HIST1H3A (Ab-6) 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. The delivery time may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timelines.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
Histone H3 is a core component of the nucleosome, the fundamental unit of chromatin. Nucleosomes wrap and compact DNA, limiting its accessibility to cellular machinery that require DNA as a template. Consequently, histones play a critical role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex interplay of post-translational modifications of histones, also known as the histone code, 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. The identification of increased expression of H3K27me3 during a patient's clinical course may be helpful for determining if tumors are heterochronous. 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 trigger a DNA damage response. PMID: 28982940
  4. Evidence suggests that the Ki-67 antigen proliferative index has significant limitations and that phosphohistone H3 (PHH3) is a viable 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. 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, the 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. PMID: 26517431
  8. Data show that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral DNA 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. This study 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 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, 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 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. 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 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) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Minichromosome maintenance protein 2 (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 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 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

Basic Research Questions

  • What is HIST1H3A and what does the HIST1H3A (Ab-6) antibody specifically recognize?

HIST1H3A is a histone variant that belongs to the histone H3 family, specifically histone cluster 1, H3a. The HIST1H3A (Ab-6) antibody specifically recognizes a peptide sequence around the threonine 6 (Thr6) site of human Histone H3.1 . Histones are small, highly basic proteins that consist of a globular domain with unstructured N- and C-terminal tails protruding from the main structure. They play a critical role in nucleosome formation, where two molecules of each of the four core histones (H2A, H2B, H3, and H4) form an octamer around which approximately 146 bp of DNA is wrapped . This antibody is useful for studying the specific N-terminal modifications of histone H3 that occur near the Thr6 residue.

  • What are the validated applications for the HIST1H3A (Ab-6) antibody?

The HIST1H3A (Ab-6) Polyclonal Antibody has been validated for multiple research applications:

ApplicationValidation Status
ELISAValidated
Western Blotting (WB)Validated
Immunohistochemistry (IHC)Validated
Immunoprecipitation (IP)Validated
Chromatin Immunoprecipitation (ChIP)Validated

For Western blot applications, this antibody typically detects a band at approximately 15 kDa, which corresponds to the molecular weight of histone H3 . The antibody has been tested for reactivity with human samples, though cross-reactivity with other species may occur due to high sequence conservation of histones across species .

  • What experimental conditions are recommended for optimal results with HIST1H3A (Ab-6) antibody in Western blotting?

For optimal Western blotting results with the HIST1H3A (Ab-6) antibody, consider these methodological recommendations:

  • Sample preparation: Use 1% SDS hot lysis method for efficient extraction of nuclear proteins . This is particularly important for histone proteins which are tightly bound to DNA.

  • Recommended dilutions: Start with a dilution range of 1:1000-1:2000 for Western blotting , but optimization may be necessary depending on your specific sample type.

  • Loading controls: When working with histones, traditional loading controls like GAPDH or β-actin may not be optimal. Consider using total H3 antibodies (like ab1791) as loading controls when studying specific histone modifications .

  • Blocking: Use 5% non-fat dry milk or BSA in TBST for blocking.

  • Buffer conditions: For histone extraction, specialized acid extraction protocols may yield better results than standard RIPA buffers .

  • Positive controls: Several cell lines can serve as positive controls, including LNCaP, HEK-293, HeLa, Jurkat, and NIH/3T3 cells, which have been validated to express detectable levels of histone H3 .

  • How should HIST1H3A (Ab-6) antibody be stored to maintain its activity?

For optimal preservation of antibody activity, store the HIST1H3A (Ab-6) antibody at -20°C for long-term storage. The antibody is typically stable for one year after shipment when properly stored . For frequent use and short-term storage (up to one month), the antibody can be kept at 4°C to avoid repeated freeze-thaw cycles which can degrade antibody performance .

The antibody is typically supplied in a storage buffer consisting of PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . Some preparations may also contain small amounts of BSA (0.1%) as a stabilizer. Aliquoting larger volumes into smaller working amounts is recommended to minimize freeze-thaw cycles for antibodies stored at -20°C.

Advanced Research Questions

  • How can I validate the specificity of HIST1H3A (Ab-6) antibody and distinguish it from other histone H3 variants in my experiments?

Validating antibody specificity for histone variants is crucial due to high sequence homology. Implement these methodological approaches:

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application. Signal disappearance confirms specificity for the target epitope.

  • Knockout/knockdown validation: Use cell lines with HIST1H3A knockdown or knockout. The antibody should show reduced or absent signal in these samples .

