ab15823: Peptide-blocking assays confirmed exclusive recognition of acetyl-K8 in histone H4, with no cross-reactivity to acetyl-K5 (H4) or acetyl-K9 (H2A) .
M14495-7: No cross-reactivity observed with unmodified K8 or other acetylated lysines (K5, K12, K16, K20, K31, K91) in histone H4 .
A7258: Detected a 11 kDa band in Western blots of TSA-treated NIH/3T3 and C6 cells, aligning with histone H4's molecular weight .
ab45166: Showed enhanced signal in Trichostatin A-treated HeLa cells via flow cytometry (1:20 dilution) and Western blot (1:5,000 dilution) .
07-328: Validated in ChIP-seq studies for poised chromatin analysis in germline development .
ab45166: Demonstrated efficacy in ChIP-seq for mapping acetylated histone H4 binding sites .
A7258: Recommended for ChIP with 5 μg antibody per 5–10 μg chromatin .
ab15823: Detected a 13 kDa band in calf thymus histone lysates, consistent with acetyl-K8 modification .
M14495-7: Identified acetyl-K8 in sodium butyrate-treated HeLa cells at 0.5 μg/mL .
A7258: Stained human, mouse, and rat kidney sections at 1:20 dilution with citrate buffer antigen retrieval .
ab15823: Produced clear nuclear staining in human breast adenocarcinoma tissues using a Leica BOND system .
| Parameter | ab45166 | ab15823 | M14495-7 |
|---|---|---|---|
| Clonality | Monoclonal | Polyclonal | Monoclonal |
| Signal Intensity | High (ChIP-seq) | Moderate (IHC) | High (WB) |
| Species Range | Broad | Limited | Vertebrates |
| Cross-Reactivity | None reported | None reported | None reported |
Neurodegeneration: Ethanol-induced neonatal neurodegeneration correlated with increased H4K8 acetylation, detected using 07-328 in mouse models .
Cancer: RM201 (M14495-7) revealed elevated H4K8ac in sodium butyrate-treated HeLa cells, implicating acetylation in chromatin remodeling .
Chromatin Dynamics: ab45166 facilitated the discovery of bivalent chromatin domains during mitosis-to-meiosis transitions .
Acetyl-HIST1H4A (K8) antibody specifically recognizes histone H4 when acetylated at lysine 8 (K8ac). Histone H4 is a core component of nucleosomes that wrap and compact DNA into chromatin, fundamentally regulating DNA accessibility to cellular machinery. This antibody binds specifically to the acetylated form of lysine 8 on histone H4 and does not cross-react with other acetylated lysine residues on histone H4 (such as K5, K12, K16, K20, K31, or K91) .
The specificity of this antibody is critical in epigenetic research as H4K8 acetylation represents a specific post-translational modification associated with transcriptionally active chromatin regions. Proper controls should be employed to verify specificity, including peptide competition assays with acetylated and non-acetylated peptides as demonstrated in validation studies .
Acetyl-HIST1H4A (K8) antibodies have been validated for multiple applications across different research platforms:
The antibody performs best with appropriate positive controls such as histone extracts from sodium butyrate-treated cells (HDAC inhibitor that increases acetylation) and negative controls including untreated cell extracts or blocking with specific peptides .
Based on the provided information, commercial Acetyl-HIST1H4A (K8) antibodies demonstrate species reactivity as follows:
The high conservation of histone H4 sequence across species facilitates cross-reactivity, though researchers should validate antibody performance in their specific experimental model. For evolutionarily distant organisms, epitope sequence verification is recommended prior to use .
