HIST1H3A (Ab-23) 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 orders. Delivery time may vary depending on the purchasing method or 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 H3A is a core component of nucleosomes, which are fundamental structural units of chromatin. Nucleosomes wrap and compact DNA, thereby limiting its accessibility to cellular machineries that utilize DNA as a template. Histones, including H3A, play a crucial role in regulating gene expression, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is tightly regulated through a complex interplay of post-translational modifications of histones, often referred to as the 'histone code,' and nucleosome remodeling processes.
Gene References Into Functions
  1. Research suggests that histone H3 ubiquitination, mediated by the E3 ubiquitin ligase NEDD4, plays a critical role in epigenetic regulation within the context of cancer. PMID: 28300060
  2. Increased expression of H3K27me3 during a patient's clinical course may indicate the presence of heterochronous tumors. PMID: 29482987
  3. JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions associated with a DNA damage response. PMID: 28982940
  4. Findings indicate that the Ki-67 antigen proliferative index has limitations, and phosphohistone H3 (PHH3) emerges as a viable alternative proliferative marker. PMID: 29040195
  5. This data suggests that cytokine-induced histone 3 lysine 27 trimethylation acts as a mechanism to stabilize gene silencing in macrophages. PMID: 27653678
  6. In the early developing human brain, HIST1H3B constitutes the predominant H3.1 transcript among H3.1 isoforms. PMID: 27251074
  7. Among 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. PMID: 26517431
  8. The histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral DNA, and deposits histone H3.3 onto these viral genomes. PMID: 28981850
  9. PHF13 specifically binds to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl), acting as a transcriptional co-regulator. PMID: 27223324
  10. The recognition of hemi-methylated CpGs DNA by UHRF1 triggers its ubiquitylation towards multiple lysines on the H3 tail, adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This research 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), which is associated with poor prognosis and influences EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation is not uncommon in adult cerebellar HGG. PMID: 28547652
  14. Lysyl oxidase-like 2 (LOXL2) acts as a histone modifier enzyme, removing 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 Dbf4 transcription levels were highest, while H3K9me3 levels were greatest during and immediately after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. The interaction 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) involves molecular recognition of secondary structure motifs within the H3 tail, potentially representing an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate the formation of the preinitiation complex. PMID: 27679476
  19. Traffic-derived airborne particulate matter exposures in leukocytes induce histone H3 modifications. PMID: 27918982
  20. Persistent histone H3 serine 10 or serine 28 phosphorylation plays a crucial 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, a predisposition to recurrence, and location in the right cerebellar hemisphere. Histone 3 mutations, on the other hand, are not commonly observed in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to efficiently control the DHRS4 gene cluster. PMID: 26864944
  23. Nuclear antigen Sp100C acts as a multifaceted sensor for histone H3 methylation and phosphorylation. PMID: 27129259
  24. Histone H3 threonine 118 phosphorylation via Aurora-A alters chromatin structure during specific phases of mitosis, promoting timely condensin I and cohesin dissociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA triggers a conformational change in UHRF1, facilitating its recognition of H3 histone. PMID: 27045799
  26. H3K9me3 plays a crucial role in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. In vivo studies using Raji cells treated with staurosporin have confirmed that histone H3 is a genuine substrate for GzmA. PMID: 26032366
  28. Circulating H3 levels correlate with mortality in sepsis patients and exhibit an inverse correlation with antithrombin levels and platelet counts. PMID: 26232351
  29. Mutations at residues L325A/D328A in the interface of KDM5B (lysine (K)-specific demethylase 5B) significantly decrease its histone H3 H3K4me2/3 demethylation activity. PMID: 24952722
  30. MCM2 binding is not required for the incorporation of histone H3.1-H4 into chromatin but plays a critical role in maintaining the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a critical role in the maintenance of leukemia stem cells (LSCs). 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 successfully employed for the analysis of histone post-translational modifications (PTMs). PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevents IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. De novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are regulated by a delicate balance of histone H3K9 acetylation and methylation. 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 acetylation at lysine 23 significant in epigenetic research?

