HIST1H3A (Ab-56) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
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 H3 is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, restricting DNA accessibility to cellular machinery that requires DNA as a template. Therefore, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. The accessibility of DNA is regulated by 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 involves 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 helpful in determining if tumors are heterochronous. PMID: 29482987
  3. Studies have revealed that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease. It mediates histone H3 N-tail proteolytic cleavage under stressful conditions that trigger a DNA damage response. PMID: 28982940
  4. Findings suggest that the Ki-67 antigen proliferative index has significant limitations. Phosphohistone H3 (PHH3) presents a viable 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. This series of 47 diffuse midline gliomas demonstrates that the histone H3-K27M mutation is mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurs with BRAF-V600E mutation and is commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Research shows that the histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. Experiments have 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. This study provides the first description of 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). This mutation is correlated with poor outcomes and has been shown to influence EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data indicates 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, while 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. Research suggests 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. This 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 have been observed. PMID: 27918982
  20. Persistent histone H3 serine 10 or serine 28 phosphorylation plays a key role in chemical carcinogenesis by 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 suggests 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. H3K9me3 plays a significant role in hypoxia, apoptosis, and the repression of APAK. PMID: 25961932
  27. Collectively, the authors verified that histone H3 is a real substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. Findings indicate that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data demonstrate that double mutations on the residues in the interface (L325A/D328A) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Research indicates that minichromosome maintenance protein 2 (MCM2) binding is not required for the 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. Data suggest that 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 the 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 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 H3K56 acetylation that HIST1H3A (Ab-56) antibody detects?

H3K56 acetylation occurs in the globular domain of histone H3 rather than in the N-terminal tail where most other histone modifications are found. This modification is particularly significant because it directly affects DNA-histone interactions by disrupting the contact between histone H3 and the DNA phosphate backbone. Unlike many histone tail modifications, H3K56Ac modifies the structural core of the nucleosome, potentially opening chromatin and enabling various DNA-templated processes .

In yeast, H3K56Ac is globally abundant and plays critical roles in multiple cellular processes including transcriptional regulation, DNA replication, DNA repair, and nucleosome assembly . While initially thought to be rare in mammals, research has confirmed the presence of H3K56Ac in human cells, particularly in embryonic stem cells where it co-localizes with pluripotency factors such as NANOG, SOX2, and OCT4 . The modification has dual functions - serving as both a genome-wide activator of transcription while also repressing promiscuous transcription after replication fork passage .

How is H3K56 acetylation regulated in the cell?

The regulation of H3K56 acetylation involves multiple enzymes and chaperone proteins in a carefully orchestrated process. In Saccharomyces cerevisiae (budding yeast), H3K56 acetylation is catalyzed by the histone acetyltransferase Rtt109 . This enzyme requires histone chaperones for its activity, with two primary complexes involved:

  • Rtt109-Asf1 complex: Preferentially acetylates H3K56

  • Rtt109-Vps75 complex: Preferentially acetylates H3K9

The histone chaperone Asf1 plays a crucial role, as it binds to H3-H4 dimers and presents them to Rtt109 for acetylation. Studies show that deletion of ASF1 causes larger reductions in H3K56Ac levels compared to H3K9Ac or H3K23Ac in vivo .

Deacetylation of H3K56Ac is primarily mediated by the sirtuins Hst3 and Hst4 in yeast. The double deletion mutant (hst3Δ hst4Δ) exhibits hyperacetylation of H3K56, with essentially all H3 molecules being K56 acetylated throughout the genome and during the entire cell cycle . This hyperacetylation is associated with severe phenotypes that can be attenuated by mutating H3K56 to a nonacetylable arginine residue, suggesting that proper regulation of H3K56Ac levels is critical for normal cellular function .

What experimental controls should be included when using HIST1H3A (Ab-56) antibody in chromatin immunoprecipitation experiments?

When performing chromatin immunoprecipitation (ChIP) experiments with HIST1H3A (Ab-56) antibody, several essential controls should be included:

  • Genetic controls: Include samples from cells with H3K56R mutation (lysine to arginine substitution that prevents acetylation) when possible, as these provide a definitive negative control for antibody specificity .