  • Cross-reactivity testing: Test against recombinant proteins of different histone H3 variants and their modifications. For HIST1H3A (Ab-6), assess potential cross-reactivity with other H3 variants (H3.2, H3.3) as the N-terminal regions can be highly similar .

  • Peptide array analysis: Commercial peptide arrays can comprehensively assess antibody binding to various histone peptides with different modifications .

  • Mass spectrometry validation: Use mass spectrometry to identify proteins immunoprecipitated by your antibody to confirm specificity.

  • Parallel antibody comparison: Compare results with other validated antibodies targeting the same epitope but from different sources or clones .

The Histone Antibody Specificity Database (http://www.histoneantibodies.com) is a valuable resource that catalogs the behavior of commercially available histone antibodies by peptide microarray , which can help in selecting properly validated antibodies.

  • What are the common pitfalls in ChIP experiments using HIST1H3A (Ab-6) antibody and how can they be addressed?

ChIP experiments with histone antibodies present several technical challenges:

  • Epitope masking: Post-translational modifications near the antibody epitope (Thr6) may interfere with antibody binding. Document existing modifications in your cell type before proceeding with ChIP experiments.

  • Cross-reactivity concerns: Some histone antibodies show unexpected cross-reactivity. For example, studies have shown that certain H3K27me3 antibodies can cross-react with H3K4me3-marked histones . Validate your HIST1H3A (Ab-6) antibody with peptide arrays or competition assays before ChIP application.

  • Fixation conditions: Optimize formaldehyde fixation time (typically 10 minutes is standard) , as over-fixation can mask epitopes while under-fixation results in poor chromatin preservation.

  • Sonication parameters: Optimize sonication conditions to achieve chromatin fragments of 200-500 bp. Insufficient fragmentation can lead to high background and poor resolution.

  • Antibody amount: Titrate antibody concentration. For ChIP applications with histone antibodies, start with 3-5 μg of antibody per 25 mg of chromatin .

  • Appropriate controls: Always include:

    • Input control (non-immunoprecipitated chromatin)

    • Negative control (non-specific IgG or no-antibody control)

    • Positive control (a known target region)

  • Data normalization: Normalize ChIP-seq data to total H3 occupancy to distinguish between changes in modification levels versus nucleosome occupancy.

  • How do modifications and variants of histone H3 impact experimental design when using HIST1H3A (Ab-6) antibody?

The complexity of histone modifications significantly impacts experimental design when using HIST1H3A antibodies:

  • Modification crosstalk: Modifications near Thr6 (such as phosphorylation of Ser10 or acetylation of Lys9) may interfere with antibody recognition . When designing experiments, consider the known modification status of your biological sample and how it might affect antibody binding.

  • Variant-specific considerations: There are multiple H3 variants (H3.1, H3.2, H3.3, CENP-A, H3t) with distinct cellular functions . HIST1H3A specifically refers to H3.1, which differs from H3.3 at key amino acid positions. The HIST1H3A (Ab-6) antibody targets the N-terminal region, so consider whether your research question requires variant-specific detection.

  • Cell cycle effects: H3.1 incorporation is primarily replication-dependent, while H3.3 can be incorporated in a replication-independent manner . This means that cellular context (proliferating vs. non-proliferating cells) may affect the interpretation of your results.

  • Developmental timing: The expression patterns of histone variants change during development . In mice, H3.3A is ubiquitously expressed during embryonic development until 13.5 days post-coitum, after which expression becomes tissue-specific .

  • Technical approach selection: Different techniques reveal different aspects of histone biology:

    • ChIP-seq identifies genomic locations

    • Mass spectrometry identifies co-occurring modifications

    • IF/IHC reveals spatial distribution

    • FRAP (Fluorescence Recovery After Photobleaching) assesses dynamics

Understanding these complexities allows for proper experimental design and accurate interpretation of results when using HIST1H3A (Ab-6) antibody.

  • How can I troubleshoot inconsistent results between different lots of HIST1H3A (Ab-6) antibody?

