The HIST1H3A (Ab-122) Antibody is a polyclonal rabbit-derived antibody designed to detect acetylation at lysine 122 (K122) on histone H3.1, a variant of the histone H3 family. This post-translational modification is critical for chromatin remodeling, gene regulation, and epigenetic signaling. The antibody is widely used in research to study histone acetylation dynamics in contexts such as cancer, DNA repair, and transcriptional regulation.
The antibody is validated for multiple techniques, with experimental data demonstrating its specificity and utility:
Target Detection: Acetylated H3K122 in human, rat, and mouse cell lysates (e.g., HeLa, Jurkat, 293, HepG2, rat brain/liver) .
Cross-Reactivity: Minimal cross-reactivity observed with unmodified histone H3 or acetylated lysines at other sites (e.g., K4, K9, K27) .
Tissue Analysis: Effective in paraffin-embedded human glioma, lung cancer, and rat brain/liver tissues .
Protocol: Compatible with formalin-fixed, paraffin-embedded (FFPE) samples and methanol-fixed cells .
Enrichment: Used to isolate acetylated H3K122 from HeLa whole-cell lysates, confirming its role in chromatin dynamics .
H3K122 acetylation is a key epigenetic mark linked to gene activation and chromatin accessibility. While the HIST1H3A (Ab-122) Antibody specifically targets acetylation, emerging research highlights the interplay between acetylation and succinylation at this site:
HAT1 as a Dual Enzyme: Histone acetyltransferase 1 (HAT1) catalyzes both acetylation (e.g., H3K27) and succinylation (e.g., H3K122), suggesting a complex regulatory role in chromatin remodeling and glycolysis .
Functional Implications: H3K122 succinylation, mediated by HAT1, is implicated in tumor progression and epigenetic regulation, though this modification is distinct from acetylation .
HIST1H3A, also known as Histone H3.1, is a core histone protein that serves as a primary building block of nucleosomes. These nucleosomes consist of DNA wrapped around histone proteins, forming the fundamental unit of chromatin structure. This packaging allows DNA to be compactly organized within the cell nucleus, enabling vast amounts of genetic information to be accommodated in a relatively small nuclear space .
Histone H3.1 plays a pivotal role in maintaining chromatin architecture and regulating gene expression. As a core component of nucleosomes, it limits DNA accessibility to cellular machineries that require DNA as a template, thereby influencing transcription regulation, DNA repair, DNA replication, and chromosomal stability .
The HIST1H3A (Ab-122) antibody is a polyclonal antibody raised against a specific peptide sequence around lysine 122 of human Histone H3.1 . This distinguishes it from other Histone H3 antibodies that may target different epitopes or modification states.
The antibody's polyclonal nature means it recognizes multiple epitopes on the target protein, potentially offering greater sensitivity than monoclonal antibodies in certain applications, though possibly with reduced specificity .
The HIST1H3A (Ab-122) antibody has been validated for human (Homo sapiens) samples . The antibody has demonstrated effectiveness in multiple applications:
| Application | Validation Status | Recommended Dilution |
|---|---|---|
| ELISA | Validated | Determined by end-user |
| Western Blot (WB) | Validated | Determined by end-user |
| Immunoprecipitation (IP) | Validated | Determined by end-user |
For optimal Western blot detection of HIST1H3A using the Ab-122 antibody, follow these methodological considerations:
Histone extraction: Use specialized histone extraction protocols that employ acid extraction (typically with 0.2N HCl or 0.4N H₂SO₄) to efficiently isolate histones from nuclear proteins.
Sample denaturation: Heat samples in loading buffer containing SDS at 95°C for 5 minutes to ensure complete denaturation.
Gel selection: Use high percentage (15-18%) SDS-PAGE gels or specialized Triton-Acid-Urea (TAU) gels for better resolution of the low molecular weight (approximately 15-17 kDa) histone proteins .
Transfer conditions: Employ a wet transfer system with methanol-containing buffer at constant amperage (typically 250-300 mA) for 60-90 minutes to ensure efficient transfer of these small proteins.
Blocking: Use 5% non-fat dry milk or BSA in TBST for blocking, with BSA often preferred for phospho-specific antibodies.
Antibody incubation: Dilute primary antibody as recommended by manufacturer (typically starting with 1:1000) and incubate overnight at 4°C for optimal binding .
Controls: Include positive controls such as MCF7, JK, K562, or HepG2 cell lysates, which have been validated for HIST1H3A detection .
The expected molecular weight for HIST1H3A is approximately 15-17 kDa, with observed bands typically appearing around 16 kDa .
When optimizing immunohistochemistry protocols for HIST1H3A detection, consider these methodological recommendations:
Tissue fixation: Use 10% neutral buffered formalin for 24-48 hours, as overfixation can mask histone epitopes.
