HIST1H3A (Ab-64) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Significance of the Target Epitope

Understanding the biological importance of lysine 64 in histone H3 is crucial for appreciating the value of the HIST1H3A (Ab-64) Antibody as a research tool.

Functional Significance of K64 Acetylation

Research has shown that acetylation of histone H3 at lysine 64 (H3K64ac) plays a significant regulatory role in chromatin function. H3K64ac has been demonstrated to:

  • Regulate nucleosome stability

  • Facilitate nucleosome eviction

  • Promote gene expression in vivo

  • Define transcriptionally active chromatin

  • Be enriched at the transcriptional start sites of active genes

These findings position H3K64ac as an important epigenetic mark that opposes its repressive counterpart, H3K64me3 (trimethylation) . The HIST1H3A (Ab-64) Antibody thus targets a modification with critical implications for gene regulation.

Research Applications and Methodologies

The HIST1H3A (Ab-64) Antibody has been validated for multiple research applications, enabling investigators to detect and quantify H3K64 modifications in various experimental contexts.

Validated Experimental Techniques

According to manufacturer specifications and research literature, the antibody has been validated for:

Enzyme-Linked Immunosorbent Assay (ELISA)

The antibody demonstrates reliable performance in ELISA applications, allowing for quantitative detection of the target protein in solution .

Immunohistochemistry (IHC)

For tissue-based research, the antibody has been validated for immunohistochemistry applications. Examples include:

  • Paraffin-embedded human liver cancer tissue

  • Paraffin-embedded human glioma tissue

The recommended dilution range for IHC-P is 1:1-100, although optimal dilutions should be determined by end-users for specific applications .

Immunofluorescence/Immunocytochemistry (IF/ICC)

The antibody has been validated for cellular localization studies with recommended dilutions of 1/1-1/10 .

Specificity Assessment

The specificity of the HIST1H3A (Ab-64) Antibody has been extensively validated. In research studies:

  • The antibody specifically detects endogenous acetylated histone H3

  • It recognizes K64-acetylated peptides with high specificity compared to other H3 acetylated lysines

  • Its recognition of H3 can be efficiently competed by the immunizing peptide, but not by other peptides containing acetylated, methylated, or unmodified histone regions

  • Limited tryptic digestion of native nucleosomes confirmed the antibody's specificity for the core region of H3

These validation studies ensure that experimental results obtained using this antibody accurately reflect the targeted histone modification.

Significant Research Findings

Scientific investigations using antibodies targeting H3K64 have yielded important insights into chromatin regulation and gene expression mechanisms.

Chromatin State and Gene Expression

Research has established that H3K64ac is enriched in euchromatin, which is the less condensed, transcriptionally active chromatin state . Chromatin immunoprecipitation (ChIP) analysis has shown that H3K64ac is preferentially associated with:

  • Pluripotency genes in embryonic stem cells

  • Active alleles at imprinting control regions (ICRs) in embryonic fibroblasts

This enrichment pattern supports the role of H3K64ac as a marker of active chromatin and transcriptionally engaged genomic regions.

Enzymatic Regulation

Studies have identified p300 and CBP as the primary histone acetyltransferases responsible for H3K64 acetylation. Key findings include:

  • Knockdown of p300 and CBP decreases steady-state levels of H3K64ac

  • This decrease is most pronounced at p300/CBP-specific genomic target regions

  • Overexpression of p300 results in increased levels of H3K64ac

  • p300 and CBP can acetylate H3K64 in vitro on free H3 and within chromatin contexts

These findings place H3K64ac within a specific enzymatic pathway, enhancing our understanding of its regulation and biological significance.

Role in Histone Variant Distribution

Interesting connections have been found between H3K64ac and histone variants. Distribution analysis of H3K64ac among the H3 variants (H3.1, H3.2, H3.3) shows differential enrichment patterns, suggesting variant-specific regulatory mechanisms .

Comparative Analysis with Related Antibodies

The HIST1H3A (Ab-64) Antibody belongs to a family of antibodies targeting different epitopes on histone H3, each with distinct applications and significance.

Related Products within the Same Family

Several related antibodies target different modifications or regions of histone H3:

  • HIST1H3A (Ab-64) Antibody [orb417605]: Validated for ELISA and IHC

  • HIST1H3A (Ab-64) Antibody [orb417613]: Validated for ELISA and IF

  • HIST1H3A (Ab-64) Antibody [orb752886]: Validated for ELISA and IHC

These antibodies share the same target region but may have slight differences in their specific epitope recognition or performance in different applications.

Comparison with Antibodies Against Other Histone H3 Modifications

The research literature mentions other important antibodies targeting different histone H3 modifications, including:

  • Histone H3K27ac Antibody: Targets acetylation at lysine 27, another important activation mark

  • Histone H3K4me1 Antibody: Targets monomethylation at lysine 4, associated with enhancer regions

Each of these antibodies provides complementary information about the epigenetic state of chromatin and allows researchers to build a comprehensive understanding of histone-mediated regulation.

