H3K64ac or unmodified H3

    • Measure FRET signal changes during salt titration or thermal denaturation

  • Single-molecule FRET: For higher resolution analysis of nucleosome dynamics:

    • Immobilize individual nucleosomes on microscope slides

    • Monitor real-time fluctuations in FRET efficiency

    • Compare "breathing" rates between H3K64ac and unmodified nucleosomes

Biochemical and Cellular Approaches:

  • Nucleosome assembly/disassembly assays: Compare rates of assembly/disassembly between H3K64ac and unmodified nucleosomes.

  • Chromatin remodeling assays: Test whether H3K64ac affects the activity of chromatin remodeling enzymes:

    • Reconstitute positioned nucleosomes with modified or unmodified H3

    • Incubate with remodelers like Chd1 or RSC

    • Analyze nucleosome repositioning by native PAGE

  • Histone eviction in cells: Express H3K64Q (acetylated lysine mimetic) or H3K64R (non-acetylatable) and measure:

    • Histone turnover rates using SNAP-tag pulse-chase

    • Chromatin accessibility by ATAC-seq

    • Transcription factor binding by ChIP

Research has shown that H3K64ac decreases nucleosome stability by approximately 60mM in salt-dependent stability assays compared to unmodified nucleosomes. This effect is distinct from H3K56ac, which was reported not to significantly affect nucleosome stability under comparable conditions .

How can I integrate H3K64ac ChIP-seq data with other genomic datasets to understand chromatin state dynamics?

Integrating H3K64ac ChIP-seq with other genomic datasets requires systematic computational approaches:

Integration with Histone Modification Data:

  • Correlation analysis: Calculate Pearson correlations between H3K64ac and other histone modifications:

    • Active marks (H3K4me3, H3K9ac, H3K27ac)

    • Enhancer marks (H3K4me1, H3K27ac)

    • Repressive marks (H3K27me3, H3K9me3, H3K64me3)

  • Chromatin state prediction: Use hidden Markov model-based approaches (e.g., ChromHMM) to define chromatin states incorporating H3K64ac with other marks.

  • Genomic feature analysis: Create meta-profiles of H3K64ac around:

    • Transcriptional start sites (grouped by expression level)

    • Enhancers (active vs. poised)

    • CTCF binding sites

    • Domain boundaries

Integration with Transcription Factor Binding and Accessibility Data:

  • Co-localization analysis: Assess overlap between H3K64ac and:

    • Transcription factor binding sites (esp. p300/CBP)

    • Chromatin accessibility regions (DNase-seq, ATAC-seq)

    • CpG islands and other DNA features

  • Motif enrichment: Identify transcription factor motifs enriched in H3K64ac peaks to identify potential regulatory mechanisms.

Integration with Expression Data:

  • Correlation with gene expression: Group genes by expression levels and analyze H3K64ac patterns.

  • Differential analysis: For treatment/condition comparisons:

    • Identify regions with differential H3K64ac

    • Correlate with differential gene expression

    • Analyze pathway enrichment for genes with concordant changes

Visualization and Analysis Tools:

  • Genome browsers (e.g., UCSC, IGV) for visual inspection

  • deepTools for generating heatmaps and profile plots

  • Bioconductor packages for statistical analysis

  • WashU Epigenome Browser for multi-sample comparisons

Research has demonstrated that H3K64ac clustering with H3K4me1 in correlation analyses indicates its role at enhancers. Additionally, when enhancers are defined by H3K4me1 peaks ±2kb away from TSSs, they can be clustered into three groups based on H3K64ac, H3K27ac, and H3K122ac patterns, revealing subclasses of regulatory elements with distinct properties .

What are emerging research areas regarding the function of H3K64ac in development and disease?

Several promising research directions are emerging regarding H3K64ac's role in biological processes:

Developmental Biology:

  • Cell fate decisions: Investigating how H3K64ac dynamics contribute to developmental transitions and lineage commitment. Preliminary data suggest H3K64ac shifts from pluripotency genes to differentiation-specific genes during development.

  • Epigenetic inheritance: Exploring whether H3K64ac patterns can be transmitted through cell divisions and contribute to epigenetic memory.

  • Imprinting regulation: Further characterizing the role of H3K64ac/H3K64me3 in maintaining imprinted gene expression patterns, as these marks show allele-specific distribution at imprinting control regions.

Disease Implications:

  • Cancer epigenetics: Investigating whether aberrant H3K64ac patterns contribute to oncogenic gene expression programs, particularly in cancers with p300/CBP mutations.

  • Neurodevelopmental disorders: Examining H3K64ac in conditions linked to chromatin dysregulation (e.g., Rubinstein-Taybi syndrome, caused by mutations in CBP).

  • Inflammatory diseases: Exploring the role of H3K64ac in rapid inflammatory gene activation, as p300/CBP are known regulators of inflammatory transcription factors.

