The HIST1H4A (Ab-16) Antibody is a polyclonal rabbit antibody targeting the Lysine 16 (K16) site of Histone H4, a core histone protein critical for chromatin structure and gene regulation. This antibody is widely used to study histone modifications, chromatin dynamics, and epigenetic mechanisms in various biological contexts. Its specificity for the K16 site makes it valuable for investigating post-translational modifications (PTMs) at this position, including potential acetylation, phosphorylation, or ubiquitination, though its primary application does not explicitly restrict to acetylated forms .
The antibody has been validated for multiple techniques, with applications varying slightly across suppliers:
| Application | Dilution Range | Key References |
|---|---|---|
| ELISA | 1:500–1:2000 | |
| Immunofluorescence (IF) | 1:50–1:200 | |
| Western Blot (WB) | 1:500–1:5000 | |
| Immunohistochemistry (IHC) | 1:20–1:200 | |
| ChIP | Not specified |
IF: Staining patterns in HeLa cells confirm nuclear localization .
IHC: Effective in paraffin-embedded human colon and cervical cancer tissues .
The antibody exhibits cross-reactivity with histone H4 from human, mouse, and rat , though specific validation data for each species should be confirmed. Its broad reactivity makes it suitable for comparative studies across mammalian models.
While the HIST1H4A (Ab-16) antibody itself does not target acetylated K16, studies on H4K16 acetylation highlight its role in:
Chromatin structure: Acetylation at K16 is critical for chromatin decondensation and transcriptional activation .
Cellular lifespan: H4K16 acetylation antagonizes Sir2-mediated deacetylation, influencing replicative aging in yeast .
Disease association: Aberrant H4K16 acetylation is linked to cancer and epigenetic dysregulation .
HIST1H4A is a histone H4 variant that serves as a core component of nucleosomes, fundamental units that wrap and compact DNA into chromatin. This compaction limits DNA accessibility to cellular machineries requiring DNA as a template. Histone H4 plays a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability through post-translational modifications that alter chromatin structure. DNA accessibility is regulated via a complex set of these modifications, collectively known as the "histone code," alongside nucleosome remodeling processes. As a core histone protein, HIST1H4A participates in genome organization and epigenetic regulation across all eukaryotic cells, making it an essential target for epigenetics research and chromatin biology studies .
Histone H4 lysine 16 acetylation (H4K16ac) represents one of the most functionally significant histone modifications with roles extending beyond general chromatin decondensation. This specific acetylation contributes substantially to transcriptional regulation by promoting chromatin accessibility and is prominently associated with active genes. H4K16ac is uniquely involved in critical cellular processes including DNA damage repair mechanisms and cellular senescence pathways. Unlike other histone H4 acetylation marks, H4K16ac shows distinct genome-wide distribution patterns, particularly enriched around transcription start sites as revealed by ChIP-seq analyses. Loss of H4K16ac has been implicated in various disease states, particularly cancer, where aberrant regulation of this modification correlates with altered gene expression profiles . The specific detection of this modification provides researchers crucial insights into both normal chromatin dynamics and disease-associated epigenetic alterations.
HIST1H4A (Ab-16) antibodies specifically recognize histone H4 acetylated at lysine 16, distinguishing them from antibodies targeting other modification sites on histone H4. These antibodies are generated using immunogens consisting of peptide sequences surrounding the acetylated Lys16 position derived from human histone H4 . Unlike antibodies targeting other histone H4 modifications such as methylation at K20 (meLys20) or acetylation at different positions (K5, K8, K12), HIST1H4A (Ab-16) antibodies are engineered for high specificity to the acetyl-Lys16 epitope. Their specificity enables precise detection of this modification in various experimental contexts. The recognition profile differs from that of H4K5ac and H4K12ac antibodies, which are associated with newly assembled chromatin, or H4K20me antibodies, which mark condensed chromatin regions . This specificity is crucial for accurate characterization of epigenetic modifications involved in transcriptional regulation and DNA repair processes.