Connections to Pathological Conditions

Research involving histone H3 lysine 64 modifications has implications for understanding certain disease states, particularly cancer.

Cancer Research Applications

The HIST1H3A (Ab-64) Antibody has been used in immunohistochemistry studies of:

  • Human liver cancer tissue

  • Human glioma tissue

These applications highlight the potential relevance of H3K64 modifications in cancer biology.

Relevance to Pediatric Glioblastoma

Research has identified gain-of-function mutations in histone H3.3, specifically G34R and K27M, as pediatric glioblastoma driver mutations that differentially affect H3.3 recruitment to activated transgenes . Though not directly targeting K64, these findings emphasize the importance of histone H3 modifications in cancer pathogenesis and suggest potential research directions for H3K64-specific studies.

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timelines.
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. Nucleosomes function to wrap and compact DNA into chromatin, limiting the accessibility of DNA to cellular machineries that require DNA as a template. Histones, therefore, play a crucial role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is regulated through a complex set of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer occurs through the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course can be useful for determining whether tumors are heterochronous. PMID: 29482987
  3. Recent findings demonstrate that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts 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 suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) offers an alternative as a proliferative marker. PMID: 29040195
  5. These results identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Research shows that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments demonstrated 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. For the first time, we describe 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 has been shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. 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. The 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 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. On the other hand, histone 3 mutations do not appear 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. Taken together, the authors verified that histone H3 is a real substrate for GzmA in vivo in Raji cells treated by staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that 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. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) serves 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. Data indicate that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that 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. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays is 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 HIST1H3A and why is the lysine 64 position significant?

HIST1H3A is a histone H3 variant (H3.1) that serves as a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries that require DNA as a template. Histones play a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability . The lysine 64 (K64) position is particularly significant because it is located on the lateral surface of the histone octamer. Modifications at this site, especially acetylation (H3K64ac), have been shown to regulate nucleosome stability and facilitate nucleosome eviction, directly impacting gene expression in vivo . Unlike many other histone modifications that occur on the N-terminal tails, K64 modifications affect the core structure of the nucleosome, potentially having more direct effects on chromatin dynamics.

How do H3K64 modifications differ from other histone marks in function?

H3K64 modifications differ fundamentally from many other histone marks because of their strategic location within the nucleosome structure. While many well-studied histone modifications (such as H3K4me3, H3K9ac, H3K27me3) occur on the N-terminal tails that extend outward from the nucleosome core, K64 is positioned on the lateral surface of the histone octamer, in closer proximity to the DNA that wraps around it . Research has demonstrated that acetylation at K64 (H3K64ac) regulates nucleosome stability and facilitates nucleosome eviction, directly impacting chromatin accessibility and gene expression . This positional difference means that H3K64 modifications may have more immediate biophysical effects on DNA-histone interactions compared to tail modifications, which often function by recruiting specific reader proteins. H3K64ac has been specifically found to be enriched at the transcriptional start sites of active genes, defining transcriptionally active chromatin regions .

What is the difference between antibodies targeting H3K64ac versus unmodified K64?

Antibodies targeting H3K64ac and unmodified K64 recognize distinctly different epitopes and serve different research purposes. H3K64ac antibodies, such as the rabbit recombinant monoclonal H3 acetyl K64 antibody [EPR20713], specifically recognize the acetylated form of lysine 64 on histone H3 . These antibodies are vital for studying the presence and distribution of this activation-associated mark in chromatin and its correlation with gene expression. In contrast, antibodies targeting unmodified K64, like the HIST1H3A (Ab-64) polyclonal antibody, recognize the unmodified lysine residue at position 64 . These antibodies are useful for detecting the baseline presence of H3 proteins and can serve as controls when studying modifications. The specificity of these antibodies is typically ensured through peptide synthesis around the K64 site of human histone H3.1 as immunogens, followed by affinity purification . When selecting between these antibodies, researchers should consider whether they're investigating the presence of the activation mark (H3K64ac) or the general distribution of H3 irrespective of its modification state.

What experimental applications are most appropriate for HIST1H3A (Ab-64) antibodies?

HIST1H3A (Ab-64) antibodies have been validated for multiple experimental applications, with varying degrees of optimization for specific techniques. Based on manufacturer specifications and research literature, these antibodies are particularly well-suited for:

ApplicationValidation StatusRecommended Dilution
Western Blotting (WB)Validated1:500 - 1:2000
Chromatin Immunoprecipitation (ChIP)Validated2-5 μg per IP
ChIP-sequencing (ChIP-seq)Validated2-5 μg per IP
Immunocytochemistry/Immunofluorescence (ICC/IF)Validated1:100 - 1:500
ELISAValidated1:1000 - 1:5000
Peptide Array (PepArr)ValidatedAssay-dependent
Immunohistochemistry (IHC)Validated for some antibodies1:100 - 1:500

When designing experiments, it's critical to consider whether you're investigating the unmodified or modified (acetylated/methylated) form of K64, as this will determine which specific antibody variant to use . For comprehensive chromatin studies, combining ChIP with high-throughput sequencing (ChIP-seq) provides genome-wide mapping of H3K64 modification patterns that can be correlated with gene expression data. For visual confirmation of nuclear localization and distribution patterns, ICC/IF approaches are recommended, particularly when co-staining with other nuclear markers.

