Target Protein: HIST1H3A (UniProt ID: P68431), a core component of nucleosomes that plays a critical role in chromatin structure and gene regulation .
Epitope: Peptide sequence around Ser10 of human histone H3.1 .
Immunogen: Synthetic peptide derived from human histone H3.1, ensuring specificity for the unmodified or post-translationally modified Ser10 region .
| Immunogen Details | Value |
|---|---|
| Species | Human |
| Modification Site | Serine 10 (Ser10) |
| Sequence Origin | Histone H3.1 (HIST1H3A) |
Validated applications include:
ChIP Validation: Demonstrated specificity in chromatin immunoprecipitation assays using Hela cells, with robust enrichment compared to control IgG .
Cancer Research: Successfully detected histone H3.1 in paraffin-embedded human cancer tissues, including colon, lung, and prostate carcinomas .
Cross-Reactivity: No reported cross-reactivity with non-histone proteins, though optimal performance requires validation in user-specific conditions .
Specificity: Confirmed via peptide competition assays and reactivity across human, mouse, and rat samples .
Western Blot: Detects a single band at ~17 kDa corresponding to histone H3 in acid-extracted cell lysates .
IF/ICC: Localizes to the nucleus in Hela cells, consistent with histone function .
Exclusively for Research: Not approved for diagnostic, therapeutic, or clinical use .
Batch Variability: As with all polyclonal antibodies, performance may vary between lots; independent validation is recommended .
Antibody Validation: Peer-reviewed studies emphasize the importance of rigorous validation for histone antibodies. The HIST1H3A (Ab-10) Antibody aligns with ENCODE standards for specificity in ChIP and WB .
Contextual Caveats: While some histone modification antibodies exhibit cross-reactivity (e.g., H3K27me3 vs. H3K4me3) , HIST1H3A (Ab-10) targets unmodified H3.1, reducing such risks .
HIST1H3A encodes Histone H3.1, one of the core histone proteins involved in chromatin structure in eukaryotic cells. Nucleosomes, which consist of DNA wrapped around histone octamers, compact DNA into chromatin, limiting DNA accessibility to cellular machinery. Histone H3.1 plays a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability . Unlike the constitutively expressed H3.3, H3.1 is primarily expressed during the S-phase of the cell cycle, making it particularly important during DNA replication .
In humans, there are several histone H3 variants, each with distinct expression patterns and functions. H3.1 (encoded by HIST1H3A and other genes) and H3.2 are expressed primarily during S-phase and are incorporated into chromatin during DNA replication. In contrast, H3.3 is expressed constitutively throughout the cell cycle . Some H3 variants have tissue-restricted expression patterns (H3.5, H3.X, H3.Y), while others like CENP-A are deposited only at centromeres . These differences in expression timing and patterns contribute to their specialized functions in chromatin regulation.
Histone H3.1 undergoes extensive post-translational modifications, particularly on its N-terminal tail that protrudes from the globular nucleosome core . These modifications include:
Acetylation (at various lysine residues including K23, K27)
Methylation (at lysine residues including K4, K27, K36)
Phosphorylation (at serine residues including S10)
Other modifications including ubiquitination and SUMOylation
These modifications constitute part of the "histone code" that regulates DNA accessibility and recruits specific protein complexes to chromatin .
Recent research has identified histone H3.1 as a chromatin-embedded redox sensor. According to a 2024 study, H3.1 can sense changes in hydrogen peroxide levels within the nucleus, with Cysteine 96 (Cys96) playing a crucial role in this sensing mechanism . When oxidized, H3.1 undergoes changes that lead to its replacement, potentially with H3.3, resulting in chromatin decompaction and transcriptional activation . This function connects cellular redox state directly to chromatin structure and gene expression regulation.
