Target: Acetylated lysine 79 on histone H3.1 (encoded by the HIST1H3A gene) .
Synonyms: H3/A, H3F3, H3FA, Histone H3.1 .
Species Reactivity: Primarily validated in Homo sapiens (human), with potential cross-reactivity in Canis familiaris (canine) based on immunogen design .
No cross-reactivity with acetylated H3K4, H3K9, H3K14, H3K18, H3K23, H3K27, H3K36, H3K56, or H3K122 .
Epitope recognition confirmed via peptide array and blocking experiments .
Transcriptional Activation: K79ac correlates with active chromatin regions, contrasting with K79 methylation (e.g., H3K79me3), which marks silent loci .
Cellular Response: Sodium butyrate treatment increases K79ac levels, suggesting a role in stress-induced chromatin remodeling .
Acetyl-HIST1H3A (K79) refers to the acetylation of lysine 79 on histone H3.1, a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries that require DNA as a template. Histones play a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability through their post-translational modifications . Acetylation at K79, similar to other histone acetylation marks, is believed to contribute to transcriptional activation by loosening chromatin structure, though its specific functions may differ from better-characterized sites like K14, which functions in transcriptional activation, chromatin accessibility, cellular identity, and epigenetic memory .
Acetyl-HIST1H3A (K79) antibodies are suitable for multiple experimental applications including:
Chromatin Immunoprecipitation (ChIP): For mapping genomic locations enriched with K79 acetylation
Western Blotting (WB): For detecting and quantifying total levels of K79 acetylation in protein extracts
Immunocytochemistry/Immunofluorescence (ICC/IF): For visualizing the nuclear distribution pattern of K79 acetylation
ELISA: For quantitative measurement of K79 acetylation levels
Peptide Array (PepArr): For testing antibody specificity against modified peptides
The working dilutions should be optimized for each application, starting with manufacturer recommendations such as 1:500-1:2000 for WB and 1:50-1:500 for ICC as reference points based on similar histone modification antibodies .
Validating antibody specificity is crucial for reliable experimental results. A systematic approach includes:
Peptide competition assays: Pre-incubating the antibody with acetylated K79 peptides should abolish signal, while incubation with unmodified or differently modified peptides should not affect signal .
Cross-reactivity testing: Using peptide arrays to confirm that the antibody does not cross-react with similar modifications (e.g., K79 methylation or acetylation at other lysine residues) .
Genetic validation: Using cells with mutations at K79 that prevent acetylation or with knockdown/knockout of relevant acetyltransferases.
Western blot analysis: Confirming a single band of appropriate molecular weight (~15-17 kDa for histone H3).
ChIP-seq with spike-in controls: Including exogenous chromatin with known K79 acetylation status as internal controls.
Based on standard practices for similar recombinant and polyclonal antibodies:
Storage temperature: Most histone modification antibodies should be stored at -20°C for long-term storage and at 4°C for short-term use .
Avoid freeze-thaw cycles: Aliquot antibodies upon receipt to minimize freeze-thaw cycles, which can degrade antibody quality.
Working dilution preparation: Dilute only the amount needed for immediate experiments in appropriate buffer containing a carrier protein (often BSA) to prevent adsorption to tubes.
Handling precautions: Follow non-reducing conditions for Western blot applications unless specifically recommended by the manufacturer.
The histone code hypothesis proposes that combinations of modifications create specific binding platforms for chromatin-associated proteins. For K79 acetylation:
Modification crosstalk: Similar to other histone acetylation marks, K79ac likely functions in concert with other modifications. The presence of K79 acetylation may influence the deposition or removal of nearby modifications, creating combinatorial patterns that dictate specific transcriptional outcomes.
Reader protein specificity: Different bromodomain-containing proteins may recognize K79ac in combination with other acetylation marks, leading to recruitment of specific transcriptional machinery.
Integration with methylation: K79 can also be methylated (mono-, di-, or tri-methylated) , suggesting a potential regulatory switch between acetylation and methylation at this residue similar to what occurs at other lysine residues like K4, K9, and K27.
Genome-wide distribution patterns: ChIP-seq analyses using K79ac antibodies can reveal how this modification co-localizes with other histone marks across the genome, providing insight into its functional roles in different chromatin contexts.
Several technical challenges are specific to studying K79 acetylation:
Antibody specificity: Due to the similar chemical nature of acetylation at different lysine residues, ensuring antibody specificity is crucial. Rigorous validation using peptide arrays and competition assays is necessary .
Modification abundance: If K79 acetylation is less abundant than other well-studied acetylation sites (like K9, K14, K27), more sensitive detection methods may be required.