ChIP experiments using Acetyl-HIST1H4A (K8) antibodies require careful optimization for accurate results:
Protocol Optimization:
Chromatin Preparation: Cross-link cells with formaldehyde (typically 1-1.5%, 10 minutes at room temperature)
Sonication: Fragment chromatin to 200-500 bp lengths (optimize cycles for your sonicator)
Antibody Amount: Use 1-5 μg per ChIP reaction as a starting point
Chromatin Amount: 25 μg of chromatin is a typical starting point
Essential Controls:
Input control (non-immunoprecipitated chromatin)
No antibody control (beads only)
Positive control regions (known H4K8ac-enriched genes)
Negative control regions (heterochromatic or inactive regions)
Quantification Method:
qPCR with primers for known regulatory regions
For ChIP-seq, include spike-in controls for normalization and use appropriate bioinformatic pipelines
For optimal ChIP results, cell treatment with HDAC inhibitors like sodium butyrate can serve as a positive control by increasing global H4K8 acetylation levels . Antibody specificity should be validated using peptide competition assays to ensure binding is specific to H4K8ac and not other acetylated lysines on H4 .
Successful Western blot detection of H4K8 acetylation requires specific sample preparation protocols:
Histone Extraction Protocol:
Acid Extraction: Use 0.2N HCl or specialized histone extraction kits
Nuclei Isolation: Alternative approach for enriching histones
Protein Quantification: Bradford or BCA assay adjusted for histones
Sample Handling: Maintain samples at 4°C and include protease and deacetylase inhibitors
Loading and Transfer Considerations:
Use 15-18% polyacrylamide gels for optimal separation of low molecular weight histones
Add 5-10 mM sodium butyrate to all buffers to prevent deacetylation during sample processing
Use PVDF membrane for transfer (0.2 μm pore size recommended)
Short transfer times (60-90 min) at lower voltage improve retention of small histone proteins
Detection Conditions:
Blocking: 5% BSA or milk (though some researchers report BSA is superior for phospho-epitopes)
Include both positive control (sodium butyrate-treated cells) and negative controls (untreated extracts)
For specificity verification, competitive blocking experiments with acetylated K8 peptides can be performed . The data shows that the H4K8ac antibody specifically recognizes sodium butyrate-treated samples with increased acetylation and this recognition can be blocked by the H4K8ac peptide but not by other acetylated histone peptides .
For optimal immunocytochemistry detection of H4K8 acetylation:
Cell Preparation:
Culture cells on appropriate coverslips or chamber slides
Fixation: 4% paraformaldehyde (10 min) or methanol:acetone (1:1)
Permeabilization: 0.1-0.5% Triton X-100 in PBS (5-10 min)
Blocking: 1-5% BSA or normal serum (1 hour)
Antibody Incubation:
Incubation time: 1-2 hours at room temperature or overnight at 4°C
Secondary antibody: Species-appropriate fluorophore-conjugated antibody (typically 1:500-1:2000)
Signal Enhancement and Controls:
For weak signals, consider signal amplification systems
Counterstain with DAPI for nuclear visualization
Include secondary-only control to assess background
Consider co-staining with other markers (e.g., actin for cell structure visualization)
Imaging Considerations:
Use confocal microscopy for precise nuclear localization
Z-stack imaging may be necessary to capture the full nuclear signal
Consistent exposure settings for comparative analysis
The search results show successful ICC applications with both polyclonal and monoclonal Acetyl-HIST1H4A (K8) antibodies, with reports of nuclear localization in HeLa cells and enhanced signal in sodium butyrate-treated samples .
H4K8 acetylation operates within a complex network of histone modifications that collectively regulate chromatin structure and gene expression:
Correlation with Other Acetylation Marks:
H4K8ac often co-occurs with other acetylation marks on histone H4 (K5, K12, K16) in transcriptionally active regions. These modifications work cooperatively to neutralize the positive charge of lysine residues, weakening histone-DNA interactions and promoting an open chromatin conformation . The patterns of co-occurrence can vary by cell type and genomic region.
Functional Relationships:
H4K8ac is generally associated with transcriptional activation
It may precede or follow other histone modifications in activation cascades
The presence of H4K8ac alongside H3K27ac often marks active enhancers
H4K8ac is typically depleted in regions with repressive marks like H3K9me3 or H3K27me3
Methodological Approaches for Co-occurrence Studies:
Sequential ChIP (Re-ChIP) to identify co-occurrence on the same nucleosomes
Parallel ChIP-seq experiments with antibodies against different modifications
Mass spectrometry analysis of modified histones to identify modification patterns
When designing experiments to study relationships between modifications, researchers should consider using antibodies that recognize specific individual modifications (like the H4K8ac-specific antibody) rather than antibodies recognizing multiple modifications (like EPR16606 which recognizes H4 acetylated at K5, K8, K12, and K16) , unless the experimental question specifically addresses the collective presence of these modifications.