HIST1H4A (Ab-16) antibodies have been validated for multiple experimental applications in epigenetic research. The primary validated applications include:
| Application | Recommended Dilution | Sample Types | Detection Method |
|---|---|---|---|
| Western Blotting (WB) | 1:200-1:2000 | Cell/tissue lysates | Chemiluminescence |
| Immunofluorescence (IF) | 1:50-1:200 | Fixed cells | Fluorescence microscopy |
| Immunocytochemistry (ICC) | 1:20-1:200 | Fixed cells | Brightfield/fluorescence |
| Chromatin Immunoprecipitation (ChIP) | Application-specific | Chromatin fragments | qPCR/sequencing |
| ELISA | Protocol-dependent | Purified histones | Colorimetric/fluorometric |
| Immunohistochemistry (IHC) | 1:1-1:10 | Fixed tissue sections | Brightfield microscopy |
These applications enable researchers to detect and quantify H4K16ac modifications in diverse experimental contexts, from protein-level detection to genome-wide distribution analysis. Each application requires specific optimization based on sample type and experimental conditions to achieve optimal signal-to-noise ratio and specificity .
ChIP-seq experiments using HIST1H4A (Ab-16) antibodies require careful optimization for successful characterization of genome-wide H4K16ac distribution. Begin with cross-linking optimization (typically 1% formaldehyde for 10 minutes) followed by chromatin fragmentation to 200-500bp fragments, verified by gel electrophoresis. Antibody specificity validation is crucial before proceeding; perform preliminary ChIP-qPCR targeting known H4K16ac-enriched regions like active promoters compared to silent genes. For immunoprecipitation, use 2-5μg of HIST1H4A (Ab-16) antibody per reaction with 25-50μg of chromatin, including IgG controls and input samples. Multiple biological replicates are essential for statistical robustness. Library preparation should include size selection and quality control steps before sequencing.
During data analysis, H4K16ac typically shows enrichment around transcription start sites of active genes and enhancers. ChIP-seq profiles from previous studies demonstrate that H4K16ac peaks correlate with DNase I hypersensitive sites and active transcription marks like H3K27ac . For experimental validation, include spike-in controls with known concentrations of modified histones. This comprehensive approach ensures reliable characterization of H4K16ac distribution patterns across the genome, providing insights into regulatory mechanisms controlling gene expression and chromatin accessibility.
For optimal immunofluorescence results with HIST1H4A antibodies, implement a two-stage fixation protocol that preserves nuclear architecture while maintaining epitope accessibility. Begin with a brief prefixation using 0.3% formaldehyde in PBS for 3 minutes at room temperature, followed by a more thorough fixation with 3% formaldehyde for 15 minutes. This approach minimizes epitope masking while adequately preserving chromatin structure. After fixation, permeabilize cells using a buffer containing 0.5% Triton X-100 in PBS for 10 minutes at room temperature, which enables antibody access to nuclear epitopes without disrupting chromatin organization.
Critical optimization parameters include:
Avoiding methanol fixation which can destroy the H4K16ac epitope
Implementing antigen retrieval (10mM sodium citrate, pH 6.0 at 95°C for 15 minutes) for formalin-fixed samples
Using blocking solutions containing 5% BSA and 0.1% Tween-20 to minimize background
Extending primary antibody incubation to overnight at 4°C at dilutions of 1:50-1:200
Including positive controls (cells treated with histone deacetylase inhibitors) and negative controls (cells treated with histone acetyltransferase inhibitors)
This optimized protocol significantly enhances signal-to-noise ratio and enables reliable visualization of H4K16ac distribution patterns in the nucleus, particularly when combined with confocal microscopy for high-resolution imaging .
Verifying antibody specificity is crucial for reliable epigenetic research. Implement a multi-tiered validation approach beginning with peptide competition assays where pre-incubation of the antibody with acetylated H4K16 peptides should abolish signal, while incubation with unmodified or differently modified peptides should not affect binding. Western blot analysis should show a single band at approximately 11-15 kDa corresponding to histone H4, with signal intensity increasing in cells treated with histone deacetylase inhibitors like trichostatin A. Critically, perform cross-reactivity testing using peptide arrays containing various histone modifications to ensure the antibody specifically recognizes H4K16ac without binding to other acetylation sites (H4K5ac, H4K8ac, H4K12ac).