Sample preparation: Optimal fixation and extraction protocols need to be established to preserve K79 acetylation while efficiently extracting chromatin-bound histones.
Epitope masking: Neighboring modifications or protein interactions might mask the K79ac epitope, leading to false-negative results in some contexts.
ChIP efficiency: Optimization of sonication conditions, antibody concentrations, and immunoprecipitation protocols specifically for K79ac ChIP experiments is necessary for high signal-to-noise ratios.
Effective ChIP-seq experimental design for K79ac includes:
Antibody selection: Choose antibodies validated specifically for ChIP applications , preferably with published ChIP-seq datasets demonstrating their performance.
Controls:
Input controls: Sequencing chromatin before immunoprecipitation
IgG controls: Using matched isotype IgG for non-specific binding assessment
Spike-in controls: Using exogenous chromatin from different species for normalization
Positive/negative genomic regions: Including primers for regions known to be enriched/depleted for K79ac
Chromatin preparation:
Optimal crosslinking time (typically 10-15 minutes with 1% formaldehyde)
Appropriate sonication to generate 200-500 bp fragments
Quality assessment of sheared chromatin
Sequencing depth: Aim for 20-40 million uniquely mapped reads per sample for histone modification ChIP-seq.
Bioinformatic analysis pipeline:
Quality control metrics specific for histone acetylation marks
Appropriate peak calling algorithms (broad vs. narrow peaks)
Integration with other genomic datasets
For rigorous quantitative comparisons:
Standardized sample processing: Process all samples simultaneously using identical protocols for histone extraction, antibody incubation, and detection methods.
Normalization strategies:
For Western blots: Normalize K79ac signal to total H3 levels
For ChIP-qPCR: Use percentage of input or normalization to invariant regions
For ChIP-seq: Implement spike-in normalization using exogenous chromatin
Technical replicates: Include multiple technical replicates to assess method variability.
Biological replicates: Analyze at least three independent biological replicates to account for biological variation.
Statistical analysis: Apply appropriate statistical tests based on experimental design and data distribution.
Dynamic range considerations: Ensure detection methods remain within linear dynamic range using standard curves if applicable.
When encountering weak or inconsistent signals:
Antibody concentration optimization: Titrate antibody concentrations to determine optimal working dilutions for each application .
Epitope retrieval methods: For fixed samples, test different antigen retrieval methods (heat-induced vs. enzymatic) to expose the K79ac epitope.
Blocking conditions: Optimize blocking reagents (BSA, non-fat milk, serum) and durations to reduce background while preserving specific signal.
Detection system sensitivity: Consider using more sensitive detection methods such as enhanced chemiluminescence for Western blots or amplification systems for immunofluorescence.
Sample preparation: Ensure histones are properly extracted and denatured; consider using histone extraction kits specifically designed to preserve acetylation marks.
Fresh antibody aliquots: Antibody activity may decrease with repeated freeze-thaw cycles; use fresh aliquots when possible.
When different antibody clones yield contradictory results:
Epitope differences: Different antibodies may recognize slightly different epitopes surrounding K79ac, leading to context-dependent detection differences. Map the exact epitope recognized by each antibody.
Clone comparison experiments: Perform side-by-side experiments using multiple antibody clones (monoclonal and polyclonal) on identical samples .
Orthogonal validation: Confirm results using non-antibody methods like mass spectrometry to directly detect K79ac.
Sequential ChIP: Perform sequential ChIP (re-ChIP) using different antibody clones to identify regions recognized by both antibodies.
Genetic validation: Use systems where K79 is mutated or where relevant acetyltransferases are depleted to confirm specificity.
Context specificity: Determine if contradictions are context-specific (e.g., cell type-dependent, locus-specific) by systematically varying experimental conditions.
For studying K79 acetylation dynamics, include:
Time-course controls: Sample collection at consistent time points across biological replicates.
Treatment controls:
Positive controls: HDAC inhibitors (like TSA or sodium butyrate) to increase global acetylation levels
Negative controls: Acetyltransferase inhibitors to reduce acetylation
Vehicle controls: For any treatments involving solvents
Cellular state markers: Monitor markers of cell cycle, differentiation status, or stress response relevant to your experimental context.
Other histone modifications: Track well-characterized histone modifications (H3K27ac, H3K9ac) as comparative controls.
Total histone levels: Monitor total H3 levels to account for potential changes in histone abundance.
Pathway validation: Include readouts of pathways known to influence histone acetylation (e.g., metabolic state indicators like Acetyl-CoA levels).
Multi-omics integration strategies include:
ChIP-seq integration with transcriptomic data: Correlate K79ac enrichment patterns with RNA-seq data to establish functional relationships with gene expression.
ATAC-seq or DNase-seq correlation: Determine relationship between K79ac and chromatin accessibility.