Researchers can choose from several types of HIST1H3A antibodies:
Polyclonal antibodies: Derived from multiple B cell lineages, recognizing multiple epitopes on H3.1
Monoclonal antibodies: Derived from a single B cell lineage, recognizing a single epitope (e.g., clones 27F2, 6F6)
Modification-specific antibodies: Recognizing specific post-translational modifications on H3.1, such as acetylation at Lys23 (acLys23), acetylation at Lys27 (acLys27), di-methylation at Lys4 (H3K4me2), di-methylation at Lys27 (H3K27me2), and tri-methylation at various lysine residues
Antibody validation is crucial for reliable research results. Consider these approaches:
Western blot analysis with positive and negative controls to confirm band size (approximately 15.4 kDa for Histone H3)
Peptide competition assays to verify epitope specificity
Knockout/knockdown experiments to confirm antibody specificity
Cross-reactivity tests against other histone variants, particularly H3.2 and H3.3, which have high sequence homology with H3.1
Dot blot or ELISA testing with modified and unmodified peptides for modification-specific antibodies
When selecting a HIST1H3A antibody, consider:
The specific post-translational modification you're studying (if applicable)
Application compatibility - check if the antibody has been validated for your application (Western blot, ChIP, immunofluorescence, etc.)
Species reactivity - ensure the antibody recognizes HIST1H3A in your experimental organism
Clonality - polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variation; monoclonal antibodies provide consistent specificity but may be more sensitive to epitope masking
Host species - consider secondary antibody compatibility and potential cross-reactivity issues in your experimental system
For successful Western blotting with HIST1H3A antibodies:
Sample preparation: Use specialized histone extraction protocols to efficiently isolate histones from nuclear fractions
Loading control: Consider total H3 or other stable nuclear proteins as loading controls
Expected size: Look for a band at approximately 15.4 kDa corresponding to Histone H3
Dilution: Typical working dilutions range from 1:5000 to 1:10000, but optimize for your specific antibody
Blocking: Use 5% non-fat dry milk or BSA in TBST, depending on the antibody specifications
Membrane choice: PVDF membranes are generally preferred for histone detection
Transfer conditions: Use low methanol concentrations and adjust transfer time to prevent small proteins like histones from transferring through the membrane
For Chromatin Immunoprecipitation (ChIP) applications:
Crosslinking: Standard formaldehyde crosslinking (1% for 10 minutes) works for most histone ChIP experiments
Sonication: Optimize sonication conditions to generate chromatin fragments of 200-500 bp
Antibody amount: Typically use 2-5 μg of antibody per ChIP reaction
Positive controls: Include primers for regions known to be enriched for your specific histone modification
Negative controls: Include IgG antibody controls and primers for regions not expected to have your modification
Wash stringency: Adjust wash buffers based on antibody specificity and background levels
Validation: Confirm enrichment through qPCR before proceeding to sequencing
For optimal immunofluorescence results:
Fixation: 4% paraformaldehyde is commonly used; avoid methanol fixation which may affect some histone epitopes
Permeabilization: Use 0.1-0.5% Triton X-100 to access nuclear antigens
Antigen retrieval: May be necessary for some fixed tissues; citrate buffer (pH 6.0) is often effective
Blocking: 5% normal serum from the same species as the secondary antibody
Antibody dilution: Start with 1:500 dilution and optimize as needed
Controls: Include no primary antibody controls and, if possible, cells with known H3.1 modification states
Counterstaining: DAPI for nuclear visualization to confirm nuclear localization of H3.1 signal
To study chromatin dynamics during the cell cycle:
Synchronize cells using methods appropriate for your cell type (double thymidine block, serum starvation/refeeding, etc.)
