The antibody is validated for diverse experimental approaches:
Sample Types: HeLa, MCF-7, HepG2, 293, NIH/3T3 whole-cell lysates .
Results: Detects a band at ~12 kDa, consistent with histone H4’s molecular weight .
Tissue Types: Human glioma, pancreatic cancer, colon, and rat colon .
Protocol: Antigen retrieval with citrate buffer (pH 6.0), blocking with goat serum, and detection via HRP-conjugated secondary antibodies .
Validation: Demonstrated enrichment of H4K12ac in euchromatin regions, as shown in ChIP-seq studies .
Potential Cross-Reactivity: HIST1H4A shares sequence homology with other histone H4 isoforms (e.g., HIST1H4B, HIST1H4C), which may result in off-target binding .
Controls: Rabbit IgG is used as a negative control to confirm specificity .
H4K12 acetylation (H4K12ac) is linked to:
Chromatin Assembly: Newly synthesized histone H4 is diacetylated at K5 and K12, facilitating nuclear import and chromatin integration .
Gene Regulation: H4K12ac is enriched at transcription start sites (TSSs), correlating with active transcription .
DNA Repair: H4K12ac may play a role in DNA damage response, though direct evidence requires further study .
A panel of monoclonal antibodies targeting H4 acetylation (K5, K8, K12, K16) and methylation (K20) revealed distinct patterns:
H4K12ac: Detected in newly assembled chromatin and associated with euchromatic regions .
H4K8ac/K16ac: Enriched near TSSs, suggesting roles in transcription initiation .
| Application | Dilution |
|---|---|
| WB | 1:500–1:2000 |
| IHC | 1:20–1:200 |
| IP | 1:200–1:2000 |
| ChIP | 5 µg per IP reaction |
HIST1H4A (histone cluster 1, H4a) is a 103 amino acid protein belonging to the histone H4 family and serves as a core component of nucleosomes. As part of the nucleosome structure, histone H4 plays a central role in chromatin organization, thereby influencing transcription regulation, DNA repair, DNA replication, and chromosomal stability . Acetylation at lysine 12 of histone H4 (H4K12ac) is a critical post-translational modification that contributes to chromatin decompaction during DNA replication, making it essential for proper gene expression and genomic stability . This specific modification is evolutionarily conserved from yeast to humans, highlighting its fundamental importance in chromatin biology . H4K12ac is particularly significant in research investigating chromatin accessibility, gene transcription regulation, and epigenetic inheritance during cell division.
The HIST1H4A (Ab-12) Antibody, which specifically targets acetylated lysine 12 on histone H4, can be utilized in multiple experimental applications:
These applications allow researchers to investigate the presence, abundance, and genomic distribution of H4K12 acetylation across different experimental contexts .
For optimal performance and longevity of the HIST1H4A (Ab-12) Antibody, proper storage and handling are crucial. The antibody should be stored at -80°C in its recommended buffer, which is typically PBS for many commercial preparations . When working with the antibody, minimize freeze-thaw cycles as they can lead to protein denaturation and reduced activity. Always centrifuge the antibody briefly after thawing to collect the solution at the bottom of the tube before opening. When diluting the antibody for experiments, use fresh, sterile buffers, and consider adding protease inhibitors to prevent degradation. For long-term storage of diluted antibody, adding stabilizing proteins such as BSA (0.1-1%) and preservatives like sodium azide (0.02%) can help maintain activity, though ensure these additives won't interfere with your specific application.
Validating antibody specificity is critical for generating reliable research data. For HIST1H4A (Ab-12) Antibody, consider implementing these validation strategies:
Peptide Competition Assay: Pre-incubate the antibody with increasing concentrations of H4K12ac peptides before application in your experiment. Specific signal should decrease proportionally with increasing peptide concentration .
Knockout/Knockdown Controls: Utilize cells with reduced HAT1 expression (the enzyme responsible for H4K12 acetylation) as a negative control, which should show decreased antibody signal .