HiC or chromosome conformation capture techniques: Investigate potential roles of K79ac in three-dimensional chromatin organization.
Single-cell approaches: Apply single-cell versions of ChIP-seq, ATAC-seq, and RNA-seq to understand heterogeneity in K79ac distribution and its functional consequences.
Computational integration: Use machine learning approaches to integrate K79ac ChIP-seq with other histone modifications to predict functional genomic elements or expression patterns.
Mass spectrometry integration: Combine antibody-based detection with proteomic approaches to identify proteins associated with K79-acetylated histones.
Cutting-edge technologies for K79ac research include:
CUT&RUN and CUT&Tag: These techniques offer higher signal-to-noise ratios than traditional ChIP-seq and require fewer cells, enabling more sensitive detection of K79ac.
CRISPR-based epigenome editing: Using dCas9 fused to acetyltransferases to specifically introduce K79ac at defined genomic loci to test functional consequences.
Live-cell imaging of acetylation dynamics: Developing specific sensors for real-time visualization of K79ac fluctuations in living cells.
Single-molecule approaches: Applying techniques like single-molecule FRET to study how K79ac affects nucleosome stability and dynamics at the molecular level.
Long-read sequencing applications: Using long-read technologies to map K79ac across repetitive regions or to link distant modifications within the same chromatin fragment.
Spatial omics approaches: Developing methods to map K79ac distribution in a spatially resolved manner within tissue contexts.
To determine K79ac functions:
Site-specific mutagenesis: Generate K79R (non-acetylatable) or K79Q (acetylation-mimicking) mutations and assess phenotypic consequences.
Acetyltransferase/deacetylase identification: Perform enzymatic activity assays or protein interaction studies to identify enzymes that regulate K79 acetylation.
Reader protein identification: Use techniques like SILAC-MS with acetylated vs. non-acetylated K79 peptide pulldowns to identify proteins that specifically recognize K79ac.
Temporal dynamics studies: Track K79ac levels during processes like cell cycle progression, differentiation, or response to stimuli to identify potential regulatory roles.
Locus-specific manipulation: Use CRISPR-based approaches to target acetyltransferases or deacetylases to specific loci to determine local effects of K79ac modulation.
Disease model studies: Examine K79ac alterations in disease states to uncover potential pathological roles.
For optimal K79ac detection:
Cell/tissue fixation: Use freshly prepared 1% formaldehyde for 10-15 minutes at room temperature, followed by quenching with glycine.
Histone extraction protocol selection:
Acid extraction: Particularly effective for preserving histone modifications
Triton extraction: Useful for nuclear fraction enrichment
Commercial histone extraction kits optimized for preserving acetylation marks
Protease and HDAC inhibitors: Include comprehensive protease inhibitor cocktails and HDAC inhibitors (sodium butyrate, TSA) during extraction to prevent degradation and deacetylation.
Gentle handling: Minimize mechanical stress and processing time to preserve labile modifications.
Storage considerations: Store extracted histones at -80°C in small aliquots to avoid repeated freeze-thaw cycles.
For K79ac ChIP-qPCR optimization:
Antibody amount titration: Test multiple antibody concentrations (2-10 μg per ChIP reaction) to determine optimal signal-to-noise ratio.
Chromatin amount optimization: Standardize input chromatin quantity (typically 20-50 μg per reaction).
Primer design considerations:
Design primers for regions expected to be enriched for K79ac (based on literature or hypothesis)
Include primers for negative regions (heterochromatic regions)
Include primers for positive control regions (known acetylation-rich promoters)
Sonication optimization: Aim for chromatin fragments of 200-500 bp, verified by gel electrophoresis.
Washing stringency: Optimize salt concentration in wash buffers to reduce background while maintaining specific signal.
Data normalization: Calculate percent input or fold enrichment over IgG control for accurate quantification.
For accurate K79ac quantification:
ELISA-based methods: Commercial ELISA kits can provide quantitative measurement with detection ranges as low as 23.5 pg/mL and sensitivity of 5.8 pg/mL .
Western blotting with standard curves: Include a dilution series of recombinant acetylated histones to create standard curves for quantification.
Mass spectrometry approaches:
Targeted MS using isotope-labeled internal standards
Multiple reaction monitoring (MRM) for sensitive and specific quantification
SILAC labeling for relative quantification between conditions
ChIP-qPCR absolute quantification: Use spike-in controls of known concentration for absolute quantification of enrichment.
Flow cytometry: For single-cell quantification of global K79ac levels using permeabilized cells and fluorescently-labeled antibodies.
Image analysis quantification: For immunofluorescence data, use appropriate image analysis software with background subtraction and internal calibration standards.