How can I design a ChIP experiment to effectively study H3K64 modifications in different cell states?

Designing an effective ChIP experiment to study H3K64 modifications requires careful planning and execution. Based on research protocols from the literature , I recommend the following approach:

  • Cell preparation and crosslinking: Harvest cells in different states of interest (e.g., before/after treatment, different cell cycle phases). Use 1% formaldehyde for 10 minutes at room temperature for protein-DNA crosslinking, followed by quenching with glycine (125 mM final concentration).

  • Nuclear isolation and sonication: Isolate nuclei by incubating cell pellets for 10 minutes in TEB buffer (0.5% Triton X-100 in PBS) . Sonicate chromatin to fragments of 200-500 bp using optimized conditions for your sonicator.

  • Antibody selection and validation: Choose highly specific antibodies for H3K64ac or other H3K64 modifications. Always validate antibody specificity using peptide competition assays or knockdown experiments. Include controls such as IgG and antibodies against well-characterized marks (H3K4me3 for active promoters, H3K27me3 for repressed regions) .

  • Immunoprecipitation: Use 2-5 μg of H3K64-specific antibody per IP reaction with chromatin from approximately 1-5×10^6 cells. Include input controls (non-immunoprecipitated chromatin) and IgG controls (non-specific binding).

  • Washing and elution: Perform stringent washing steps to reduce background. For H3K64ac ChIP, use wash buffers with increasing salt concentrations as described in published protocols .

  • Analysis: For targeted analysis, use qPCR with primers for regions of interest (promoters, enhancers). For genome-wide profiling, proceed to library preparation for ChIP-seq. When analyzing H3K64ac ChIP-seq data, look for enrichment at transcriptional start sites of active genes as this modification has been shown to define transcriptionally active chromatin .

  • Data interpretation: Compare H3K64 modification patterns with other histone marks (H3K4me3, H3K9ac, H3K27ac) and gene expression data to understand functional correlations .

For differential analysis between cell states, ensure consistent chromatin preparation and IP conditions across samples, and include spike-in controls for normalization if possible.

What controls should be included when validating the specificity of H3K64 antibodies?

When validating the specificity of H3K64 antibodies, comprehensive controls are essential to ensure reliable and interpretable results. Based on best practices in the field, I recommend including the following controls:

  • Peptide competition assays: Pre-incubate the antibody with the peptide used as an immunogen (peptide sequence around site of Lys-64 derived from Human Histone H3.1) . A specific antibody should show significantly reduced or eliminated signal when the competing peptide is present.

  • Modified vs. unmodified peptide arrays: Test the antibody against peptide arrays containing the target site in various modification states (unmodified, acetylated, methylated, etc.) to confirm specific recognition of the intended modification state .

  • Western blot validation:

    • Use recombinant histone H3 proteins (wild-type and K64A mutant) as controls to confirm specificity

    • Include histones from cells treated with histone deacetylase inhibitors (for acetyl-specific antibodies) which should increase the signal

    • Test against histones from HAT enzyme overexpression systems to confirm increased signal for H3K64ac antibodies

  • Immunofluorescence controls:

    • Include secondary antibody-only controls to exclude non-specific binding

    • Use peptide competition in parallel samples

    • Include cells with known modification states as positive and negative controls

  • Genetic validation: When possible, use cell lines with H3.1/H3.3 K64R or K64A mutations that cannot be acetylated/methylated at this position as negative controls .

  • Cross-reactivity testing: Test against other histone modifications, particularly those at nearby residues, to ensure the antibody doesn't cross-react with similar epitopes.

  • Species specificity validation: Confirm reactivity across relevant species if working with non-human models, as the H3K64 region is highly conserved but may have subtle sequence differences that affect antibody recognition .

How should I interpret different signal intensities of H3K64ac across genomic regions?

Interpreting H3K64ac signal intensities across genomic regions requires careful consideration of the biological context and technical aspects of your experiment. H3K64ac is a modification that has been shown to regulate nucleosome stability and facilitate nucleosome eviction, directly affecting gene expression . When analyzing ChIP-seq or similar data for H3K64ac, consider the following interpretation framework:

  • Promoter enrichment: H3K64ac has been found to be specifically enriched at transcriptional start sites (TSS) of active genes . Strong signals at promoters typically indicate actively transcribed genes. Compare these signals with RNA-seq data to confirm the correlation between H3K64ac enrichment and gene expression levels.

  • Correlation with other active marks: H3K64ac should positively correlate with other activation-associated histone marks like H3K4me3, H3K9ac, and H3K27ac . Regions with co-enrichment of these marks can be confidently classified as active chromatin domains.

  • Cell type-specific patterns: Different cell types may show distinct H3K64ac distribution patterns reflecting their specific transcriptional programs. These differences are biologically meaningful and should be interpreted in the context of cell identity and function.

