While direct data on HIST1H3A (Ab-115) is sparse, related histone H3.1 antibodies (e.g., CSB-RA010418A0HU , AP61415 ) are validated for:
Western Blot (WB): Detection of histone H3.1 in cell lysates.
Immunohistochemistry (IHC): Localization of nuclear histones in tissue sections.
Immunofluorescence (IF): Visualizing chromatin structure in fixed cells.
Histone H3.1 antibodies, including those targeting HIST1H3A, are critical for studying:
Chromatin Remodeling: H3.1 is involved in DNA replication and repair, as it integrates into nucleosomes during S-phase .
Epigenetic Regulation: Acetylation or methylation at specific residues (e.g., K115 , K18 ) modulates gene expression.
Heterogeneity in Single Cells: H3.1 antibodies reveal variability in histone incorporation patterns across cell populations .
Modification-Specific Recognition: Antibodies like AP61415 (AcK115) distinguish acetylated H3.1, enabling studies on transcriptional activation .
For context, other histone H3.1 antibodies include:
HIST1H3A (Ab-115) antibody is a polyclonal antibody raised in rabbits against a specific peptide sequence around the lysine 115 residue derived from human Histone H3.1 protein. This antibody recognizes the HIST1H3A protein, also known as Histone H3.1, which is a core component of nucleosomes. Histone H3.1 plays a fundamental role in shaping the epigenetic landscape of the genome, influencing gene expression, and maintaining genomic integrity through its dynamic interactions with DNA and various proteins . The antibody targets a specific region that allows it to distinguish H3.1 from other histone variants, making it valuable for investigating histone variant distribution and function in chromatin research .
The HIST1H3A (Ab-115) antibody has been validated for several experimental applications with specific recommended dilutions to achieve optimal results:
| Application | Recommended Dilution |
|---|---|
| Western Blot (WB) | 1:100-1:1000 or 1:500-1:5000 |
| Immunohistochemistry (IHC) | 1:50-1:500 |
| Immunofluorescence (IF) | 1:30-1:200 |
| ELISA | As per manufacturer protocol |
These dilution ranges provide starting points for optimization. Researchers should perform titration experiments to determine the optimal concentration for their specific experimental conditions, sample types, and detection methods. Proper validation is essential when applying the antibody to new experimental systems or cell types .
The production of HIST1H3A antibody involves a sophisticated multi-step process:
Initial immunization of rabbits with a synthesized peptide derived from human HIST1H3A protein (specifically containing the region around lysine 115).
Extraction of genes encoding the HIST1H3A antibody from the immunized rabbits.
Integration of these antibody genes into specialized expression vectors.
Introduction of modified vectors into host suspension cells.
Cultivation of the cells to stimulate antibody expression and secretion.
Purification of the antibody using affinity chromatography techniques, which effectively isolate the antibody from the cell culture supernatant .
The final product is antigen-affinity purified to ensure high specificity and minimal cross-reactivity with other proteins. This rigorous purification process is critical for experimental applications requiring high specificity, such as detecting subtle differences between histone variants .
The heterogeneous immunofluorescence (IF) staining pattern observed with H3.1 antibodies among single cells can be attributed to several biological and technical factors:
Cell cycle-dependent incorporation: Histone H3.1 is primarily incorporated during DNA replication in S phase. Cells at different stages of the cell cycle will show varying levels and distributions of newly incorporated H3.1, resulting in heterogeneous staining patterns .
Chromatin state variations: Cells may have different chromatin condensation states depending on their transcriptional activity, which affects antibody accessibility to histone epitopes.
Post-translational modifications (PTMs): H3.1 can undergo various PTMs that might mask the epitope recognized by the antibody. The pattern of these modifications can vary between cells and chromatin regions .
Replication timing: Different genomic regions replicate at different times during S phase, resulting in asynchronous incorporation of H3.1 throughout the genome.