Antigen retrieval: Perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) under high pressure, which is crucial for exposing histone epitopes that may be masked during fixation .
Blocking: Block with 10% normal serum (from the species of the secondary antibody) for 30 minutes at room temperature to reduce background staining .
Primary antibody incubation: Begin with a 1:300 dilution in 1% BSA and incubate overnight at 4°C for optimal sensitivity and specificity .
Detection system: Use a polymer-based detection system rather than avidin-biotin complexes to minimize background, as endogenous biotin can be abundant in some tissues.
Counterstaining: Use light hematoxylin counterstaining to avoid obscuring the nuclear signals where histones are predominantly localized.
Positive tissue controls: Include human gastric cancer or salivary gland tissue sections, which have been validated for strong nuclear HIST1H3A staining .
For successful immunofluorescence detection of HIST1H3A, researchers should consider these critical methodological factors:
Cell fixation: Fix cells in 4% paraformaldehyde for 15 minutes at room temperature to preserve cellular architecture while maintaining epitope accessibility .
Permeabilization: Use 0.2% Triton X-100 in PBS for 10 minutes to allow antibody access to nuclear proteins without excessive extraction of histones.
Blocking: Block with 5% normal serum and 1% BSA in PBS for 1 hour at room temperature to reduce non-specific binding.
Primary antibody dilution: Begin with a 1:500 dilution for Histone H3 antibodies and optimize as needed .
Co-staining recommendations: Combine with cytoskeletal markers (like phalloidin) and DNA stains (DAPI) to provide context for the nuclear histone staining .
Mounting media: Use anti-fade mounting media containing DAPI to preserve fluorescence and counterstain nuclei.
Imaging parameters: Employ confocal microscopy with appropriate negative controls to distinguish specific nuclear staining from background. Expect strong nuclear localization of HIST1H3A signal.
Distinguishing between histone H3 variants (H3.1, H3.2, H3.3, etc.) presents a significant challenge due to their high sequence homology. Advanced researchers should consider these approaches:
Variant-specific antibodies: Some commercial antibodies claim variant specificity, though these should be rigorously validated using knockout or knockdown controls.
Mass spectrometry validation: Use mass spectrometry to confirm the identity of immunoprecipitated histones, which can distinguish variants based on their subtle sequence differences.
Immunoprecipitation followed by sequencing: Perform ChIP-seq to identify genomic locations enriched for specific variants, as H3.3 tends to be enriched at actively transcribed regions while H3.1/H3.2 are incorporated during DNA replication.
Sequential immunoprecipitation: Perform sequential IP using antibodies against specific modifications followed by variant-specific antibodies to enrich for specifically modified variants.
Expression analysis: In certain experimental settings, leverage the fact that canonical H3.1/H3.2 genes are primarily expressed during S-phase while H3.3 is expressed throughout the cell cycle.
Cross-reactivity is a common challenge with histone antibodies due to the high conservation of histone proteins. Researchers should implement these methodological strategies:
Peptide competition assays: Pre-incubate the antibody with excess target peptide to confirm specificity; specific binding should be blocked while non-specific binding will remain.
Knockout/knockdown validation: Use CRISPR-engineered cells lacking the target histone variant or cells with siRNA-mediated knockdown as negative controls.
Dot blot analysis: Test antibody specificity against a panel of histone peptides with different modifications to assess potential cross-reactivity with other modified forms.
Multiple antibody approach: Use multiple antibodies targeting different epitopes of the same protein and compare results for consistency.
Recombinant protein standards: Include purified recombinant histones as positive controls to establish expected signal intensity and molecular weight.
Species control samples: When working with antibodies claimed to have multi-species reactivity, include samples from each species to verify cross-reactivity claims .
For quantitative analysis of histone H3 levels, researchers should employ these rigorous methodologies:
Standardized loading controls: Use total protein staining (e.g., Ponceau S, SYPRO Ruby) rather than typical loading controls like GAPDH, as histone content may change independently of housekeeping proteins.
Absolute quantification: Employ recombinant histone H3 protein standards at known concentrations to generate standard curves for absolute quantification.
Normalization approaches: Normalize H3 signals to total H4 levels (another core histone) or to total histone content for more accurate comparisons between samples with different chromatin compositions.
Multi-technique validation: Combine Western blot quantification with other techniques such as ELISA or mass spectrometry for cross-validation.
Image analysis software: Use appropriate software (ImageJ, Image Lab, etc.) with background subtraction and consistent region-of-interest selection for densitometric analysis.
Statistical analysis: Apply appropriate statistical tests (t-test, ANOVA) to determine if observed differences are significant, with multiple biological replicates (n≥3).