HIST1H3A (Histone Cluster 1, H3a) is a core component of nucleosomes that wrap and compact DNA into chromatin. Nucleosomes play a central role in limiting DNA accessibility to cellular machineries requiring DNA as a template. Histones are critical for transcription regulation, DNA repair, DNA replication, and chromosomal stability .

The acetylation at lysine 23 (acLys23) is a specific post-translational modification that contributes to the histone code, which regulates DNA accessibility. This modification is particularly important because:

  • It occurs in the globular domain of histone H3

  • It influences nucleosome stability

  • It serves as a marker for transcriptionally active chromatin regions

  • It interacts with specific reader proteins that mediate downstream effects

This specific modification allows researchers to track particular chromatin states associated with gene expression and epigenetic regulation.

What applications has the HIST1H3A (Ab-23) antibody been validated for?

The HIST1H3A (Ab-23) antibody has been rigorously validated across multiple applications:

ApplicationValidation StatusSpecies Reactivity
ELISAValidatedHuman, Mouse, Rat
Western Blotting (WB)ValidatedHuman, Mouse
Immunohistochemistry (IHC)ValidatedHuman
Immunofluorescence (IF)ValidatedHuman, Mouse
Chromatin Immunoprecipitation (ChIP)ValidatedHuman

The antibody is particularly valuable for chromatin immunoprecipitation studies, making it an essential tool for researchers investigating histone modifications and their role in gene regulation .

How does HIST1H3A (Ab-23) antibody differ from other histone H3 modification-specific antibodies?

The HIST1H3A (Ab-23) antibody specifically recognizes the acetylation at lysine 23 on histone H3.1, distinguishing it from antibodies targeting other modifications such as:

  • H3K27me3 (trimethylation at lysine 27), which is associated with gene silencing

  • H3K4me3 (trimethylation at lysine 4), which is associated with active promoters

  • H3K9me3 (trimethylation at lysine 9), which is associated with heterochromatin

  • H3K36me3 (trimethylation at lysine 36), which is enriched in actively transcribed regions

This specificity is crucial as different modifications can have opposing effects on chromatin structure and gene expression. The antibody was developed using a peptide sequence specifically around the site of Lys (23) derived from Human Histone H3.1, ensuring high selectivity for this particular modification .

What are the optimal conditions for using HIST1H3A (Ab-23) antibody in ChIP experiments?

For optimal ChIP results with HIST1H3A (Ab-23) antibody, follow these guidelines:

Chromatin Preparation:

  • Cross-link cells with 1% formaldehyde for 10 minutes at room temperature

  • Quench with 125 mM glycine for 5 minutes

  • Lyse cells and sonicate chromatin to fragments of 200-500 bp

  • Verify fragment size by agarose gel electrophoresis

Immunoprecipitation:

  • Use 2-5 μg of HIST1H3A (Ab-23) antibody per ChIP reaction

  • Incubate with chromatin overnight at 4°C with rotation

  • Add protein A/G magnetic beads and incubate for 2-3 hours

  • Perform stringent washing steps to reduce background

  • Elute bound chromatin and reverse cross-links

Controls:

  • Include a non-specific IgG control

  • Use input chromatin (pre-immunoprecipitation) as a reference

  • Consider including a positive control targeting a highly abundant histone mark

This protocol has been validated in human cell lines including HeLa and U2OS, showing specific enrichment at transcriptionally active regions .

How should sample preparation be optimized for Western blotting with HIST1H3A (Ab-23) antibody?

For successful Western blotting with HIST1H3A (Ab-23) antibody:

Histone Extraction:

  • Use acid extraction methods (e.g., 0.2N HCl or triton extraction) to isolate histones

  • Include HDAC inhibitors (e.g., sodium butyrate, TSA) in lysis buffers to preserve acetylation marks

  • Add protease inhibitors to prevent degradation

  • Include phosphatase inhibitors if examining phosphorylation marks simultaneously

Gel Electrophoresis and Transfer:

  • Use 15-18% SDS-PAGE gels for optimal histone separation

  • Consider using Triton-Acid-Urea (TAU) gels for better resolution of modified histones

  • Transfer to PVDF membranes at low voltage (30V) overnight for efficient transfer of small proteins

Antibody Incubation:

  • Block with 5% BSA (not milk, which contains bioactive proteins)

  • Use antibody at 1:500 to 1:2000 dilution

  • Incubate overnight at 4°C for best results

Detection:

  • Use ECL or fluorescent secondary antibodies for detection

  • Expected band size is approximately 15-17 kDa

This approach has been validated on human and mouse cell lines and tissue samples, showing specific detection of H3K23ac modification .