  • Enzyme inhibition controls: Samples from cells treated with inhibitors of H3K56 acetyltransferases (e.g., cells lacking Rtt109 or Asf1 in yeast models) will have decreased H3K56Ac levels and should show reduced signal .

  • Peptide competition assay: Pre-incubating the antibody with acetylated H3K56 peptides should block specific binding during ChIP.

  • Immunoblot validation: Prior to ChIP experiments, validate antibody specificity using western blot analysis of nuclear extracts from wild-type cells versus cells with H3K56R mutation or cells lacking the relevant acetyltransferases.

  • Input controls: Include input chromatin controls (pre-immunoprecipitation) to normalize ChIP signals and account for differences in chromatin preparation efficiency.

  • IgG control: Use non-specific IgG antibodies matched to the host species of the HIST1H3A (Ab-56) antibody to assess background binding.

  • Cell cycle synchronization: Given that H3K56Ac levels fluctuate during the cell cycle (being highest during S phase), synchronized cell populations at different cell cycle stages can serve as biological controls for expected signal variation .

How can I optimize detection of H3K56Ac in mammalian cells where the modification may be less abundant than in yeast?

Detecting H3K56Ac in mammalian cells presents unique challenges due to its relatively lower abundance compared to yeast. Mass spectrometry analysis has detected H3K56Ac in human cells at approximately 0.04% of total H3 molecules, which is significantly lower than the abundance of other H3 N-terminal acetylation sites (3.8% to 22.9%) in HeLa cells . To optimize detection:

  • Increase starting material: Use larger amounts of starting chromatin material to compensate for lower abundance of the modification.

  • Cell type selection: Focus on cell types where H3K56Ac has been demonstrated to be more abundant, such as embryonic stem cells where it plays a role in pluripotency regulation .

  • Crosslinking optimization: Titrate formaldehyde concentration (0.5-2%) and crosslinking times (5-20 minutes) to optimize epitope accessibility while maintaining chromatin integrity.

  • Sonication parameters: Carefully optimize sonication parameters to generate consistent fragment sizes (200-500bp) without damaging epitopes.

  • Antibody concentration: Perform antibody titration experiments to determine the optimal antibody-to-chromatin ratio for maximum specific signal with minimal background.

  • Extended incubation: Consider longer antibody incubation times (overnight at 4°C) to improve capture of low-abundance modifications.

  • Sequential ChIP: For highly specific detection, perform sequential ChIP (re-ChIP) using antibodies against general H3 followed by H3K56Ac-specific antibody.

  • Signal amplification methods: Consider employing amplification methods in downstream detection steps, such as using enhanced chemiluminescence substrates for western blots or amplification steps in ChIP-seq library preparation.

What is the relationship between H3K56Ac and other histone modifications, and how can this be analyzed experimentally?

H3K56Ac exists within a complex landscape of histone modifications that collectively regulate chromatin structure and function. Analyzing the relationships between H3K56Ac and other modifications requires sophisticated experimental approaches:

  • Sequential ChIP (re-ChIP): This technique can determine co-occurrence of H3K56Ac with other modifications on the same nucleosome. For example, sequential immunoprecipitation with H3K56Ac antibody followed by antibodies against modifications like H3K4me3 or H3K9Ac can identify genomic regions where these marks co-exist.

  • Mass spectrometry analysis: Tandem mass spectrometry of histone extracts can quantitatively assess the co-occurrence of H3K56Ac with other modifications on the same histone molecule. This approach can reveal synergistic or antagonistic relationships between modifications.

  • Combinatorial genetic approaches: Analyzing cells with mutations in enzymes responsible for different histone modifications (e.g., Rtt109 for H3K56Ac, Set1 for H3K4me) can reveal functional interactions between these pathways.

  • Genome-wide correlation analyses: Comparing ChIP-seq datasets for H3K56Ac and other histone marks can identify patterns of co-occurrence or mutual exclusivity across the genome. Research has shown that in human embryonic stem cells, H3K56Ac overlaps with pluripotency factors at both active and inactive promoters, displaying a distribution pattern that differs from other active histone marks such as H3K4me3 and H3K9Ac .

  • Chromatin state analysis: Integrating multiple histone modification datasets (including H3K56Ac) can define specific chromatin states associated with different functional genomic elements.