Technical considerations: Fixation methods, permeabilization conditions, and antibody concentration can all influence the observed staining pattern .
Researchers investigating this heterogeneity should consider synchronizing cells, performing co-staining with cell cycle markers, and comparing patterns with other histone variant antibodies to distinguish biological variation from technical artifacts .
The ability of HIST1H3A (Ab-115) antibody to distinguish between newly incorporated and existing histone H3.1 depends on several factors:
Recent findings indicate that newly incorporated H3.1 may have distinct post-translational modification patterns compared to pre-existing H3.1, particularly during DNA replication and repair processes. These differences in modification states could potentially affect epitope accessibility and antibody recognition .
For precise tracking of newly incorporated histones, researchers should consider:
Combining antibody detection with SNAP-tag or other labeling technologies for pulse-chase experiments
Using dual immunofluorescence with antibodies against specific replication-associated PTMs
Correlating H3.1 staining patterns with replication markers such as PCNA or EdU
Employing chromatin immunoprecipitation combined with nascent DNA capture methods
This approach provides more comprehensive insights into histone deposition dynamics during chromatin assembly and remodeling processes .
The structural differences between H3.1 and other H3 variants have significant implications for antibody specificity and experimental design:
H3.1 and H3.2 differ by only a single amino acid substitution (S96C), where H3.1 contains cysteine at position 96 while H3.2 has serine. This subtle difference creates distinct molecular properties that affect antibody recognition. The cysteine residue in H3.1 is located within a hydrophobic pocket encompassing F67, A95, and L100 in H3 and L58, F61, and L62 in helix 2 of H4. This substitution enhances the stability of this hydrophobic cage and subsequently leads to the stabilization of the H3/H4 dimer .
More pronounced differences exist between canonical H3.1 and the testis-specific H3.1T variant, which differs by four substitutions (A24V, V71M, A98S, and A111V). These modifications confer distinct properties to H3.1T, including in vitro and in vivo instability, weaker association with H2A/H2B dimers, defective incorporation into nucleosomes by Nap1, and more rapid exchange in nucleosomes of living cells .
When using HIST1H3A (Ab-115) antibody for variant-specific detection:
Verify epitope conservation across variants of interest
Perform control experiments with cells/tissues known to express specific variants
Consider potential cross-reactivity with highly similar variants
Use complementary techniques like mass spectrometry to validate findings
Be aware that post-translational modifications near the epitope region may affect recognition
These considerations are particularly important when studying specialized cell types or developmental processes where multiple histone variants may be expressed simultaneously.
When using HIST1H3A (Ab-115) antibody in chromatin immunoprecipitation (ChIP) experiments, researchers may encounter several technical challenges:
Cross-reactivity with other histone variants: Due to the high sequence similarity between H3.1 and other H3 variants (particularly H3.2 which differs by only one amino acid), ensuring specificity can be challenging. The S96C substitution that distinguishes H3.1 from H3.2 affects molecular properties, making H3.1 distinguishable from H3.2 in HPLC elution profiles, but this subtle difference may still result in cross-reactivity in ChIP experiments .
Epitope masking by post-translational modifications: The region around lysine 115 may be subject to post-translational modifications that could mask the epitope and reduce antibody binding efficiency. Researchers should be aware that the chromatin state and modification profiles of their experimental system might affect antibody recognition .
Fixation conditions: Over-fixation can mask epitopes while under-fixation may not preserve protein-DNA interactions adequately. Optimization of formaldehyde concentration and fixation time is essential.
Chromatin fragmentation: Excessive or insufficient sonication can affect epitope accessibility and ChIP efficiency. Standardizing fragmentation conditions is critical for reproducible results.
Antibody concentration: Insufficient antibody leads to poor enrichment while excess antibody may increase non-specific binding. Titration experiments should be performed to determine optimal concentration .
To address these issues, researchers should include appropriate controls (such as IgG control, input control, and positive control regions), optimize fixation and sonication conditions, and validate results with alternative methods or antibodies targeting different epitopes of H3.1.