For genetic validation, employ cells with genetic knockdown of histone acetyltransferases specific for H4K16 (such as hMOF/KAT8) which should show reduced signal. Additionally, validate using histone mutants where lysine 16 is substituted with arginine (K16R) to prevent acetylation. Research has demonstrated that high-quality HIST1H4A (Ab-16) antibodies show minimal cross-reactivity with other acetylation sites on histone H4, though some commercial antibodies may exhibit 20-25% validation failure rates . This comprehensive validation ensures experimental results accurately reflect H4K16ac biology rather than antibody artifacts.
The performance and longevity of HIST1H4A antibodies depend significantly on proper storage and handling practices. Store antibodies in small aliquots (10-20 μl) at -80°C for long-term storage to minimize freeze-thaw cycles, which can cause protein denaturation and epitope recognition loss. For working solutions, maintain at 4°C for no more than two weeks, adding preservatives like 0.02% sodium azide to prevent microbial growth. Glycerol addition (final concentration 30-50%) helps prevent freeze-damage during storage. Avoid repeated freeze-thaw cycles; each cycle typically reduces activity by 10-15% based on experimental measurements of binding efficiency.
When handling these antibodies, maintain sterile conditions and use low-protein binding tubes to prevent adsorption-based losses. Temperature transitions should be gradual; thaw frozen aliquots on ice rather than at room temperature. Centrifuge briefly before opening tubes to collect any solution from the cap. Performance validation through regular quality control testing is essential; check antibody function with positive controls at least every three months. Data from stability studies show that properly stored HIST1H4A antibodies maintain >90% of their initial activity for approximately 12 months, after which gradual decline may occur. Implement a consistent batch testing protocol when obtaining new lots to ensure experimental continuity and reproducibility .
Sample preparation methodology critically influences epitope recognition and signal quality when using HIST1H4A (Ab-16) antibodies. Fixation protocols substantially impact epitope accessibility, with over-fixation using formaldehyde (>4% or >20 minutes) causing significant epitope masking through protein cross-linking. Studies show that acetylation-specific antibodies like those targeting H4K16ac are particularly sensitive to fixation conditions, with optimal results typically achieved using 1-2% formaldehyde for 10 minutes at room temperature. Importantly, avoid methanol fixation which can destroy acetylation marks through dehydration effects.
For protein extraction in Western blotting applications, acid extraction methods (0.2N HCl or triton extraction) preserve histone modifications better than conventional RIPA buffers, which can lead to deacetylase activity and epitope loss. Sample heating should be limited to 95°C for 5 minutes maximum, as extended boiling reduces antibody recognition by 40-60%. For immunoprecipitation, native chromatin preparations often yield better results than crosslinked samples due to reduced epitope masking.
Antigen retrieval methods significantly impact immunohistochemistry and immunofluorescence applications; citrate buffer (pH 6.0) typically outperforms Tris-EDTA (pH 9.0) for preserving H4K16ac epitopes in fixed tissues. Comparative analysis of different extraction protocols demonstrates that specialized histone extraction kits yield 30-40% higher signal intensity compared to generic protein extraction methods when analyzed by Western blotting with HIST1H4A (Ab-16) antibodies .
When confronted with conflicting results between different detection methods using HIST1H4A antibodies, implement a systematic analytical approach. First, consider method-specific technical limitations: ChIP-seq provides genome-wide distribution but may suffer from antibody efficiency variations, while Western blotting detects total protein levels but lacks spatial resolution. Immunofluorescence offers subcellular localization information but may be affected by fixation artifacts. Different methods have varying detection sensitivities; Western blotting can detect approximately 1-5% changes in global H4K16ac levels, while ChIP-qPCR might require 2-3 fold enrichment for statistical significance.
To resolve discrepancies, implement orthogonal validation using alternative antibodies targeting the same modification or employ mass spectrometry for unbiased modification analysis. Consider biological context—H4K16ac shows cell cycle-dependent fluctuations with highest levels in G1 phase and lowest during S phase. For example, a study found that while Western blotting showed minimal global changes in H4K16ac following certain treatments, ChIP-seq revealed significant redistribution of this mark across the genome rather than absolute level changes. Additionally, examine experiment-specific variables such as crosslinking efficiency for ChIP, extraction methods for Western blotting, or fixation protocols for microscopy. Document all experimental parameters meticulously, as minor protocol variations can significantly impact histone modification detection results .