Lot-to-lot variability is a significant challenge in histone antibody research. Here's a methodological approach to troubleshooting:

  • Validation for each lot: For each new lot, perform basic validation experiments:

    • Western blot against known positive controls (HeLa or NIH/3T3 cells)

    • Peptide competition assay

    • Comparison with previous lots on identical samples

  • Standardization protocol:

    • Document detailed conditions for each experiment

    • Use the same positive controls across experiments

    • Maintain consistent sample preparation methods

    • Apply identical blocking conditions and incubation times

  • Reference standards:

    • Establish an internal reference sample that works well

    • Process this reference alongside test samples for direct comparison

    • Consider using recombinant histone standards

  • Technical variables to control:

    • Storage conditions (avoid repeated freeze-thaw cycles)

    • Incubation temperature consistency

    • Buffer preparation methods

    • Sample handling procedures

  • Cross-reference with other antibodies:

    • When possible, validate findings with antibodies targeting different epitopes of the same protein

    • Consider using commercially available histone modification antibody panels that have been validated together

  • Peptide array analysis:

    • If inconsistencies persist, consider submitting different lots to peptide array analysis to map exact binding specificities

Document all observations, as this information may be valuable to both your research group and the antibody manufacturer for quality improvement.

  • How should I design experiments to study the cross-talk between different histone modifications using HIST1H3A (Ab-6) and other histone antibodies?

Designing experiments to study histone modification cross-talk requires careful planning:

  • Sequential ChIP (Re-ChIP) methodology:

    • First, immunoprecipitate chromatin with HIST1H3A (Ab-6) antibody

    • Elute the immunoprecipitated material

    • Perform a second immunoprecipitation with antibodies against other modifications

    • This identifies genomic regions containing both modifications simultaneously

  • Mass spectrometry approach:

    • Immunoprecipitate histones with HIST1H3A (Ab-6) antibody

    • Analyze by mass spectrometry to identify co-occurring modifications

    • Use histone peptides with known modifications as standards

  • Combinatorial antibody testing:

    • Compare ChIP-seq profiles using antibodies against different modifications

    • Analyze overlapping and distinct genomic regions

    • Correlate with gene expression data

  • Genetic manipulation experiments:

    • Use cells with mutations in histone modifying enzymes

    • Analyze how disrupting one modification affects others

    • Compare results between wild-type and mutant cells

  • Imaging approaches:

    • Use immunofluorescence with multiple histone antibodies

    • Perform co-localization analysis

    • Consider super-resolution microscopy for detailed nuclear localization

  • Time-course experiments:

    • Study the temporal order of histone modifications during cellular processes

    • Sample at multiple time points after stimulus

    • Track modification changes during differentiation or gene activation

  • Controls for antibody specificity:

    • Use peptide competition with modified and unmodified peptides

    • Include genetic controls (knockout/knockdown of modifying enzymes)

    • Test antibodies on peptide arrays containing combinatorial modifications

This multifaceted approach can provide comprehensive insights into histone modification cross-talk while controlling for technical variables that might affect interpretation.

  • What emerging technologies are enhancing the utility of HIST1H3A antibodies in epigenetic research?

Recent technological advances are expanding the applications of histone antibodies in epigenetic research:

  • CUT&RUN and CUT&Tag:

    • These techniques offer alternatives to traditional ChIP with higher sensitivity and lower background

    • Require fewer cells than conventional ChIP (as few as 1,000 cells)

    • Provide higher resolution mapping of histone modifications

    • Can be combined with HIST1H3A (Ab-6) antibody for precise localization studies

  • Single-cell epigenomics:

    • Single-cell ChIP-seq and CUT&Tag protocols allow analysis of histone modifications at single-cell resolution

    • Enable study of cellular heterogeneity in epigenetic states

    • Require highly specific antibodies with low background

  • CRISPR-based epigenome editing:

    • dCas9 fused to epigenetic modifiers can introduce specific histone modifications at targeted loci

    • HIST1H3A antibodies are crucial for validating the efficacy of these modifications

    • Allows causal studies of histone modification function

  • Live-cell imaging of histone dynamics:

    • Antibody-derived nanobodies conjugated to fluorescent proteins

    • Enable real-time tracking of histone modifications in living cells

    • Provide spatiotemporal information about epigenetic changes

  • Mass cytometry (CyTOF) with histone antibodies:

    • Allows simultaneous detection of multiple histone modifications at single-cell level

    • Enables correlation of histone states with cell surface markers

    • Provides high-dimensional data for complex epigenetic profiling

  • Highly multiplexed immunofluorescence:

    • Techniques like CODEX or Imaging Mass Cytometry allow visualization of multiple histone marks simultaneously

    • Provide spatial context for histone modifications within tissue architecture

    • Require highly validated antibodies with minimal cross-reactivity

  • Integrative multi-omics approaches:

    • Combine ChIP-seq using HIST1H3A antibodies with RNA-seq, ATAC-seq, and DNA methylation data

    • Provide comprehensive view of epigenetic regulation

    • Machine learning algorithms help identify patterns and correlations

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.