Peptide Microarray Analysis: Test antibody against a panel of modified and unmodified histone peptides to assess cross-reactivity with other H4 modifications and neighboring PTMs .
Multiple Antibody Comparison: Employ at least two different H4K12ac antibodies from different vendors or clones in parallel experiments to confirm consistent results .
Mass Spectrometry Correlation: If possible, correlate antibody-based detection with mass spectrometry analysis of histone modifications to confirm specificity .
Research has demonstrated that some H4K12ac antibodies may cross-react with other acetylation sites on H4, particularly when multiple acetylation marks are present, showing enhanced signal on peptides containing H4K12ac alongside other H4 acetylation sites . Therefore, rigorous validation is essential for accurate interpretation of experimental data.
Several factors can influence the binding specificity of HIST1H4A (Ab-12) Antibody, potentially leading to misinterpretation of experimental results:
Adjacent Post-Translational Modifications: Neighboring modifications on histone H4 can significantly impact antibody recognition. Research has shown that H4 acetyl antibodies often display enhanced binding to epitopes with increased acetylation content at multiple sites . For example, the presence of acetylation at K5, K8, or K16 on the same H4 tail may enhance or interfere with the detection of K12ac.
Modification State Specificity: Some antibodies may not accurately distinguish between different modification states (e.g., mono-, di-, or tri-methylation) or may cross-react with similar modifications on different lysine residues .
Fixation Methods: In immunostaining applications, different fixation protocols can alter epitope accessibility or structure, affecting antibody binding.
Sample Preparation: Harsh extraction methods may disrupt the native structure of histone proteins, potentially exposing or concealing the H4K12ac epitope.
Antibody Concentration: Using too high or too low antibody concentrations can lead to non-specific binding or insufficient signal, respectively.
Studies using peptide microarrays have identified that many commercial H4 acetyl antibodies show preferential binding to epitopes with iterative increases in acetylation content, rather than binding exclusively to their target modification . This highlights the importance of thorough validation and careful experimental design.
The acetylation pattern at H4K12 shows interesting differences across histone complexes, particularly between canonical H3.1-H4 and variant H3.3-H4 tetramers:
Preferential Acetylation: HAT1-RbAp46 (the enzyme complex responsible for H4K5,12 acetylation) acetylates H4 in H3.1-H4 complexes more efficiently than H4 in H3.3-H4 complexes . This differential acetylation may contribute to the distinct functional roles of these histone variants.
Chaperone Association: The HAT1 holoenzyme and associated proteins show stronger association with H3.1 than with H3.3, which correlates with increased H4K5,12ac on H4 that co-purifies with H3.1 .
Functional Consequences: This differential acetylation pattern impacts nucleosome assembly of H3.1 and H3.3 differently, potentially contributing to their distinct genomic distributions and functions .
These findings suggest that H4K12ac may serve as a modification that helps distinguish between replication-dependent nucleosome assembly (primarily H3.1-containing) and replication-independent processes (primarily H3.3-containing), making it an important modification to study in the context of chromatin dynamics and cell cycle progression.
Optimizing ChIP-seq experiments with HIST1H4A (Ab-12) Antibody requires careful consideration of several parameters to ensure high-quality, reproducible data:
Antibody Validation for ChIP: Before proceeding with genome-wide analyses, validate the antibody specifically for ChIP applications using qPCR at known H4K12ac-enriched regions. ENCODE projects and epigenomics roadmap efforts rely on properly validated histone PTM antibodies for accurate genomic mapping .
Chromatin Fragmentation: Optimize sonication conditions to achieve DNA fragments between 200-500 bp, which is ideal for high-resolution mapping of H4K12ac distribution.
Antibody Titration: Determine the optimal antibody-to-chromatin ratio through a titration series. Use the minimum amount of antibody that gives maximum enrichment to reduce background.
Controls: Always include:
Cross-linking Conditions: Optimize formaldehyde concentration and cross-linking time to preserve protein-DNA interactions while maintaining epitope accessibility.