Several factors contribute to false results when using HIST1H4A (Ab-16) antibodies. False positives commonly arise from cross-reactivity with similar histone modifications, particularly other acetylated lysines on histone H4 (K5, K8, K12) which share sequence context similarities. Studies show approximately 15-20% of commercial antibodies exhibit cross-reactivity with these related modifications . Non-specific binding to denatured or exposed histone epitopes in improperly prepared samples represents another source of false positives. Inadequate blocking (particularly when using milk, which contains bioactive proteins) can contribute to background signal mistakenly interpreted as positive.
False negatives frequently result from epitope masking during sample preparation, particularly over-fixation with formaldehyde which can reduce antibody accessibility to the acetylated lysine 16. Technical research demonstrates that extending formaldehyde fixation from the recommended 10 minutes to 30 minutes can reduce signal detection by up to 70% . Post-translational modification interdependence also influences antibody recognition; neighboring modifications like phosphorylation at serine 1 can sterically hinder antibody access to the K16ac epitope. Sample degradation through active histone deacetylases during extraction represents another significant concern, with studies showing that omitting deacetylase inhibitors during extraction can result in 40-60% signal loss within 30 minutes at room temperature. Implementing appropriate controls, including peptide competition assays and samples with known modification status, helps distinguish true signals from artifacts .
Distinguishing biological variation from technical artifacts requires implementing comprehensive control strategies. Begin by establishing cell type-specific baselines using multiple antibody clones targeting H4K16ac, as different cell types naturally exhibit varying H4K16ac levels based on their transcriptional programs. Internal controls are essential: analyze housekeeping genes known to maintain consistent H4K16ac levels across cell types as technical references. Employ spike-in normalization using chromatin from a different species (e.g., Drosophila chromatin in human samples) to provide an invariant reference for quantifying technical variability.
For sequencing applications, prepare biological replicates processed in different batches to identify batch effects versus true biological variation. Statistical approaches including principal component analysis help separate technical noise from biological signal. Cell cycle normalization is crucial—synchronize cells or use cell cycle markers in analysis, as H4K16ac levels naturally fluctuate during the cell cycle, potentially masking or exaggerating cell type differences. Research has shown that S-phase cells typically display reduced H4K16ac compared to G1-phase cells from the same population .
When analyzing multiple cell types, prepare all samples simultaneously using identical reagent lots and protocols. For challenging comparisons, orthogonal validation using mass spectrometry quantification of H4K16ac provides antibody-independent verification. Consider environmental factors affecting epigenetic marks; cell culture conditions including confluence levels can alter H4K16ac patterns by 15-20% based on quantitative analyses. This systematic approach helps differentiate genuine biological differences from methodology-induced variations .
Advanced multiplexed analysis of histone modifications using HIST1H4A (Ab-16) antibodies enables comprehensive epigenetic profiling beyond single-modification studies. Implement sequential ChIP (re-ChIP) protocols, where chromatin is immunoprecipitated with H4K16ac antibodies followed by a second immunoprecipitation with antibodies against other modifications. This approach identifies genomic regions where H4K16ac co-occurs with other marks, revealing modification interdependencies. For example, studies have uncovered that H4K16ac frequently co-localizes with H3K4me3 at active promoters but shows mutual exclusivity with H4K20me3 repressive marks .
For microscopy applications, employ antibodies conjugated to spectrally distinct fluorophores, such as Alexa Fluor 647-conjugated anti-H4K16ac antibodies combined with different fluorophore-conjugated antibodies against other histone marks . This multiplexed immunofluorescence approach preserves spatial relationships between modifications within nuclear architecture. Mass spectrometry-based approaches complement antibody-based methods by enabling quantitative analysis of combinatorial histone modifications; use targeted mass spectrometry with heavy isotope-labeled peptide standards containing H4K16ac to quantify this modification in relation to others on the same histone tail.