Novel Mechanistic Questions:

  • Reader proteins: Identifying potential proteins that specifically recognize H3K64ac, if any exist, despite challenges in detecting specific readers.

  • Interplay with DNA methylation: Investigating how H3K64ac affects or is affected by DNA methylation patterns, particularly at regulatory elements.

  • Chromatin higher-order structure: Determining how H3K64ac influences higher-order chromatin organization and nuclear compartmentalization.

  • Crosstalk with other lateral surface modifications: Exploring functional relationships between H3K64ac and other lateral surface modifications like H3K56ac and H3K122ac.

Early findings show that H3K64ac functions without identified specific readers, suggesting a direct biophysical effect on chromatin structure. This mechanism is distinct from many histone tail modifications that function primarily through reader protein recruitment .

How might technological advances improve detection and functional analysis of H3K64ac?

Emerging technologies hold promise for advancing H3K64ac research:

Next-Generation Antibody Technologies:

  • Recombinant antibodies: Development of recombinant monoclonal antibodies with enhanced specificity and lot-to-lot consistency.

  • Nanobodies/single-domain antibodies: Smaller antibody fragments that may access H3K64ac more efficiently within compact chromatin structures.

  • CUT&Tag/CUT&RUN adaptations: Optimizing these techniques specifically for H3K64ac to achieve higher resolution and sensitivity with less input material.

Mass Spectrometry Advances:

  • Targeted MS approaches: Developing sensitive, targeted MS methods to quantify H3K64ac absolute levels across conditions.

  • Middle-down MS: Analyzing larger histone fragments to understand combinatorial patterns of H3K64ac with other modifications.

  • Crosslinking MS: Identifying proteins interacting with H3K64ac regions through crosslinking approaches.

Single-Cell Technologies:

  • Single-cell ChIP-seq adaptations: Developing protocols to map H3K64ac in individual cells to understand heterogeneity.

  • Multi-omics integration: Combining single-cell H3K64ac profiling with transcriptomics and other epigenetic marks.

  • Live-cell imaging: Creating systems to visualize H3K64ac dynamics in living cells, potentially using engineered reader domains or antibody fragments.

Genome Engineering Approaches:

  • Base editing technologies: Precise editing of H3K64 residue in endogenous histones to study functional consequences.

  • Targeted modification systems: Developing tools to target acetyltransferases or deacetylases specifically to genomic loci to manipulate H3K64ac locally.

  • Degron-based approaches: Controlling histone variant levels and turnover to study H3K64ac dynamics.

These technological advances would address current limitations in studying H3K64ac, including improved sensitivity for detecting low-abundance modifications, better understanding of combinatorial patterns, and enhanced spatial and temporal resolution of H3K64ac dynamics .

What is the current understanding of the interplay between H3K64ac and other lateral surface modifications?

The relationship between H3K64ac and other lateral surface modifications represents an emerging area of research:

Comparative Analysis of Lateral Surface Modifications:

ModificationLocationEffect on NucleosomeAssociated withCatalyzed by
H3K64acLateral surface, α1 helixDecreases stabilityActive transcriptionp300/CBP
H3K64me3Lateral surface, α1 helixIncreases stabilityHeterochromatinUnknown
H3K56acLateral surface, entry/exitDNA breathing at entry/exitReplication, DNA repairGcn5, p300
H3K122acLateral surface, dyad axisDisrupts histone-DNA bindingActive transcription, enhancersp300/CBP
H3K115acLateral surface, near dyadWeakens histone-DNA contactsActive chromatinUnknown

Functional Interrelationships:

  • Co-occurrence patterns: H3K64ac and H3K122ac frequently co-occur at active regulatory elements, particularly at enhancers and promoters, suggesting cooperative functions.

  • Distinct mechanisms: Despite similar genomic distributions, these modifications affect nucleosome dynamics through different mechanisms:

    • H3K64ac: Located near the inner gyre of DNA, affecting DNA-histone contacts

    • H3K56ac: Located near DNA entry/exit points, affecting DNA unwrapping

    • H3K122ac: Located at the dyad axis, directly disrupting central histone-DNA contacts

  • Differential effects on remodeling: H3K64ac enhances Chd1-mediated nucleosome repositioning but not RSC activity, suggesting specificity in remodeler interactions.

Combinatorial Effects:
Recent research suggests that combinations of lateral surface modifications may have synergistic effects on nucleosome dynamics. For example:

  • H3K64ac+H3K122ac may destabilize nucleosomes more effectively than either modification alone

  • The presence of multiple lateral surface acetylations may create a more permissive chromatin state than achievable by tail modifications

Biological Context Specificity:

  • H3K64ac appears particularly important for developmental gene regulation

  • H3K56ac is strongly linked to replication and DNA repair processes

  • H3K122ac has been identified as marking a novel class of active enhancers lacking H3K27ac

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.