Sequencing Depth: For histone modifications like H4K12ac, aim for at least 20 million uniquely mapped reads per sample to ensure adequate coverage.
Bioinformatic Analysis: Employ appropriate peak-calling algorithms (e.g., MACS2 for broad peaks) and consider the genomic distribution patterns typical of H4K12ac (often enriched near transcription start sites) when interpreting results .
Studies comparing ChIP-seq results with different antibodies targeting the same modification have revealed that antibody cross-reactivity can contribute to inaccurate mapping of histone modifications in genome-wide analyses . Therefore, validating results with orthogonal approaches or multiple antibodies is highly recommended.
When studying H4K12ac across different experimental systems, researchers should consider these methodological aspects:
Cell Cycle Synchronization: Since H4K12ac levels fluctuate during the cell cycle (particularly during S-phase when new histones are synthesized and deposited), synchronizing cells is crucial for comparative studies . Methods include:
Double thymidine block for G1/S synchronization
Nocodazole treatment for mitotic arrest
Serum starvation/release for G0/G1 transition
Species-Specific Considerations: While H4K12ac is evolutionarily conserved, the regulatory mechanisms and genomic distributions may vary between organisms:
Distinguishing Cytoplasmic vs. Nuclear H4K12ac: Different fractionation protocols allow separation of:
Cytoplasmic H4K12ac (newly synthesized, not chromatin-associated)
Nuclear soluble H4K12ac (nuclear but not chromatin-bound)
Chromatin-associated H4K12ac (incorporated into nucleosomes)
Quantification Methods:
Context-Dependent Interpretation: H4K12ac can have different functional implications depending on:
Genomic location (promoters vs. gene bodies)
Co-occurrence with other histone marks
Cell type and developmental stage
Research in Arabidopsis has revealed that H4K12ac influences chromatin accessibility differently near transcription start sites (enhanced accessibility) versus transcription termination sites (reduced accessibility) , highlighting the importance of context-specific analysis.
When encountering potential false results with HIST1H4A (Ab-12) Antibody, implement these troubleshooting strategies:
Cross-Reactivity Assessment: Commercial H4K12ac antibodies may cross-react with other acetylated lysines on H4. Studies have shown that many H4 acetyl antibodies recognize epitopes with multiple acetylation marks with stronger signal than single modifications . To address this:
Signal Verification: Confirm signals using orthogonal methods:
If ChIP-seq shows enrichment, validate with ChIP-qPCR
If Western blot shows a band, confirm with mass spectrometry
If immunostaining shows nuclear signal, verify with fractionation followed by Western blot
Epitope Masking: H4K12ac may be obscured by:
Protein-protein interactions masking the epitope
Adjacent modifications affecting antibody accessibility
Fixation methods altering epitope structure
Antibody Sensitivity: Some antibodies have detection thresholds:
Increase antibody concentration or incubation time
Try more sensitive detection systems (e.g., enhanced chemiluminescence)
Use signal amplification methods for low-abundance modifications
Technical Validation:
Include positive controls (cell types known to have high H4K12ac levels)
Try different antibody lots or suppliers
Optimize extraction methods to preserve the modification
Research has demonstrated that different H4K12ac antibodies can yield varying results in the same experimental system. For example, when tracking H3S10 phosphorylation (another histone PTM) through cell cycle progression, two different antibodies gave contradictory results, with one showing specific detection of mitotic enrichment while another showed non-specific binding . Similar issues may affect H4K12ac detection, emphasizing the importance of rigorous controls.