Cutting-edge CUT&RUN or CUT&Tag protocols offer higher sensitivity than traditional ChIP, using protein A-micrococcal nuclease or protein A-Tn5 transposase fusions to generate modification-specific DNA fragments with minimal background. These techniques reduce input requirements to approximately 1,000-50,000 cells compared to the millions needed for conventional ChIP, enabling analysis of rare cell populations or clinical samples with limited material availability .
Studies utilizing HIST1H4A antibodies have revealed critical insights into H4K16ac's role in various disease states, particularly cancer and neurodegenerative disorders. In cancer biology, comprehensive analyses using these antibodies have demonstrated global hypoacetylation of H4K16 across multiple cancer types, with an average 25-40% reduction compared to matched normal tissues. This reduction correlates with decreased expression of KAT8/hMOF acetyltransferase, which specifically targets H4K16. ChIP-seq studies have further identified that loss of H4K16ac occurs predominantly at tumor suppressor gene loci, directly linking this epigenetic alteration to oncogenic gene expression programs. Interestingly, quantitative analysis shows progressive loss of H4K16ac correlating with advancing cancer stages, suggesting potential as a prognostic biomarker.
In neurodegenerative disorders, particularly Alzheimer's disease, immunohistochemical studies using H4K16ac antibodies have revealed significant alterations in this modification in post-mortem brain tissues. Neurons in affected brain regions show up to 60% reduction in H4K16ac compared to age-matched controls. Mechanistically, research has demonstrated that amyloid-β exposure leads to rapid deacetylation of H4K16 through SIRT1 activation, affecting expression of neuronal survival genes. Furthermore, H4K16ac levels are significantly altered in cellular models of DNA damage and aging, with immunofluorescence studies showing this modification is redistributed to sites of DNA damage, consistent with its role in maintaining genomic stability. These findings collectively highlight the potential of targeting H4K16ac regulatory mechanisms for therapeutic intervention in multiple disease contexts .
HIST1H4A antibodies have enabled significant advances in understanding chromatin assembly dynamics through their ability to distinguish specific modification patterns associated with newly synthesized histones. Pulse-chase experiments combined with immunofluorescence using these antibodies have revealed that newly synthesized histone H4 is predominantly acetylated at K5 and K12 but lacks K16 acetylation during initial chromatin deposition . This specific modification pattern serves as a signature for newly assembled nucleosomes. ChIP-seq studies utilizing HIST1H4A (Ab-16) antibodies alongside other modification-specific antibodies have mapped the genome-wide transition from assembly-associated modifications to mature chromatin marks, demonstrating that H4K16ac is established post-deposition through the activity of specific acetyltransferases, primarily KAT8/hMOF.
Quantitative time-course experiments have established that H4K16ac acquisition occurs with distinct kinetics across different genomic regions—appearing within 20-30 minutes at active gene promoters but taking several hours to accumulate at other regulatory regions. This differential timing suggests context-dependent regulation of acetyltransferase recruitment. Super-resolution microscopy combined with HIST1H4A antibodies has visualized the spatial organization of H4K16ac domains during chromatin maturation, revealing that this modification helps establish and maintain euchromatic territories within nuclear architecture.
In cell cycle studies, HIST1H4A antibodies have demonstrated that H4K16ac levels fluctuate predictably, with maximum levels in G1 phase followed by reduction during S phase when newly synthesized, unacetylated histones dilute the existing modified population. These modifications are then progressively re-established as chromatin matures in G2, creating a dynamic modification landscape that influences gene expression patterns throughout the cell cycle .
Single-cell epigenomic technologies incorporating HIST1H4A antibodies are revolutionizing our understanding of cellular heterogeneity in histone modification patterns. Single-cell CUT&Tag protocols utilizing HIST1H4A (Ab-16) antibodies can now map H4K16ac distribution in individual cells, revealing previously undetectable subpopulations with distinct epigenetic signatures. Technical advances have reduced input requirements to single cells while maintaining specificity for H4K16ac. Studies employing these techniques have identified distinct H4K16ac patterns correlating with cell states in developmental processes and tissue differentiation. For example, single-cell analysis of neural differentiation revealed progressive establishment of H4K16ac at neuron-specific enhancers, with intermediate progenitor cells showing heterogeneous modification patterns not detectable in bulk analyses.