The data suggest that H3K56Ac serves unique functions distinct from other active histone marks. For instance, while H3K56Ac overlaps with active marks at transcriptionally active regions, it also plays roles in replication-coupled nucleosome assembly that are not shared by marks like H3K4me3 .

How does H3K56Ac influence DNA repair processes, and what experimental setups can best investigate this function?

H3K56Ac plays a significant role in DNA repair processes, particularly in facilitating access of repair machinery to damaged DNA. To investigate this function:

  • AP site repair assays: Studies have shown that H3K56Ac enhances AP endonuclease 1 (APE1) activity in base excision repair (BER). In in vitro studies using acetylated, well-positioned nucleosome core particles (H3K56Ac-601-NCPs), APE1 strand incision was enhanced compared to unacetylated wild-type NCPs . To study this:

    • Generate defined nucleosome substrates containing site-specific DNA lesions (e.g., AP sites)

    • Compare repair efficiency in nucleosomes containing H3K56Ac versus unmodified H3

    • Measure repair kinetics using purified repair enzymes and quantitative assays

  • Double-strand break repair analysis: Design experiments to analyze recruitment of repair factors to induced double-strand breaks (DSBs) in the presence or absence of H3K56Ac:

    • Use systems like I-SceI or CRISPR-Cas9 to induce site-specific DSBs

    • Compare repair factor recruitment and repair efficiency in wild-type cells versus cells with H3K56R mutation or lacking Rtt109

    • Measure chromatin accessibility at repair sites using assays like ATAC-seq

  • DNA damage sensitivity assays: Compare sensitivity of wild-type cells versus cells with defects in H3K56 acetylation to various DNA damaging agents:

    • UV radiation (nucleotide excision repair)

    • Methyl methanesulfonate (base excision repair)

    • Ionizing radiation (double-strand break repair)

    • Hydroxyurea (replication stress)

  • Interactions with repair proteins: Investigate how H3K56Ac affects the recruitment and activity of specific repair proteins:

    • The high-mobility group box 1 protein (HMGB1) enhances APE1 activity in wild-type nucleosomes, but this effect is not observed in H3K56Ac-containing nucleosomes

    • This suggests H3K56Ac may modulate interactions between repair proteins and chromatin, which can be studied using biochemical interaction assays and functional repair assays

What role does H3K56Ac play in nucleosome assembly, and how can ChIP-seq data with HIST1H3A (Ab-56) antibody inform our understanding of this process?

H3K56Ac plays a critical role in replication-coupled nucleosome assembly through its interactions with histone chaperones. ChIP-seq data using HIST1H3A (Ab-56) antibody can provide valuable insights into this process:

  • Histone chaperone interactions: H3K56Ac enhances the association of histone H3 with the histone chaperones CAF-1 and Rtt106, which are crucial for nucleosome assembly. Studies show that:

    • H3 molecules associated with CAF-1, Rtt106, and Asf1 are acetylated at K56

    • The H3K56R mutation significantly reduces the binding of H3 to CAF-1 and Rtt106 but not to Asf1

    • In cells lacking H3K56Ac (rtt109Δ or asf1Δ mutants), the association of H3 with CAF-1 and Rtt106 is dramatically reduced

    • In vitro binding assays show that purified CAF-1 and Rtt106 bind more efficiently to H3 acetylated at K56 than to unacetylated H3

  • ChIP-seq experimental design for studying nucleosome assembly:

    • Perform ChIP-seq for H3K56Ac in synchronized cell populations at different cell cycle stages, focusing on S phase when replication-coupled assembly occurs

    • Compare H3K56Ac distribution to the localization of replication machinery (e.g., PCNA)

    • Conduct parallel ChIP-seq for histone chaperones (CAF-1, Rtt106) to identify sites of co-localization with H3K56Ac

    • Include cell cycle synchronization controls to distinguish replication-coupled from replication-independent nucleosome assembly

  • Data analysis approaches:

    • Analyze H3K56Ac enrichment patterns relative to replication origins

    • Compare H3K56Ac distribution in G1 versus S phase to identify replication-specific patterns

    • Integrate with nascent strand sequencing data to correlate H3K56Ac with active replication

    • Perform metagene analysis to assess H3K56Ac patterns across gene bodies and regulatory elements

  • Functional validation experiments:

    • Complement ChIP-seq with nucleosome occupancy assays (MNase-seq) in wild-type versus H3K56 acetylation-deficient cells

    • Use FRAP (Fluorescence Recovery After Photobleaching) to measure histone incorporation rates at specific genomic loci

    • Employ pulse-chase experiments with tagged histones to track newly synthesized histones during replication

How can I accurately determine H3K56Ac dynamics during cellular differentiation using HIST1H3A (Ab-56) antibody?