H4K12ac functions within a complex network of histone modifications that collectively regulate chromatin structure and function:
Co-occurrence Patterns: H4K12ac frequently co-occurs with other activating modifications:
Antagonistic Relationships: Some modifications may be mutually exclusive with H4K12ac:
H4K12 acetylation and methylation cannot occur simultaneously on the same residue
High levels of H3K9me3 or H3K27me3 (repressive marks) often correlate with reduced H4K12ac in heterochromatic regions
Sequential Modification Patterns: Research suggests a temporal order of modifications:
Functional Cross-talk: The presence of H4K12ac can influence the deposition or removal of other marks:
H4K12ac may enhance the recruitment of H3K4 methyltransferases at promoters
H4K12ac may inhibit the activity of certain histone deacetylases on neighboring residues
This complex interplay creates a "histone code" that regulates DNA accessibility to cellular machinery involved in transcription, replication, and repair . Recent research emphasizes that accurate interpretation of this code depends on antibodies that can distinguish specific modifications without cross-reactivity to neighboring PTMs .
H4K12ac contributes to numerous biological processes and has been implicated in several pathological conditions:
DNA Replication and Cell Cycle Progression:
H4K12ac, along with H4K5ac, is enriched on newly synthesized histones during S-phase
These marks facilitate nucleosome assembly by promoting histone chaperone interactions
The HAT1-RbAp46 complex preferentially acetylates H4 in H3.1-H4 tetramers (replication-dependent histones) over H3.3-H4 (replication-independent histones)
Transcriptional Regulation:
Epigenetic Memory and Inheritance:
Pathological Implications:
Dysregulation of H4K12ac has been associated with:
Cancer progression (aberrant gene activation)
Neurodegenerative disorders (memory formation defects)
Aging (global changes in acetylation patterns)
Species-Specific Functions:
Integrating H4K12ac data with other epigenomic datasets provides a more comprehensive understanding of chromatin regulation:
Multi-omics Data Integration Approaches:
| Data Type | Integration Purpose | Technical Considerations |
|---|---|---|
| ChIP-seq for other histone marks | Identify co-occurrence patterns | Use same chromatin preparation for all ChIPs when possible |
| ATAC-seq/DNase-seq | Correlate H4K12ac with chromatin accessibility | Consider sequential IP-then-accessibility assays on same samples |
| RNA-seq | Link H4K12ac to gene expression | Match time points and conditions precisely |
| DNA methylation data | Examine relationship with DNA epigenetic marks | Cell sorting may be necessary for heterogeneous populations |
| Hi-C/chromosome conformation | Connect H4K12ac to 3D genome organization | Requires specialized bioinformatic pipelines |
Computational Analysis Strategies:
Correlation analyses between H4K12ac and other epigenetic marks across genomic regions
Machine learning approaches to identify combinatorial patterns and predictive features
Trajectory analyses to understand temporal dynamics during biological processes
Network analyses to identify regulatory hubs where H4K12ac interacts with other factors
Visualization Tools and Approaches:
Genome browsers with multiple tracks for different epigenetic marks
Heatmaps centered on genomic features (TSS, enhancers, etc.)
Chromatin state segmentation models that incorporate H4K12ac data
Principal component analysis to identify major sources of variation
Validation of Integrated Findings:
Functional studies using CRISPR-based manipulation of HAT1/H4K12ac
Mass spectrometry to verify co-occurrence of modifications on the same histone tails
Single-cell approaches to address cellular heterogeneity
When integrating H4K12ac data from antibody-based experiments, it's crucial to consider potential antibody cross-reactivity issues. For example, studies using antibodies against different methylation states of H3K4 found overlapping signals due to antibody cross-reactivity, potentially confounding the interpretation of their distinct regulatory functions . Similar considerations apply to H4K12ac integration with other histone acetylation data.