Collect cells at different cell cycle stages (confirmed by flow cytometry or cyclin expression)
Perform ChIP-seq or immunofluorescence with HIST1H3A antibodies to track H3.1 incorporation into chromatin
Compare with replication timing data to correlate H3.1 deposition with DNA synthesis
Use pulse-chase experiments with tagged histones to differentiate new deposition from existing H3.1
Consider dual immunofluorescence with cell cycle markers (e.g., PCNA for S-phase) to correlate H3.1 patterns with cell cycle stages
To investigate relationships between H3.1 modifications and gene expression:
Perform ChIP-seq with modification-specific H3.1 antibodies (e.g., H3K27me3, H3K4me3, acLys23)
Integrate with RNA-seq data from the same cell type/condition
Use sequential ChIP (re-ChIP) to identify genomic regions with co-occurring modifications
Compare ChIP-seq profiles before and after perturbations (e.g., inhibitors of histone-modifying enzymes)
Correlate with chromatin accessibility data (ATAC-seq, DNase-seq)
Perform reporter assays with mutated histone H3.1 to confirm functional relationships
Utilize CUT&RUN or CUT&Tag as alternative approaches for higher signal-to-noise ratio in mapping histone modifications
Based on recent findings about H3.1's role as a redox sensor , researchers can:
Utilize oxidation-resistant H3.1 mutants (e.g., H3.1(C96S)) to study the impact of redox sensing on chromatin structure
Use DCP-Bio1 or similar reagents to label oxidized Cys-SOH residues in H3.1 following oxidative stress
Perform ChIP-seq before and after oxidative stress to map changes in H3.1 occupancy
Compare with H3.3 ChIP-seq to assess histone variant exchange during redox responses
Combine with transmission electron microscopy to visualize changes in chromatin compaction
Utilize fluorescence microscopy with nuclear size measurements to quantify chromatin decompaction
Perform RNA-seq to correlate transcriptional changes with histone oxidation and replacement
To study cancer-associated histone H3 mutations:
Use mutation-specific antibodies (if available) or tagged mutant histones to track their incorporation
Perform ChIP-seq to map genome-wide distribution of mutant histones
Compare chromatin accessibility (ATAC-seq) in cells with wild-type versus mutant H3.1
Investigate changes in other histone modifications that may be affected by H3 mutations
Study protein interactions using immunoprecipitation followed by mass spectrometry
Assess impact on gene expression using RNA-seq
Correlate findings with cancer patient data to establish clinical relevance
To investigate H3.1 turnover and dynamics:
SNAP-tag or CLIP-tag labeling of H3.1 for pulse-chase experiments
FRAP (Fluorescence Recovery After Photobleaching) with fluorescently tagged H3.1
Stable isotope labeling with amino acids in cell culture (SILAC) combined with mass spectrometry
Inducible expression systems for temporal control of tagged H3.1 variants
ChIP-seq time courses following perturbations that affect histone deposition
Single-molecule tracking of tagged histones to measure diffusion rates and binding dynamics
Targeted degradation approaches (e.g., auxin-inducible degron system) to study rapid H3.1 depletion effects
Common challenges and solutions include:
Cross-reactivity with other H3 variants: Use highly specific antibodies or epitope tags; validate with peptide arrays
High background in Western blots: Increase blocking time/concentration; optimize antibody dilution; use more stringent washes
Poor signal in ChIP: Optimize chromatin fragmentation; increase antibody amount; ensure epitope is not masked by other modifications
Inconsistent results across experiments: Use lot-controlled antibodies; standardize protocols; include positive controls
Non-specific bands in Western blots: Use recombinant H3.1 as positive control; perform peptide competition assays
Epitope masking by adjacent modifications: Consider using alternative antibodies targeting different regions of H3.1
Poor reproducibility in ChIP-seq: Implement rigorous quality control; use spike-in controls; standardize bioinformatic pipelines
For integrative analysis:
Start with Western blotting to confirm antibody specificity and H3.1 modification levels
Perform immunofluorescence to determine cellular and subcellular localization patterns
Use ChIP-seq to map genome-wide distribution of H3.1 and its modifications
Integrate with gene expression data (RNA-seq) to correlate chromatin states with transcriptional output
Add chromatin accessibility data (ATAC-seq, DNase-seq) to understand functional consequences
Validate key findings with orthogonal approaches (e.g., CUT&RUN/CUT&Tag to complement ChIP)
Use computational approaches to integrate multiple datasets and identify statistically significant patterns
Essential controls include:
Input controls for ChIP experiments to normalize for DNA abundance
IgG controls from the same species as the primary antibody to assess non-specific binding
Peptide competition controls to confirm antibody specificity
Positive controls: genomic regions known to be enriched for your histone modification
Negative controls: regions not expected to have your modification
Technical replicates to assess experimental variability
Biological replicates to account for natural biological variation
Treatment controls (e.g., histone modification enzyme inhibitors) to validate antibody specificity