What considerations are important when using HIST1H3A (Ab-23) antibody for immunofluorescence?

For optimal immunofluorescence with HIST1H3A (Ab-23) antibody:

Fixation Methods:

  • 4% paraformaldehyde (10 minutes at room temperature) preserves most epitopes

  • Methanol fixation (-20°C for 10 minutes) may better expose some nuclear epitopes

  • Avoid over-fixation, which can mask the epitope

Permeabilization:

  • Use 0.1-0.5% Triton X-100 for nuclear antigens

  • Include a permeabilization step (10 minutes at room temperature)

Antigen Retrieval:

  • Heat-mediated antigen retrieval with citrate buffer (pH 6.0) may improve signal

  • For tissue sections, enzymatic retrieval using proteinase K can be effective

Antibody Incubation:

  • Dilute antibody 1:100 to 1:500 in blocking buffer

  • Incubate overnight at 4°C for best results

  • Consider adding 0.1% Tween-20 to reduce background staining

Controls:

  • Include a negative control (primary antibody omitted)

  • Consider a peptide competition assay to validate specificity

Researchers have successfully used these conditions for detecting H3K23ac in human cell lines including HeLa and 22Rv1, with strong nuclear staining patterns corresponding to euchromatic regions .

How can I distinguish between true H3K23ac signals and non-specific binding in my experiments?

To ensure signal specificity when using HIST1H3A (Ab-23) antibody:

Validation Approaches:

  • Peptide Competition Assay: Pre-incubate antibody with excess H3K23ac peptide before use. True signals should disappear.

  • Knockout/Knockdown Controls: Use cells where the H3 acetyltransferase responsible for K23 acetylation is depleted.

  • HDAC Inhibition: Treatment with HDAC inhibitors should increase global H3K23ac levels.

  • Correlation With Known Marks: H3K23ac should positively correlate with other active chromatin marks (H3K4me3, H3K27ac).

Common False Positives:

  • Cross-reactivity with other acetylated lysines on H3 (K18, K27)

  • Non-specific binding to highly charged nuclear proteins

  • Inadequate blocking leading to high background

Data Analysis Considerations:

  • In ChIP-seq, compare enrichment patterns with published datasets

  • For imaging, quantify nuclear/cytoplasmic signal ratios

  • For Western blots, verify molecular weight and compare with positive controls

When properly validated, H3K23ac signals should show nuclear localization and enrichment at actively transcribed genes and enhancers .

What are the most common issues encountered with HIST1H3A (Ab-23) antibody and how can they be resolved?

IssuePossible CausesSolutions
Weak or no signal in Western blotInsufficient histone extraction; Degraded acetylation marksUse acid extraction; Add HDAC inhibitors to all buffers; Verify protein transfer
High background in IFInadequate blocking; Too high antibody concentrationIncrease blocking time; Optimize antibody dilution; Include 0.1% Tween-20 in wash buffers
Poor enrichment in ChIPInefficient chromatin fragmentation; Suboptimal antibody concentrationOptimize sonication; Titrate antibody amount; Increase incubation time
Non-specific bands in Western blotCross-reactivity; Sample degradationUse peptide competition controls; Add protease inhibitors; Optimize washing conditions
Variable results between experimentsBiological variation in acetylation levels; Technical variabilityStandardize cell culture conditions; Include internal controls; Normalize to total H3

These troubleshooting approaches have been validated across multiple research settings and can significantly improve experimental outcomes when working with this antibody .

How does the dynamic nature of histone acetylation affect experimental design with HIST1H3A (Ab-23) antibody?