When designing ChIP experiments with HIST1H3A antibodies, researchers should consider these methodological approaches:
Crosslinking optimization: For total H3 occupancy studies, use 1% formaldehyde for 10 minutes at room temperature, as over-crosslinking can mask epitopes and reduce immunoprecipitation efficiency.
Sonication parameters: Optimize sonication conditions to generate chromatin fragments of 200-500 bp, which is optimal for resolution while maintaining sufficient epitope integrity.
IP controls: Include:
Input samples (non-immunoprecipitated chromatin)
IgG negative controls (same species as the primary antibody)
Positive controls using antibodies against abundantly marked regions
Quantitative PCR primer design: Design primers targeting regions with known H3 enrichment (active genes, silenced regions) and depleted regions (such as certain enhancers) as internal controls.
Sequential ChIP: For studying specific H3 variant distribution in combination with certain modifications, perform sequential ChIP with modification-specific antibodies followed by variant-specific antibodies.
ChIP-seq considerations: For genome-wide studies, ensure sufficient sequencing depth (typically 20-30 million uniquely mapped reads) and include spike-in controls for normalization between samples.
To investigate relationships between HIST1H3A and its post-translational modifications (PTMs), researchers should consider these experimental approaches:
Combined immunoprecipitation strategy: Use the HIST1H3A antibody for initial immunoprecipitation, followed by Western blotting with antibodies specific for various PTMs (acetylation at K122 , methylation at K122 , etc.).
Mass spectrometry analysis: Perform IP with HIST1H3A antibody followed by mass spectrometry to identify and quantify the complete pattern of modifications present on the immunoprecipitated histones.
ChIP-reChIP approach: Perform sequential ChIP (first with HIST1H3A antibody, then with modification-specific antibodies) to identify genomic regions where HIST1H3A carries specific modifications.
Inhibitor studies: Combine HIST1H3A analysis with treatments using specific inhibitors of histone-modifying enzymes (HDACs, HATs, methyltransferases) to study the dynamics of these modifications.
Enrichment protocols: Use HIST1H3A antibody in combination with PTM-specific antibodies in a multiplexed IP approach to study how modifications change across different cellular conditions or disease states.
Time-course experiments: Design time-course experiments following cellular perturbations to track how HIST1H3A modifications change temporally, providing insights into modification dynamics.
When studying HIST1H3A in disease contexts, consider these methodological recommendations:
Sample preservation: For clinical samples, optimize fixation protocols (preferably using neutral buffered formalin for 24-48 hours) to preserve histone epitopes while maintaining tissue architecture.
Tissue microarrays: Create tissue microarrays containing multiple patient samples and controls to enable high-throughput comparative analysis while minimizing technical variation.
Quantitative analysis approaches:
For IHC: Use digital pathology with standardized scoring systems
For WB: Employ densitometry with appropriate normalization
For IF: Quantify nuclear intensity through automated image analysis
Cell line models: Select appropriate disease-relevant cell lines that recapitulate the histone dynamics of interest, validating findings through multiple cell lines when possible.
Control selection: Include both healthy tissue controls and disease-relevant controls (e.g., non-malignant adjacent tissue for cancer studies) to distinguish disease-specific changes from tissue-specific patterns.
Correlation with clinical data: Design studies to correlate HIST1H3A levels or modifications with clinical parameters (survival, treatment response) using appropriate statistical methods.
Multi-omics integration: Combine histone analysis with transcriptomic, genomic, or proteomic data to develop integrated models of how histone alterations contribute to disease mechanisms.
To ensure reliable results when working with HIST1H3A antibodies, implement these quality control measures:
Antibody validation: Verify antibody specificity through:
Lot-to-lot consistency: Test new antibody lots against previous lots to ensure consistent performance, as antibody quality can vary between manufacturing batches.
Storage conditions: Store antibodies according to manufacturer recommendations (typically aliquoted and stored at -20°C or -80°C) to maintain activity over time .
Controls for each experiment:
Positive controls: Cell lines or tissues with known HIST1H3A expression
Negative controls: Primary antibody omission, isotype controls
Loading controls: Total protein stains for Western blots
Signal specificity verification:
For WB: Confirm single band at expected molecular weight
For IHC/IF: Verify expected nuclear localization pattern
For IP: Confirm enrichment over IgG control
To rigorously validate HIST1H3A antibody specificity, researchers should implement these methodological approaches:
Multi-antibody validation: Use multiple antibodies targeting different epitopes of HIST1H3A and compare staining patterns and signal intensities.
