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.
The HIST1H3A (Ab-23) antibody has been rigorously validated across multiple applications:
| Application | Validation Status | Species Reactivity |
|---|---|---|
| ELISA | Validated | Human, Mouse, Rat |
| Western Blotting (WB) | Validated | Human, Mouse |
| Immunohistochemistry (IHC) | Validated | Human |
| Immunofluorescence (IF) | Validated | Human, Mouse |
| Chromatin Immunoprecipitation (ChIP) | Validated | Human |
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 .
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 .
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 .
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 .
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 .
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 .
| Issue | Possible Causes | Solutions |
|---|---|---|
| Weak or no signal in Western blot | Insufficient histone extraction; Degraded acetylation marks | Use acid extraction; Add HDAC inhibitors to all buffers; Verify protein transfer |
| High background in IF | Inadequate blocking; Too high antibody concentration | Increase blocking time; Optimize antibody dilution; Include 0.1% Tween-20 in wash buffers |
| Poor enrichment in ChIP | Inefficient chromatin fragmentation; Suboptimal antibody concentration | Optimize sonication; Titrate antibody amount; Increase incubation time |
| Non-specific bands in Western blot | Cross-reactivity; Sample degradation | Use peptide competition controls; Add protease inhibitors; Optimize washing conditions |
| Variable results between experiments | Biological variation in acetylation levels; Technical variability | Standardize 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 .
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.
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.
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 .
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 .
A detailed comparison between polyclonal HIST1H3A (Ab-23) antibody and monoclonal options:
| Characteristic | Polyclonal HIST1H3A (Ab-23) | Monoclonal H3K23ac Antibodies |
|---|---|---|
| Epitope Recognition | Multiple epitopes around K23ac | Single epitope, highly specific |
| Batch-to-Batch Variability | Moderate to high | Minimal (especially for recombinant) |
| Signal Intensity | Generally stronger signal | May require signal amplification |
| Tolerance to Fixation | More robust to different fixation methods | More sensitive to fixation conditions |
| Cross-Reactivity | Potential cross-reactivity with similar modifications | Less cross-reactivity, higher specificity |
| Cost | Generally more economical | Typically more expensive |
| Applications | Excellent for ChIP, WB, IF | Superior for highly specific applications |
| Sensitivity to Epitope Changes | Can recognize partially denatured epitopes | More sensitive to epitope accessibility |
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 .
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 .