Investigating H3K56Ac dynamics during cellular differentiation requires careful experimental design:

  • Differentiation system selection: Choose a well-characterized differentiation system, such as:

    • Embryonic stem cells (ESCs) to defined lineages (neural, cardiac, etc.)

    • Induced pluripotent stem cells (iPSCs) to various cell types

    • Hematopoietic stem cells to blood cell lineages

  • Temporal sampling strategy:

    • Collect samples at multiple timepoints during differentiation (undifferentiated, early, intermediate, and terminally differentiated states)

    • Include biological replicates at each timepoint for statistical robustness

    • Maintain parallel cultures for validation of differentiation status using established markers

  • Integrated ChIP-seq approach:

    • Perform ChIP-seq for H3K56Ac at each differentiation timepoint

    • In parallel, conduct ChIP-seq for:

      • Pluripotency factors (NANOG, SOX2, OCT4) in ESC systems

      • Lineage-specific transcription factors

      • Other histone modifications (H3K4me3, H3K27me3) for comparison

    • Include RNA-seq to correlate H3K56Ac dynamics with gene expression changes

  • Data analysis strategies:

    • Identify sites where H3K56Ac changes during differentiation

    • Perform Gene Ontology analysis of H3K56Ac-associated genes

    • Conduct motif analysis to identify transcription factor binding sites associated with H3K56Ac

    • Integrate with published datasets on chromatin accessibility (ATAC-seq) and chromosome conformation (Hi-C)

  • Validation experiments:

    • Confirm ChIP-seq findings with ChIP-qPCR at select loci

    • Use CRISPR-based approaches to alter H3K56Ac at specific loci and assess impact on differentiation

    • Employ reporter assays to test the functional significance of H3K56Ac at developmental gene promoters

Research has shown that in human embryonic stem cells, H3K56Ac is present at both active and inactive promoters where it colocalizes with key pluripotency regulators NANOG, SOX2, and OCT4. Notably, during cellular differentiation, H3K56Ac relocates to developmental genes, suggesting a functional role in cell fate determination . This pattern makes H3K56Ac a more accurate reflection of the epigenetic differences between hESCs and somatic cells than other active histone marks such as H3K4 trimethylation and K9 acetylation .

What is the relationship between H3K56Ac and cellular aging, and how can this be investigated experimentally?

H3K56 acetylation has been implicated in cellular aging processes, particularly in yeast models. Research shows that proper regulation of H3K56Ac levels is critical for normal lifespan:

  • Impact on replicative lifespan:

    • Deletion of genes encoding the H3K56 acetyltransferase machinery (ASF1, RTT109) results in shortened replicative lifespan in yeast

    • The replicative lifespan was indistinguishable between strains deleted for ASF1, RTT109, or both together, indicating they function in the same pathway to promote achievement of a full lifespan

    • Yeast strains with the H3K56R mutation (preventing acetylation) show drastically reduced lifespan

    • Interestingly, overexpression of Asf1 (which increases H3K56Ac levels) led to reduced median lifespan, with a 19% reduction when induced with 0.5% galactose and a 28% reduction when induced with 1% galactose

  • Experimental approaches to study H3K56Ac in aging:

    • Replicative lifespan assays: Monitor cell division capacity of individual yeast cells with varying H3K56Ac levels

    • H3K56Ac quantification during aging: Measure changes in H3K56Ac levels in young versus aged cells using both global approaches (Western blotting, mass spectrometry) and locus-specific methods (ChIP-qPCR)

    • Genetic manipulation: Compare lifespan effects of H3K56 mutants (H3K56R, H3K56Q) and enzyme deletions (asf1Δ, rtt109Δ, hst3Δ hst4Δ)