Microfluidic platforms combined with barcoding strategies enable high-throughput processing of thousands of individual cells for H4K16ac profiling, generating comprehensive atlases of acetylation patterns across tissues. Computational integration of single-cell H4K16ac data with other epigenetic marks and transcriptional profiles has established multi-dimensional maps of cellular states, revealing how H4K16ac variations contribute to cell fate decisions. Recent technological developments have enabled combined single-cell genome and epigenome profiling, allowing researchers to correlate genetic variation with H4K16ac patterns in the same cell. This integrated approach has particular relevance for cancer heterogeneity studies, where genetic subclones within tumors display distinct H4K16ac landscapes that influence their aggressive phenotypes and therapeutic responses .
Recent advances in antibody engineering have significantly enhanced the performance characteristics of HIST1H4A (Ab-16) antibodies. Recombinant monoclonal antibody technology has largely supplanted traditional polyclonal approaches, providing superior batch-to-batch consistency and specificity. For instance, rabbit recombinant monoclonal antibodies like EPR1004 demonstrate approximately 30-40% higher specificity for H4K16ac compared to polyclonal alternatives when tested against peptide arrays . These engineered antibodies incorporate targeted amino acid substitutions in complementarity-determining regions (CDRs) that enhance both affinity and specificity for the acetylated lysine 16 epitope.
Fragment antibody (Fab) and single-chain variable fragment (scFv) derivatives offer improved tissue penetration for applications like whole-mount immunofluorescence and thick-section imaging, reducing background while maintaining specificity. Novel conjugation chemistries enabling site-specific attachment of fluorophores or enzymes at defined positions away from antigen-binding regions preserve full binding capacity. This represents a significant improvement over traditional random conjugation methods that can reduce effective binding by 15-25% due to modification of critical binding residues.
Bispecific antibody formats that simultaneously recognize H4K16ac and another histone modification enable direct detection of modification co-occurrence without sequential immunoprecipitation steps. For super-resolution microscopy applications, specialized mini-antibodies with reduced size (approximately one-third the size of conventional IgGs) provide improved resolution by decreasing the distance between the fluorophore and the target epitope. Additionally, nanobody-based detection systems derived from camelid antibodies offer similar advantages through their compact size (15kDa vs 150kDa for conventional antibodies) while maintaining high specificity and affinity for H4K16ac targets .
Integrative multi-omics approaches incorporating HIST1H4A antibody data enable comprehensive epigenetic analysis that contextualizes H4K16ac patterns within broader regulatory networks. Begin by implementing coordinated experimental designs that maximize compatibility between ChIP-seq data from H4K16ac immunoprecipitation and other genomic datasets. For optimal integration with transcriptomic data, perform RNA-seq and ChIP-seq on matched samples, ideally from the same cell population, to establish direct correlations between H4K16ac occupancy and gene expression levels. Research has demonstrated that genes with high H4K16ac enrichment at promoters show approximately 3-4 fold higher expression levels compared to genes lacking this modification .
For chromatin accessibility integration, combine H4K16ac ChIP-seq with ATAC-seq or DNase-seq, revealing how this modification correlates with open chromatin states. Computational approaches including multivariate hidden Markov models effectively integrate these datasets to define chromatin states characterized by specific combinations of histone modifications and accessibility. For three-dimensional chromatin organization analysis, integrate H4K16ac profiles with Hi-C or ChIA-PET data to examine how this modification influences higher-order chromatin structure and long-range interactions.
Advanced computational frameworks like multimodal deep learning algorithms can now integrate H4K16ac ChIP-seq with DNA methylation data from WGBS, transcription factor binding profiles, and nucleosome positioning maps to build predictive models of gene regulation. These integrated analyses have revealed that H4K16ac serves as a stronger predictor of active enhancers when combined with H3K27ac data than either modification alone. For clinical applications, integration of H4K16ac profiles with patient genetic data and clinical outcomes enables identification of epigenetic signatures associated with disease progression and treatment response, highlighting the translational potential of integrated epigenomic analyses incorporating HIST1H4A antibody data .