  • Dynamics during cellular processes: Changes in H3K64ac levels during processes like differentiation or response to stimuli reflect dynamic regulation of chromatin accessibility. Increasing signals often precede or accompany gene activation.

  • Broad vs. narrow peaks: H3K64ac may appear as either focused peaks at regulatory elements or broader domains across transcribed regions. The pattern provides insight into how this modification contributes to local chromatin environment.

For accurate interpretation, always normalize your H3K64ac signal to appropriate controls, including input DNA and, if possible, total H3 distribution to account for nucleosome occupancy variations. Remember that the absolute signal intensity is influenced by antibody efficiency, chromatin preparation, and sequencing depth, so relative enrichment patterns are often more informative than absolute values .

What are common technical challenges when working with H3K64 antibodies and how can they be addressed?

Working with H3K64 antibodies presents several technical challenges that can affect experimental outcomes. Based on research practices and literature, here are the most common issues and recommended solutions:

  • Epitope masking in fixed samples:

    • Challenge: Formaldehyde fixation may obscure the K64 epitope, which is located on the lateral surface of the histone octamer.

    • Solution: Optimize fixation time (try shorter crosslinking, 5-8 minutes instead of 10-15) or use epitope retrieval methods for IHC/IF applications. For ChIP applications, test different crosslinking conditions to find the optimal balance between chromatin preservation and epitope accessibility .

  • Cross-reactivity with other histone modifications:

    • Challenge: Antibodies may recognize similar modification sites on other histones or other modifications at nearby residues.

    • Solution: Always validate antibody specificity using peptide competition assays and dot blots with modified peptides. For critical applications, use antibodies that have been validated by multiple methods including in K64 mutant backgrounds .

  • Batch-to-batch variability in polyclonal antibodies:

    • Challenge: Polyclonal HIST1H3A (Ab-64) antibodies may show lot-to-lot variation in specificity and sensitivity.

    • Solution: When possible, use recombinant monoclonal antibodies like EPR20713 for H3K64ac . If using polyclonal antibodies, validate each new lot against previous standards and consider purchasing larger lots for long-term projects.

  • Low signal-to-noise ratio in ChIP experiments:

    • Challenge: H3K64 modifications may have lower abundance than some well-studied histone marks.

    • Solution: Increase chromatin input, optimize antibody concentration (typically 2-5 μg per IP), extend incubation times (overnight at 4°C), and use more stringent washing conditions. Sequential ChIP (re-ChIP) can also increase specificity for studying co-occurrence with other marks .

  • Inefficient nuclear permeabilization for immunofluorescence:

    • Challenge: The K64 position is less accessible than tail modifications in intact nuclei.

    • Solution: Use more robust permeabilization protocols (0.5% Triton X-100 in PBS for 20 minutes) and consider including 0.1-0.2% SDS in the permeabilization buffer for improved nuclear penetration .

  • Quantification challenges in western blots:

    • Challenge: Determining relative levels of K64 modifications accurately.

    • Solution: Always normalize to total H3 levels from the same samples. For H3 variants, consider using HA-tagged systems for precise quantification as described in the literature: "Quantification was done using the ImageJ software and the ratio between specific H3K64ac and HA signals (loading control) was calculated" .

How does H3K64 acetylation interact with other histone modifications in regulating gene expression?

H3K64 acetylation operates within a complex network of histone modifications that collectively regulate chromatin structure and gene expression. Understanding these interactions is crucial for advanced chromatin research. Evidence from multiple studies reveals several key interaction patterns:

  • Co-occurrence with activation-associated marks: H3K64ac positively correlates with other active histone marks, particularly H3K4me3, H3K9ac, and H3K27ac at promoters and enhancers of actively transcribed genes . This suggests a coordinated mechanism where multiple acetylation events work together to create a permissive chromatin environment. The unique position of K64 on the lateral surface of the histone octamer complements the function of tail modifications by directly affecting nucleosome stability.

  • Antagonistic relationship with repressive marks: H3K64ac shows mutually exclusive patterns with repressive modifications like H3K9me3 and H3K27me3 . Regions marked with H3K64ac typically show depletion of these repressive marks, indicating a binary switch in chromatin states.

  • Sequential modification patterns: Emerging evidence suggests that H3K64ac may be established after certain tail modifications. The p300 co-activator has been identified as an enzyme that acetylates H3K64 , and its recruitment often follows pioneer factor binding and initial enhancer marking. This suggests H3K64ac may function as a secondary modification that reinforces and stabilizes active chromatin states initiated by other modifications.

  • Cross-talk with histone variant incorporation: H3K64ac patterns can differ between canonical H3.1/H3.2 and variant H3.3 histones, adding another layer of regulation. These differences may reflect distinct roles in replication-dependent versus replication-independent chromatin assembly pathways.

  • Functional consequences of combinatorial modifications: The combination of H3K64ac with specific tail modifications appears to have functional consequences beyond those of individual marks. For example, nucleosomes carrying both H3K64ac and H3K27ac may be particularly unstable and prone to eviction during transcriptional activation .