    • Correlation with age-associated transcriptional changes: Perform RNA-seq in conjunction with H3K56Ac ChIP-seq in young and aged cells

  • Balance is critical:

    • The data suggest that both the inability to acetylate H3K56 and excessive/persistent H3K56 acetylation lead to lifespan shortening

    • This indicates a delicate balance of H3K56Ac levels is required for optimal lifespan, with both too little and too much being detrimental

  • Connection to histone levels during aging:

    • Research suggests that histone protein levels drop during aging

    • The short lifespan of cells that fail to acetylate H3 on K56 might be related to the requirement of H3K56Ac and Asf1 for accurate expression of cell cycle regulated histone genes

How does H3K56Ac influence transcription, and what techniques with HIST1H3A (Ab-56) antibody can best reveal these mechanisms?

H3K56Ac plays complex roles in transcriptional regulation, functioning as both an activator and repressor in different contexts. Advanced techniques using HIST1H3A (Ab-56) antibody can help elucidate these mechanisms:

  • Dual roles in transcription:

    • H3K56Ac acts as a genome-wide activator of transcription in yeast

    • It has major impact on transcription initiation but also appears to promote elongation and/or termination

    • Conversely, H3K56Ac represses promiscuous transcription that occurs immediately following replication fork passage, in this case by promoting efficient nucleosome assembly

  • Nascent RNA sequencing approaches:

    • Net-seq (native elongating transcript sequencing) combined with H3K56Ac ChIP-seq can reveal the immediate impact of H3K56Ac on transcription

    • This approach has shown that H3K56Ac has different effects on transcription depending on the cell cycle stage and genomic context

    • Time-course experiments throughout the cell cycle can reveal how H3K56Ac differentially affects transcription in G1, S, and G2 phases

  • ChIP-seq analysis techniques:

    • Compare H3K56Ac distribution to RNA polymerase II occupancy and phosphorylation states

    • Analyze H3K56Ac enrichment at promoters versus gene bodies to distinguish initiation from elongation effects

    • Assess H3K56Ac dynamics at genes with different expression levels and transcriptional responses

  • Nucleosome dynamics studies:

    • Combine H3K56Ac ChIP-seq with assays of nucleosome positioning and turnover

    • H3K56Ac enhances promoter-proximal nucleosome turnover, which can be measured using techniques like CATCH-IT (Covalent Attachment of Tags to Capture Histones and Identify Turnover)

    • Correlate nucleosome dynamics with transcriptional activity in wild-type versus H3K56 acetylation-deficient cells

  • Functional transcription assays:

    • Reporter assays with promoters containing different nucleosome configurations

    • In vitro transcription assays using chromatin templates with or without H3K56Ac

    • Measurement of transcription factor binding affinity to nucleosomes containing H3K56Ac versus unmodified nucleosomes

This comprehensive analysis reveals that H3K56Ac functions in context-dependent ways to regulate transcription, with its effects depending on genomic location, cell cycle stage, and interaction with other chromatin factors.

What are the cutting-edge applications of HIST1H3A (Ab-56) antibody in current epigenetic research?

HIST1H3A (Ab-56) antibody is becoming increasingly valuable in cutting-edge epigenetic research due to the unique properties of H3K56 acetylation as a core nucleosomal modification with diverse biological functions. Current applications include:

  • Single-cell epigenomics: Adapting ChIP protocols for single-cell analysis of H3K56Ac to understand cell-to-cell variation in stem cell populations and during differentiation. This approach can reveal how H3K56Ac heterogeneity contributes to cell fate decisions and developmental plasticity.

  • Spatial epigenomics: Combining H3K56Ac detection with methods like Slide-seq or spatial transcriptomics to understand how this modification varies across tissues and contributes to regional specialization during development.

  • Therapeutic epigenetic targeting: Using H3K56Ac profiles to identify targetable epigenetic vulnerabilities in diseases like cancer, where abnormal H3K56Ac patterns may contribute to pathogenesis.

  • Synthetic biology applications: Engineering chromatin with defined H3K56Ac patterns to control gene expression and cellular identity in regenerative medicine applications.

  • Environmental epigenetics: Investigating how environmental factors affect H3K56Ac distribution and how these changes may contribute to disease susceptibility and adaptive responses.

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.