Validating the specificity of HIST1H3A (Ab-115) antibody requires a multi-faceted approach:
These validation steps should be documented and included in publications to support the reliability of experimental findings .
HIST1H3A (Ab-115) antibody provides a valuable tool for investigating the role of histone H3.1 variants in cancer development and progression:
Expression profiling: The antibody can be used to assess H3.1 expression levels across different cancer types and stages. Research has shown that H3.1 is differentially regulated in various cancers, including HER2-positive breast cancer and is negatively regulated by ERβ1. It has also been found to be highly expressed at the cancer stem-like stage and overexpressed in aromatase inhibitor-resistant estrogen receptor-positive (ER+) breast cancer .
Chromatin immunoprecipitation sequencing (ChIP-seq): This technique can map genome-wide distribution of H3.1 in cancer cells versus normal cells, identifying regions where altered H3.1 deposition may contribute to dysregulated gene expression. The differential distribution of H3.1 across genomic regions may reveal insights into cancer-specific chromatin states .
Correlation with cancer phenotypes: Immunohistochemistry with the antibody can be used to correlate H3.1 levels or nuclear distribution patterns with clinical parameters like tumor grade, treatment response, and patient outcomes. According to the data, H3.1 expression patterns can distinguish between different gastric cancer grades and normal tissue .
Epigenetic therapy monitoring: The antibody can track changes in H3.1 distribution following epigenetic therapies, as H3.1 has been found to be altered in response to epigenetic therapy in certain cancers .
Cancer stem cell studies: Since H3.1 is highly expressed at the cancer stem-like stage, the antibody can help identify and characterize cancer stem cell populations in tumor samples .
To effectively use this antibody in cancer research, investigators should consider combining it with antibodies against cancer-specific markers and employing multiparametric analysis techniques to correlate H3.1 patterns with specific cancer phenotypes or molecular subtypes .
Investigating the relationship between histone H3.1 modifications and cancer-related phenotypes requires sophisticated methodological approaches:
Sequential ChIP (ChIP-reChIP): This technique can determine the co-occurrence of H3.1 and specific post-translational modifications on the same nucleosomes. By first immunoprecipitating with HIST1H3A (Ab-115) antibody and then with antibodies against specific modifications (or vice versa), researchers can identify genomic regions where modified H3.1 is present .
Mass spectrometry-based proteomics: Using HIST1H3A antibody for immunoprecipitation followed by mass spectrometry analysis allows for comprehensive characterization of post-translational modifications specific to H3.1 in cancer cells versus normal cells. This approach can identify cancer-specific modification patterns on H3.1 .
Single-cell immunofluorescence analysis: Combining HIST1H3A (Ab-115) antibody with antibodies against specific histone modifications in multiplexed immunofluorescence can reveal heterogeneity in H3.1 modification patterns at the single-cell level within tumors. This is particularly important given that H3.1 shows heterogeneous staining patterns among single cells .
CRISPR-Cas9 modification of H3.1 genes: Creating cells with mutations at specific modification sites in H3.1-encoding genes can help establish causal relationships between particular modifications and cancer phenotypes .
Integrated genomics approach: Correlating ChIP-seq data (using HIST1H3A antibody) with RNA-seq, ATAC-seq, and clinical data can provide insights into how H3.1 distribution and modifications affect gene expression programs relevant to cancer progression .
Drug response studies: Using the antibody to track changes in H3.1 modification patterns following treatment with epigenetic modulators can identify potential therapeutic targets. For example, H3.1 has been found to be downregulated in chronic myelogenous leukemia stem cells and altered in response to epigenetic therapy in certain cancers .
These approaches should be complemented with functional assays (proliferation, migration, invasion, etc.) to establish mechanistic links between specific H3.1 modifications and cancer-related phenotypes .