For researchers investigating these interactions, sequential ChIP (re-ChIP) experiments using antibodies against H3K64ac and other modifications can reveal co-occurrence patterns at specific genomic locations. Mass spectrometry approaches can also quantify combinatorial modification states on the same histone molecules, providing insights not accessible through standard ChIP approaches.

What role does H3K64 acetylation play in nucleosome dynamics and chromatin accessibility?

H3K64 acetylation plays a distinctive and mechanistically significant role in nucleosome dynamics and chromatin accessibility. Unlike many histone tail modifications that primarily function as binding platforms for reader proteins, H3K64ac directly affects the core biophysical properties of the nucleosome due to its strategic location:

  • Direct effects on nucleosome stability: Research has demonstrated that H3K64ac regulates nucleosome stability by weakening histone-DNA interactions . This occurs because lysine 64 is positioned at the lateral surface of the histone octamer where it directly contacts the DNA wrapped around the nucleosome. Acetylation neutralizes the positive charge of lysine, reducing electrostatic interactions with the negatively charged DNA backbone. This charge neutralization destabilizes the nucleosome structure, making DNA more accessible to transcription factors and RNA polymerase.

  • Facilitation of nucleosome eviction: Studies have shown that H3K64ac facilitates nucleosome eviction during transcriptional activation . This property is particularly important at promoters and enhancers where nucleosome displacement is often necessary for gene activation. The enrichment of H3K64ac at transcriptional start sites of active genes supports this functional role in creating accessible chromatin regions .

  • Interaction with chromatin remodeling complexes: Emerging evidence suggests that H3K64ac may enhance the activity of ATP-dependent chromatin remodeling complexes. These complexes, which slide or evict nucleosomes to regulate DNA accessibility, may recognize H3K64ac-containing nucleosomes as preferred substrates due to their already destabilized nature.

  • Impact on higher-order chromatin structure: Beyond effects on individual nucleosomes, H3K64ac may influence higher-order chromatin folding by altering internucleosomal interactions. Regions enriched for H3K64ac tend to adopt more open, accessible conformations that facilitate transcription.

  • Dynamics during transcriptional activation: Temporal studies suggest that H3K64ac levels increase during transcriptional activation, preceding or accompanying the onset of gene expression . This supports a model where H3K64ac is part of the mechanism that converts chromatin from a repressed to an active state.

For researchers studying these dynamics, techniques such as ATAC-seq (Assay for Transposase-Accessible Chromatin) can be combined with H3K64ac ChIP-seq to correlate this modification with chromatin accessibility genome-wide. Additionally, in vitro nucleosome assembly and stability assays using recombinant histones with K64 modifications can provide direct biophysical measurements of how this modification affects nucleosome properties .

How can I integrate H3K64 modification data with other epigenomic and transcriptomic datasets?

Integrating H3K64 modification data with other epigenomic and transcriptomic datasets requires systematic analytical approaches to reveal functional relationships. This integration is essential for understanding how H3K64 modifications fit within the broader epigenetic landscape and influence gene expression. Based on current research practices, I recommend the following strategies:

  • Multi-omics correlation analysis:

    • Correlate H3K64ac ChIP-seq profiles with RNA-seq data to establish direct relationships between this modification and gene expression levels

    • Calculate correlation coefficients between H3K64 modifications and other histone marks across genomic regions

    • Create heatmaps centered on transcription start sites (TSS) or enhancers showing the co-occurrence patterns of multiple histone modifications including H3K64ac

  • Chromatin state modeling:

    • Use computational tools like ChromHMM or Segway to define chromatin states based on combinatorial patterns of histone modifications including H3K64ac

    • Analyze how regions with H3K64ac are classified in these models and their relationship to functional genomic elements

    • Example finding: "H3K64ac is enriched at the transcriptional start sites of active genes and it defines transcriptionally active chromatin"

  • Transcription factor binding integration:

    • Overlap H3K64ac peaks with transcription factor ChIP-seq data to identify potential functional interactions

    • Investigate whether specific transcription factors are enriched at H3K64ac-marked regions

    • Pay special attention to p300/CBP binding sites, as p300 has been identified as an enzyme that acetylates H3K64

  • Nucleosome positioning analysis:

    • Integrate H3K64ac data with MNase-seq or ATAC-seq to correlate this modification with nucleosome positioning and stability

    • Analyze nucleosome occupancy changes in regions with dynamic changes in H3K64ac

  • Three-dimensional chromatin organization:

    • Correlate H3K64ac patterns with Hi-C or other chromosome conformation capture data to understand how this modification relates to higher-order chromatin structure

    • Examine whether H3K64ac-enriched regions show preferences for specific chromatin interaction patterns

  • Computational implementation:

    • Utilize R/Bioconductor packages (such as DiffBind, ChIPseeker, and clusterProfiler) or Python libraries (such as deepTools and pyGenomeTracks) for integrated analysis

    • Normalize datasets appropriately before integration to account for technical variations

    • Implement appropriate statistical methods to identify significant correlations while controlling for confounding factors

  • Visualization strategies:

    • Create browser tracks showing multiple data types aligned to the same genomic coordinates

    • Generate aggregation plots showing average profiles of different features around H3K64ac peaks

    • Develop circular visualization plots (Circos) for genome-wide integration of multiple data types

For researchers new to integrative analysis, I recommend starting with focused analyses of promoters and enhancers where H3K64ac has been shown to play important functional roles , before expanding to genome-wide integration.