Histone acetylation is highly dynamic, which creates several experimental considerations:

Temporal Factors:

  • Acetylation patterns can change rapidly (within minutes) in response to stimuli

  • Consider time-course experiments to capture dynamics

  • Synchronize cells when studying cell cycle-dependent changes

  • The half-life of H3K23ac may vary between cell types and conditions

Environmental Considerations:

  • Serum starvation can alter global acetylation patterns

  • Cell density affects histone modification levels

  • Stress responses can rapidly change the epigenetic landscape

  • Consider controlling for circadian effects in animal studies

Experimental Approaches:

  • Use HDAC inhibitors as positive controls for increased acetylation

  • Consider pulse-chase experiments with labeled histones

  • For ChIP-seq, validate findings with orthogonal methods (e.g., CUT&RUN)

Research has shown that H3.3 deposition and modification, which includes K23 acetylation, can be induced by stimuli such as interferon and remains stable in non-dividing cells while being diminished in dividing cells . This dynamic nature means experimental conditions must be carefully controlled and timed to ensure reproducibility.

How can HIST1H3A (Ab-23) antibody be used to study the relationship between histone variants and modifications?

The HIST1H3A (Ab-23) antibody can be leveraged to investigate the complex interplay between histone variants and their modifications:

Sequential ChIP Approaches:

  • Perform ChIP-reChIP with antibodies against histone variants (H3.1, H3.3) followed by H3K23ac

  • This can reveal whether K23 acetylation preferentially occurs on specific H3 variants

Mass Spectrometry Integration:

  • Use the antibody for immunoprecipitation followed by mass spectrometry

  • This allows identification of co-occurring modifications on the same histone tail

  • Can reveal "modification signatures" specific to certain genomic regions

Genomic Approaches:

  • Combine ChIP-seq for H3K23ac with other techniques like ATAC-seq

  • This provides insights into chromatin accessibility in relation to this modification

  • Correlation with transcriptomic data (RNA-seq) can reveal functional impacts

Research has shown that H3.3 variant carries higher levels of active marks like K36me3 compared to H3.1, and similar patterns may exist for K23ac . Understanding these relationships is crucial for deciphering the histone code and its biological implications.

What role does H3K23 acetylation play in cell cycle regulation and DNA damage response?

H3K23 acetylation has emerging roles in cell cycle and DNA damage pathways:

Cell Cycle Dynamics:

  • H3.1 deposition occurs primarily during S-phase, but modifications like K23ac show cell-cycle dependent patterns

  • p53 ensures normal behavior and modification of H3.1 during G1/S transition

  • HIST1H3A (Ab-23) antibody can track these changes through cell cycle phases

  • Different from H3.3, which is deposited throughout the cell cycle independent of replication

DNA Damage Response:

  • H3K23ac levels change in response to genotoxic stress

  • May function as a platform for recruitment of DNA repair factors

  • Can be studied using HIST1H3A (Ab-23) antibody before and after damage induction

  • Often works in concert with other modifications like H3S10 phosphorylation

Experimental Approaches:

  • Synchronize cells at different cell cycle stages and analyze H3K23ac patterns

  • Induce DNA damage with agents like neocarzinostatin or UV and track H3K23ac changes

  • Combine with cell cycle markers (e.g., PCNA for S-phase) in co-localization studies

These applications allow researchers to understand how this specific modification contributes to maintaining genomic integrity across different cellular states .

How can HIST1H3A (Ab-23) antibody be incorporated into multi-omics approaches for comprehensive epigenetic profiling?