Genetic validation approaches:
siRNA/shRNA knockdown of HIST1H3A to demonstrate reduced signal
CRISPR/Cas9-mediated knockout as negative control
Overexpression systems to confirm increased signal
Peptide blocking experiments: Pre-incubate antibody with excess immunizing peptide to demonstrate signal reduction in specific binding.
Orthogonal technique confirmation: Validate findings using independent techniques (e.g., mass spectrometry to confirm Western blot findings).
Cross-reactivity assessment: Test the antibody against recombinant H3 variants to assess potential cross-reactivity with other histone variants.
Application-specific controls:
For ChIP: Include IgG controls and known positive/negative genomic regions
For immunostaining: Include competing peptide controls and secondary-only controls
Histone H3.1 (HIST1H3A) plays sophisticated roles in epigenetic regulation and disease through these mechanisms:
Replication-dependent incorporation: Unlike the variant H3.3, H3.1 is predominantly incorporated during DNA replication, creating a foundation for maintaining epigenetic memory through cell divisions .
Modification patterns: H3.1 carries specific patterns of post-translational modifications that differ from other variants, including specific acetylation and methylation at K122, which influence chromatin accessibility and transcription .
Disease associations: Alterations in H3.1 levels or modifications have been implicated in:
Cancer progression, where global changes in histone modifications correlate with clinical outcomes
Developmental disorders resulting from mutations in histone genes or modifying enzymes
Neurodegenerative conditions where chromatin regulation is disrupted
Therapeutic targeting: Emerging therapies target histone-modifying enzymes to restore normal modification patterns on histones including H3.1, with applications in cancer, neurological disorders, and autoimmune diseases.
Histone replacement dynamics: The balance between H3.1 and H3.3 incorporation influences cellular plasticity and differentiation potential, with H3.1 generally associated with more stable, repressive chromatin states.
To investigate HIST1H3A dynamics throughout the cell cycle, researchers should consider these advanced methodological approaches:
Cell synchronization techniques:
Double thymidine block for G1/S boundary synchronization
Nocodazole treatment for M-phase arrest
Serum starvation-release protocols for G0/G1 synchronization
Live-cell imaging approaches:
SNAP-tag or Halo-tag fusion constructs of H3.1 for pulse-chase experiments
Fluorescence recovery after photobleaching (FRAP) to measure H3.1 mobility in different cell cycle phases
Photoactivatable GFP-H3.1 to track newly synthesized histones
Quantitative cell cycle analysis:
Combine EdU labeling (for S-phase) with HIST1H3A staining and flow cytometry
Multi-parameter flow cytometry using cell cycle markers (cyclin B1, phospho-histone H3 (Ser10)) with HIST1H3A staining
ChIP-seq across cell cycle:
Perform H3.1-specific ChIP-seq at different cell cycle stages to map genome-wide incorporation patterns
Integrate with replication timing data to correlate H3.1 deposition with replication timing domains
Nascent chromatin capture:
Use pulse-chase approaches with biotin-dUTP followed by streptavidin pulldown to isolate newly replicated DNA and associated H3.1
Mass spectrometry time-course:
Perform quantitative proteomics across synchronized cell populations to track H3.1 abundance and modification changes through the cell cycle
For advanced analysis of HIST1H3A genomic distribution data, researchers should implement these computational approaches:
ChIP-seq analysis pipeline:
Quality control: FastQC for read quality assessment
Alignment: Bowtie2 or BWA with parameters optimized for histone ChIP
Peak calling: MACS2 with broad peak settings appropriate for histone distribution
Visualization: Generate bigWig files for browser visualization using deepTools
Differential binding analysis:
Use DiffBind or DESeq2 to identify regions with significant changes in H3.1 occupancy between conditions
Apply appropriate normalization methods (TMM, RLE) to account for library size differences
Integration with other genomic datasets:
Correlate H3.1 distribution with:
Replication timing data
Transcriptional activity (RNA-seq)
Other histone modifications
Chromatin accessibility (ATAC-seq, DNase-seq)
Motif analysis:
Use MEME, HOMER, or similar tools to identify DNA sequence motifs enriched in H3.1-occupied regions
Compare with transcription factor binding sites databases
Chromatin state segmentation:
Apply ChromHMM or similar algorithms to integrate H3.1 data with other histone marks to define chromatin states
Correlate chromatin states with functional genomic elements
3D chromatin structure integration:
Correlate H3.1 distribution with Hi-C or similar 3D genomic data to understand relationship with higher-order chromatin organization
Analyze distribution within topologically associating domains (TADs)
Visualization and reporting:
Generate comprehensive visualization using packages like EnrichedHeatmap, ComplexHeatmap in R
Use genome browsers (UCSC, IGV) with multiple tracks for integrated visualization
Apply dimensionality reduction techniques (PCA, t-SNE) to identify patterns across multiple datasets