What is the optimal protocol for histone extraction when studying H3K64 modifications?

The optimal protocol for histone extraction when studying H3K64 modifications must preserve both the integrity of histones and their post-translational modifications while ensuring high yield and purity. Based on published methodologies, I recommend the following optimized protocol specifically tailored for H3K64 studies:

  • Cell preparation and harvesting:

    • Harvest 1-5×10^6 cells in their desired state (treatment conditions, time points)

    • Wash cells twice with ice-cold PBS containing protease inhibitors and HDAC inhibitors (5 mM sodium butyrate and 10 mM nicotinamide) to preserve acetylation marks including H3K64ac

  • Nuclei isolation:

    • Gently resuspend cell pellet in TEB buffer (0.5% Triton X-100, 2 mM PMSF, 0.02% NaN₃ in PBS) with protease and deacetylase inhibitors

    • Incubate on ice for 10 minutes with gentle mixing

    • Centrifuge at 2000 rpm for 10 minutes at 4°C

    • Wash the nuclear pellet once more with TEB buffer

  • Acid extraction of histones:

    • Resuspend nuclei in 0.2 N HCl (100-200 μl per 10^6 cells)

    • Incubate overnight at 4°C with rotation to extract histones

    • Centrifuge at maximum speed (>10,000 g) for 10 minutes at 4°C

    • Carefully transfer the supernatant containing acid-soluble histones to a new tube

  • Concentration and desalting (optional but recommended):

    • Neutralize the acid with 1/10 volume of 2 M NaOH

    • Precipitate histones by adding TCA to a final concentration of 20% and incubating on ice for 30 minutes

    • Centrifuge at maximum speed for 10 minutes, wash pellet with acetone containing 0.1% HCl, then pure acetone

    • Air-dry and resuspend in water or desired buffer

  • Quality control and quantification:

    • Determine protein concentration using the Bradford method

    • Analyze 1-2 μg by SDS-PAGE followed by Coomassie staining to verify purity and integrity

    • For specific H3K64 modification analysis, perform Western blot using appropriate antibodies

  • Special considerations for H3K64:

    • Since K64 is in the globular domain of H3, ensure complete denaturation of histones before SDS-PAGE by heating samples to 95°C for 5 minutes in sample buffer containing 5% SDS

    • For mass spectrometry analysis of H3K64 modifications, consider using propionylation to block unmodified lysines and improve detection specificity

This protocol has been validated for the extraction of histones with preserved K64 modifications as demonstrated in studies focusing on H3K64ac and related modifications . For researchers requiring higher purity, HPLC fractionation of histone variants can be performed as an additional step after acid extraction.

How can I optimize immunofluorescence protocols for detecting H3K64 modifications in different cell types?

Optimizing immunofluorescence (IF) protocols for detecting H3K64 modifications requires special consideration due to the location of K64 within the nucleosome structure. Based on published methods and technical expertise, I recommend the following optimized protocol with specific adjustments for different cell types:

  • General protocol optimization:

    a) Fixation and permeabilization:

    • Fix cells in 4% paraformaldehyde/2% sucrose for 15 minutes at room temperature

    • Wash three times in cold PBS (5 minutes each)

    • Perform enhanced permeabilization with 0.5% Triton X-100 in PBS for 20 minutes at room temperature

    • For difficult-to-permeabilize cells, consider adding 0.1% SDS to the permeabilization buffer

    b) Epitope retrieval (critical for K64 detection):

    • For formalin-fixed tissue sections or stubborn samples, add a heat-mediated antigen retrieval step

    • Use 10 mM sodium citrate buffer (pH 6.0) at 95°C for 20 minutes, then cool to room temperature

    c) Blocking and antibody incubation:

    • Block in 3% BSA in PBS for 1 hour at room temperature

    • Dilute primary H3K64 antibodies in blocking solution (1:100 to 1:500 depending on antibody and cell type)

    • Incubate overnight at 4°C in a humidified chamber

    • Wash 3×5 minutes with PBS + 0.05% Tween-20

    • Incubate with appropriate fluorescent secondary antibodies (1:500-1:1000) for 1 hour at room temperature

    • Include DAPI (1:1000) during secondary antibody incubation or as a separate step

  • Cell type-specific optimizations:

    a) Adherent cells (e.g., HeLa, fibroblasts):

    • Grow cells on poly-L-lysine coated coverslips for better adhesion

    • Standard protocol works well with 0.5% Triton X-100 permeabilization

    b) Suspension cells (e.g., lymphocytes):

    • Cytospin cells onto slides (300 rpm, 3 minutes) or use poly-L-lysine coating

    • Extend permeabilization time to 25-30 minutes

    c) Stem cells and primary tissues:

    • For embryonic stem cells: use shorter fixation (10 minutes) to prevent over-crosslinking

    • For tissues: section at 5-7 μm thickness and use antigen retrieval as described above

    • For mouse testis tubules: follow specialized protocols as described in the literature

  • Critical quality controls:

    • Include a peptide competition control (pre-incubate antibody with immunizing peptide)

    • Use cell types with known high levels of the target modification as positive controls

    • Include secondary antibody-only controls to assess background

    • Consider dual staining with antibodies against total H3 to normalize signal

  • Image acquisition and analysis:

    • Use confocal microscopy with a 63× or 100× oil objective for optimal resolution

    • Acquire Z-stacks (0.5 μm intervals) to capture the full nuclear volume

    • For quantitative analysis, maintain identical acquisition settings across all samples

    • Use appropriate software (ImageJ/FIJI) for quantification, normalizing H3K64 modification signals to total H3 or DAPI

This optimized protocol has been successfully applied to detect H3K64 modifications in various cell types and tissues, enabling both qualitative assessment of nuclear distribution patterns and quantitative analysis of modification levels .

What are the best practices for designing H3K64-specific ChIP-seq experiments for genome-wide profiling?

Designing H3K64-specific ChIP-seq experiments for genome-wide profiling requires careful consideration of several critical factors to ensure high-quality, interpretable data. Based on published methodologies and technical expertise, I recommend the following best practices:

  • Experimental design considerations:

    a) Controls and replicates:

    • Include input DNA controls (non-immunoprecipitated chromatin) for each condition

    • Use IgG control immunoprecipitations to establish background levels

    • Perform at least 3 biological replicates per condition for statistical robustness

    • Consider spike-in normalization using chromatin from a different species (e.g., Drosophila) for quantitative comparisons between conditions

    b) Antibody selection and validation:

    • Use highly specific antibodies validated for ChIP applications

    • Perform western blot validation on the same chromatin preparation

    • Consider using multiple antibodies targeting the same modification (from different vendors or clones) for confirmation of key findings

    • For H3K64ac, the rabbit recombinant monoclonal antibody [EPR20713] has been validated for ChIP-seq applications

  • Optimized ChIP protocol for H3K64 modifications:

    a) Chromatin preparation:

    • Crosslink cells with 1% formaldehyde for 10 minutes at room temperature

    • Isolate nuclei using TEB buffer (0.5% Triton X-100 in PBS)

    • Sonicate chromatin to 200-500 bp fragments (optimize conditions for your cell type)

    • Verify fragment size distribution by agarose gel electrophoresis before proceeding

    b) Immunoprecipitation optimization:

    • Use 2-5 μg of H3K64-specific antibody per IP reaction

    • Include HDAC inhibitors (sodium butyrate, nicotinamide) in all buffers when studying H3K64ac

    • Extend antibody incubation to overnight at 4°C with rotation

    • Use protein A/G magnetic beads for efficient capture and washing

    • Perform stringent washing steps with increasing salt concentrations to reduce background

    c) Library preparation considerations:

    • Use 5-10 ng of ChIP DNA for library preparation (quantify by fluorometric methods)

    • Select size range of 200-500 bp during library preparation

    • Include unique molecular identifiers (UMIs) to control for PCR duplicates

    • Sequence to a minimum depth of 20-30 million uniquely mapped reads per sample

  • Bioinformatic analysis best practices:

    a) Quality control metrics:

    • Assess library complexity (non-redundant fraction)

    • Calculate fraction of reads in peaks (FRiP score) - aim for >1% for H3K64ac

    • Evaluate peak width distribution (H3K64ac typically shows sharper peaks near TSS)

    • Perform correlation analysis between replicates (Pearson r > 0.7)

    b) Specialized analysis for H3K64 modifications:

    • Focus analysis on TSS regions where H3K64ac is known to be enriched

    • Generate average profile plots centered on TSS of genes grouped by expression level

    • Correlate H3K64ac signal with gene expression data and other histone marks

    • For differential analysis between conditions, use specialized tools like DiffBind or MACS2 with bdgdiff

    c) Biological interpretation:

    • Perform gene ontology and pathway analysis of genes associated with H3K64 modifications

    • Integrate with transcription factor binding data to identify potential regulatory relationships

    • Correlate changes in H3K64 modifications with changes in gene expression during biological processes

This comprehensive approach has been successfully applied to profile H3K64ac and other modifications genome-wide, revealing their distribution patterns and functional associations with transcriptional activity .

What are emerging research areas involving H3K64 modifications beyond transcriptional regulation?

While H3K64 modifications, particularly acetylation, have been primarily studied in the context of transcriptional regulation, several emerging research areas are expanding our understanding of their functions. These frontier areas represent exciting opportunities for researchers:

  • DNA damage response and repair: Recent evidence suggests that H3K64 modifications may be dynamically regulated during DNA damage response. The structural position of K64 near the DNA entry/exit points on the nucleosome makes it strategically positioned to influence chromatin accessibility for repair factors. Researchers are beginning to investigate whether H3K64ac facilitates chromatin relaxation at damage sites and whether other modifications (like methylation) might play opposing roles in this process.