Integrating HIST1H3A (Ab-23) antibody into multi-omics frameworks:

Combined Genomic Approaches:

  • ChIP-seq for H3K23ac paired with:

    • ATAC-seq (chromatin accessibility)

    • RNA-seq (transcriptome)

    • DNA methylation analysis (methyl-seq)

    • Chromosome conformation capture (Hi-C)

Single-Cell Applications:

  • Single-cell ChIP-seq adaptations for H3K23ac

  • CUT&Tag for improved sensitivity in limited samples

  • Integration with single-cell RNA-seq for direct correlation with gene expression

Spatial Epigenomics:

  • Combine immunofluorescence with RNA-FISH

  • Use multiplexed antibody imaging with H3K23ac and other histone marks

  • Spatially resolved chromatin profiling technologies

Data Integration Strategies:

  • Machine learning approaches to identify epigenetic signatures

  • Network analysis to understand regulatory connections

  • Trajectory analysis for developmental or disease progression studies

This integrated approach provides a comprehensive understanding of how H3K23 acetylation fits within the broader epigenetic landscape and its functional consequences on gene regulation .

How does the sensitivity and specificity of polyclonal HIST1H3A (Ab-23) antibody compare to monoclonal alternatives?

A detailed comparison between polyclonal HIST1H3A (Ab-23) antibody and monoclonal options:

CharacteristicPolyclonal HIST1H3A (Ab-23)Monoclonal H3K23ac Antibodies
Epitope RecognitionMultiple epitopes around K23acSingle epitope, highly specific
Batch-to-Batch VariabilityModerate to highMinimal (especially for recombinant)
Signal IntensityGenerally stronger signalMay require signal amplification
Tolerance to FixationMore robust to different fixation methodsMore sensitive to fixation conditions
Cross-ReactivityPotential cross-reactivity with similar modificationsLess cross-reactivity, higher specificity
CostGenerally more economicalTypically more expensive
ApplicationsExcellent for ChIP, WB, IFSuperior for highly specific applications
Sensitivity to Epitope ChangesCan recognize partially denatured epitopesMore sensitive to epitope accessibility

What emerging technologies will enhance the utility of HIST1H3A (Ab-23) antibody in future epigenetic research?

Emerging technologies poised to expand HIST1H3A (Ab-23) antibody applications:

CUT&Tag and CUT&RUN:

  • Higher sensitivity than traditional ChIP

  • Requires fewer cells and less antibody

  • Better signal-to-noise ratio for H3K23ac detection

  • Works with difficult-to-sonicate samples

Proximity Ligation Assays (PLA):

  • Detect interactions between H3K23ac and reader proteins

  • Visualize spatial relationships between different histone modifications

  • Single-molecule resolution of modification patterns

Mass Cytometry (CyTOF):

  • Multiplexed analysis of dozens of histone modifications

  • Single-cell resolution of epigenetic states

  • Metal-conjugated antibodies eliminate spectral overlap issues

Live-Cell Imaging:

  • Development of H3K23ac-specific intrabodies

  • Real-time tracking of acetylation dynamics

  • FRET-based sensors for histone modification changes

Nanopore Sequencing:

  • Direct detection of modified nucleosomes

  • Long-read epigenetic analysis

  • Simultaneous detection of DNA methylation and histone modifications

These technologies will dramatically expand our understanding of H3K23 acetylation dynamics and function in various biological contexts .

How can HIST1H3A (Ab-23) antibody contribute to understanding disease-associated epigenetic dysregulation?

The HIST1H3A (Ab-23) antibody has significant potential in disease research:

Cancer Epigenetics:

  • Altered H3K23ac patterns in various cancers

  • Association with oncogene activation or tumor suppressor silencing

  • Biomarker potential for cancer subtyping and prognosis

  • Target for epigenetic therapies (HDAC inhibitors)

Neurodegenerative Disorders:

  • Disrupted H3K23ac in Alzheimer's and Parkinson's disease

  • Memory formation involves dynamic histone acetylation

  • HIST1H3A (Ab-23) antibody can track disease progression in brain tissues

Inflammatory Conditions:

  • H3K23ac changes during immune cell activation

  • Association with dysregulated inflammation

  • Potential for monitoring therapeutic interventions

  • Interferon response genes show H3.3 deposition and modification

Research Approaches:

  • Patient-derived samples compared to healthy controls

  • Animal models of disease progression

  • Drug screening for compounds affecting H3K23ac

  • Integration with genetic risk factors

Understanding how H3K23 acetylation patterns change in disease states may provide new insights into pathogenesis and identify novel therapeutic targets .

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.