The HIST1H3A (Ab-115) antibody offers significant potential for elucidating the dynamic roles of histone H3.1 variants during cellular reprogramming and differentiation processes:
Temporal profiling during differentiation: By using the antibody at different time points during cell differentiation, researchers can track changes in H3.1 deposition patterns that correlate with cell fate decisions. This approach can reveal how H3.1 incorporation changes as cells transition between pluripotent and differentiated states .
Genome-wide mapping during reprogramming: ChIP-seq using HIST1H3A antibody during induced pluripotent stem cell (iPSC) generation can identify genomic regions where H3.1 deposition changes precede or follow transcriptional changes associated with pluripotency acquisition.
Lineage-specific patterns: The antibody can help identify tissue-specific patterns of H3.1 distribution that may be essential for maintaining cell identity. For instance, comparing H3.1 patterns in different cell lineages derived from the same stem cell population can highlight regions critical for lineage specification .
Co-occupancy studies: Combining ChIP-seq data for H3.1 (using HIST1H3A antibody) with data for lineage-specific transcription factors can identify regulatory elements where H3.1 and these factors interact to control gene expression during differentiation.
Comparison with variant-specific patterns: Comparing the distribution of H3.1 with other histone variants (such as H3.3, which is incorporated independently of replication) during differentiation can reveal how variant-switching contributes to chromatin remodeling during cell fate changes .
Modification state transitions: Investigating how the post-translational modification landscape of H3.1 evolves during differentiation can provide insights into epigenetic mechanisms governing cell fate decisions. The S96C substitution that distinguishes H3.1 from H3.2 may affect modification patterns and protein interactions relevant to differentiation .
These approaches can significantly advance our understanding of the epigenetic basis of development, regeneration, and disease, particularly in contexts where aberrant differentiation contributes to pathological states .
Emerging technologies are enabling increasingly sophisticated analyses of histone H3.1 dynamics using antibodies like HIST1H3A (Ab-115):
CUT&RUN and CUT&Tag: These techniques offer higher signal-to-noise ratios than traditional ChIP-seq and require fewer cells, making them valuable for analyzing rare cell populations or clinical samples. Using HIST1H3A antibody with these methods can provide higher-resolution maps of H3.1 distribution .
Single-cell ChIP-seq: Applying HIST1H3A antibody in single-cell ChIP-seq protocols can reveal cell-to-cell variation in H3.1 distribution patterns within heterogeneous populations, offering insights into how chromatin states vary at the individual cell level. This is particularly relevant given the observed heterogeneous staining patterns of H3.1 antibodies among single cells .
Live-cell imaging of H3.1 dynamics: Combining HIST1H3A antibody with cell-penetrating peptide technology or developing Fab fragments allows for tracking H3.1 in living cells, revealing real-time dynamics during processes like DNA replication and repair.
Proximity ligation assays (PLA): Using HIST1H3A antibody in conjunction with antibodies against chromatin-modifying enzymes or transcription factors in PLA can identify specific protein-protein interactions involving H3.1 in situ.
CRISPR-Cas9 epitope tagging of endogenous H3.1: As mentioned in the literature, CRISPR-Cas9-mediated gene editing techniques allow for epitope tagging of histone isoforms in their endogenous loci, permitting ChIP-seq analysis to determine whether histone isoforms display unique patterns of genomic localization .
Super-resolution microscopy: Applying advanced microscopy techniques with HIST1H3A antibody can visualize the spatial organization of H3.1-containing nucleosomes at nanoscale resolution, potentially revealing domain-specific patterns not detectable with conventional microscopy.
Mass cytometry (CyTOF): Combining metal-conjugated HIST1H3A antibody with antibodies against other histone marks and cellular proteins in CyTOF analysis allows for high-dimensional profiling of histone variant patterns across cell populations.
These cutting-edge approaches offer unprecedented opportunities to understand how H3.1 dynamics contribute to normal development and disease processes, potentially leading to novel diagnostic markers or therapeutic targets .