  • Cellular reprogramming and development: The role of H3K64 modifications in cell fate transitions is an emerging area of interest. Given that H3K64ac correlates with active chromatin states , its dynamics during cellular reprogramming (such as iPSC generation or transdifferentiation) may provide insights into chromatin barriers and facilitators of cell identity changes. Developmental studies tracking H3K64 modifications through embryogenesis could reveal critical transition points in chromatin organization.

  • Three-dimensional genome organization: The impact of H3K64 modifications on higher-order chromatin structure remains largely unexplored. Since these modifications affect nucleosome stability , they likely influence how chromatin folds in three-dimensional space. Integration of H3K64ac ChIP-seq data with Hi-C or similar techniques could reveal whether regions enriched for this modification show distinct interaction patterns or topological preferences.

  • Aging and age-related diseases: Changes in histone modification patterns are hallmarks of aging, but H3K64 modifications have been understudied in this context. Given their fundamental role in regulating chromatin structure, age-associated changes in H3K64 modification patterns could contribute to the chromatin disorganization observed in aging tissues and age-related diseases.

  • Non-coding RNA regulation: Preliminary evidence suggests potential crosstalk between H3K64 modifications and the function of long non-coding RNAs in chromatin regulation. This emerging field explores whether lncRNAs influence the deposition or removal of H3K64 modifications, or conversely, whether these modifications affect lncRNA binding to chromatin.

  • Metabolic regulation of chromatin states: Since acetylation requires acetyl-CoA, a key metabolic intermediate, researchers are beginning to explore how cellular metabolism influences H3K64ac levels. This connection could provide novel insights into how metabolic states impact chromatin structure and gene expression through H3K64 modifications.

For researchers interested in these emerging areas, combining H3K64-specific antibodies with new methodologies such as CUT&RUN, CUT&Tag, or single-cell ChIP-seq could provide unprecedented insights into the diverse functions of these modifications beyond transcriptional regulation.

How might advances in antibody technology improve the study of H3K64 and related histone modifications?

Advances in antibody technology are poised to significantly enhance the study of H3K64 and related histone modifications, enabling more precise, quantitative, and comprehensive analyses. Several promising developments with direct relevance to H3K64 research include:

  • Recombinant monoclonal antibody engineering:

    • The shift from polyclonal to recombinant monoclonal antibodies, like the EPR20713 antibody for H3K64ac , eliminates batch-to-batch variability and improves reproducibility

    • Site-specific mutagenesis of antibody paratopes can further enhance specificity for H3K64 modifications versus similar epitopes

    • Protein engineering approaches may produce antibodies with improved binding kinetics and affinity for low-abundance H3K64 modifications

  • Combinatorial modification-specific antibodies:

    • Development of antibodies that specifically recognize H3K64 modifications in combination with other nearby modifications (e.g., H3K64ac+K56ac) would enable studies of modification crosstalk

    • Such antibodies could be generated using synthetic peptides with defined modification patterns as immunogens

    • These tools would allow researchers to directly investigate combinatorial histone codes involving H3K64

  • Single-chain antibody fragments (scFvs) and nanobodies:

    • Smaller antibody formats like scFvs and camelid-derived nanobodies offer improved nuclear penetration for imaging applications

    • Their reduced size may provide better access to the H3K64 position within compact chromatin structures

    • These formats are amenable to intracellular expression as "chromobodies" for live-cell tracking of H3K64 modifications

  • Proximity-labeling antibody conjugates:

    • Antibodies linked to enzymes like APEX2, BioID, or TurboID can identify proteins in proximity to H3K64 modifications

    • This approach could reveal modification-specific protein interactions and complexes that recognize H3K64ac

    • Implementation would involve ChIP using these conjugates followed by mass spectrometry

  • Quantitative multiplexed detection systems:

    • Antibodies conjugated to unique DNA barcodes (similar to CITE-seq) would enable simultaneous quantification of multiple histone modifications including H3K64ac

    • Mass cytometry (CyTOF) with metal-labeled antibodies could provide single-cell quantification of H3K64 modifications alongside other epigenetic and cellular markers

    • These approaches would facilitate comprehensive epigenetic profiling that includes H3K64 status

  • Integration with CRISPR technologies:

    • CRISPR-based approaches using catalytically inactive Cas9 (dCas9) fused to antibody-recruiting domains could enable locus-specific pulldown of chromatin containing H3K64 modifications

    • This would allow researchers to study the genomic context of H3K64 modifications at specific loci without genome-wide ChIP

  • Structurally validated antibodies:

    • Determination of antibody-epitope co-crystal structures for H3K64 modification-specific antibodies would provide molecular insights into recognition specificity

    • This structural information could guide rational antibody engineering for improved performance

    • Cryo-EM studies of antibodies bound to nucleosomes containing H3K64 modifications would reveal recognition in the native chromatin context

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.