Emerging technologies offer new approaches to studying H4K12ac with improved specificity and resolution:
Mass Spectrometry-Based Approaches:
Targeted MS methods can quantify H4K12ac with high precision
Middle-down proteomics allows analysis of combinatorial modifications on intact histone tails
Crosslinking MS can identify proteins that specifically interact with H4K12ac-modified histones
Engineered Readers and Binders:
Recombinant antibody fragments optimized for H4K12ac specificity
Designed proteins that bind specifically to H4K12ac-containing nucleosomes
Nanobodies with high specificity for H4K12ac for live-cell imaging
CRISPR-Based Epigenome Editing:
Targeted recruitment of HAT1 to specific genomic loci
Precise engineering of H4K12 to non-acetylatable residues at specific genes
Optogenetic control of H4K12ac deposition or removal
Single-Cell and Spatial Technologies:
Single-cell CUT&Tag for profiling H4K12ac in heterogeneous populations
Spatial omics techniques combining H4K12ac detection with tissue localization
Live-cell sensors for monitoring H4K12ac dynamics in real-time
Computational Approaches:
Deep learning models to predict H4K12ac patterns from DNA sequence and other epigenetic marks
Systems biology frameworks integrating H4K12ac into regulatory network models
Evolutionary analyses of H4K12ac conservation and divergence across species
These advanced technologies help address limitations of traditional antibody-based approaches, including the cross-reactivity issues documented in microarray studies of histone PTM antibodies . By providing orthogonal and complementary data, they contribute to a more accurate understanding of H4K12ac biology.
Ensuring reproducibility in H4K12ac research requires standardized approaches and thorough reporting:
Antibody Validation and Reporting:
Document complete antibody information (vendor, catalog number, lot, clone)
Report all validation experiments performed (peptide arrays, knockout controls, etc.)
Consider using antibodies validated by consortia like ENCODE or validated through databases like the one for histone antibody specificity assessment
Experimental Protocol Standardization:
Provide detailed protocols including buffer compositions, incubation times, and temperatures
Report cell density, passage number, and synchronization methods if applicable
Document chromatin preparation methods, sonication parameters, and fragment size verification
Controls and Quality Metrics:
Include both positive and negative controls in all experiments
Report quality control metrics (signal-to-noise ratios, enrichment at positive control regions)
Use spike-in controls (e.g., H4K12ac-containing designer nucleosomes) for quantitative experiments
Data Processing Transparency:
Share raw data in public repositories
Provide detailed bioinformatic pipelines including software versions and parameters
Document normalization methods and statistical approaches
Cross-Laboratory Validation:
Consider repeating key experiments in different labs before publication
Participate in community standard-setting efforts for histone PTM research
Compare results with orthogonal techniques when possible
The scientific community has recognized issues with histone antibody quality control, including off-target recognition and influence by neighboring PTMs . Addressing these challenges through rigorous validation and transparent reporting is essential for continued progress in understanding H4K12ac biology.
Several promising research directions will advance our understanding of H4K12ac biology:
Single-Cell Epigenomics of H4K12ac:
Investigating cell-to-cell variation in H4K12ac patterns
Tracking how H4K12ac changes during cellular differentiation at single-cell resolution
Correlating H4K12ac heterogeneity with transcriptional variability
Temporal Dynamics and Kinetics:
Measuring rates of H4K12ac deposition and removal during the cell cycle
Investigating how quickly H4K12ac responds to cellular signals
Understanding the temporal relationship between H4K12ac and transcriptional activation
Mechanistic Studies of Reader Proteins:
Identifying specific proteins that recognize and bind to H4K12ac
Determining how H4K12ac influences chromatin compaction at the molecular level
Investigating how H4K12ac affects higher-order chromatin structure
Therapeutic Targeting of H4K12ac Pathways:
Developing small molecules to modulate HAT1 activity with improved specificity
Exploring the potential of H4K12ac modulation in treating diseases with epigenetic dysregulation
Creating targeted approaches to restore normal H4K12ac patterns in disease states
Evolutionary Conservation and Divergence:
Comparing H4K12ac functions across diverse species
Understanding how H4K12ac regulatory mechanisms have evolved
Identifying species-specific aspects of H4K12ac biology
Integration with Metabolic Regulation:
Exploring how cellular metabolism influences H4K12ac through acetyl-CoA availability
Investigating connections between nutrient sensing and H4K12ac dynamics
Understanding how metabolic diseases affect H4K12ac patterns