Integrating H4K8ac ChIP-seq data with other genomics datasets requires systematic computational approaches:
Data Integration Framework:
Quality Control: Assess ChIP-seq data quality (fragment size distribution, library complexity, signal-to-noise ratio)
Peak Calling: Use appropriate algorithms (MACS2, HOMER) with input control normalization
Genome Visualization: Import data tracks into genome browsers (UCSC, IGV) alongside other genomic features
Multi-omics Correlation:
RNA-seq for gene expression correlation
ATAC-seq/DNase-seq for chromatin accessibility
Other histone marks (H3K27ac, H3K4me3, etc.)
Transcription factor binding sites
Analytical Methods:
Heatmap clustering of multiple histone modifications around transcription start sites
Correlation analysis between H4K8ac signal and gene expression levels
Motif enrichment analysis within H4K8ac peaks to identify associated transcription factors
Pathway enrichment analysis of genes associated with H4K8ac peaks
Machine learning approaches to predict functional outcomes from combinatorial histone modification patterns
Validation Strategies:
Confirm key findings with orthogonal methods (qPCR, ICC)
Test functional relationships through perturbation experiments
Use CRISPR-based approaches to modify specific regulatory regions
When performing ChIP-seq with Acetyl-HIST1H4A (K8) antibodies, researchers should follow established guidelines for antibody validation in ChIP applications, including peptide competition assays and testing on known positive/negative genomic regions . The specificity of the antibody for K8ac over other acetylated lysines is critical for accurate interpretation of the genomic distribution data .
Distinguishing between global and gene-specific H4K8 acetylation changes requires multi-level analytical approaches:
Global H4K8ac Assessment Methods:
Western Blotting: Quantifies total H4K8ac levels relative to total H4
ELISA/Luminex: Quantitative measurement of H4K8ac in histone extracts
Mass Spectrometry: Accurate quantification of multiple histone PTMs
Provides relative abundance of H4K8ac vs. unmodified H4
Can identify combinatorial modifications on the same histone tail
Gene-Specific H4K8ac Assessment:
ChIP-qPCR: Targeted approach for candidate loci
Normalize to input and/or to unchanged reference regions
Include a panel of genes representing different regulation patterns
ChIP-seq: Genome-wide distribution of H4K8ac
Differential peak calling between conditions
Spike-in normalization for accurate quantitative comparisons
CUT&RUN/CUT&Tag: Higher resolution, lower background alternatives
May offer improved sensitivity for detecting subtle changes
Integration Strategies:
Correlate global changes (Western blot) with the number and intensity of ChIP-seq peaks
Cluster genes based on H4K8ac response patterns
Integrate with transcriptomic data to correlate acetylation changes with expression changes
Compare with other histone acetylation marks to identify modification-specific effects
When interpreting results, researchers should consider that HDAC inhibitors like sodium butyrate typically induce global H4K8 acetylation increases , while gene-specific transcription factors might recruit HATs to specific loci. Proper controls, including both technical (antibody specificity) and biological (untreated vs. treated) , are essential for accurate interpretation.
ChIP experiments with Acetyl-HIST1H4A (K8) antibodies can encounter several challenges:
Common Pitfalls and Solutions:
High Background/Low Specificity
Poor Enrichment
Variable Results Between Replicates
Issue: Inconsistent immunoprecipitation or chromatin preparation
Solution: Standardize chromatin preparation protocol, maintain consistent antibody:chromatin ratios, use automated systems where possible, include spike-in controls for normalization
False Negative Results
Cross-Reactivity with Other Acetylated Lysines
Critical Quality Controls:
Include mock IP (no antibody) control
Use IgG isotype control
Test known positive regions (housekeeping genes) and negative regions
Validate key findings with orthogonal methods
The specificity tests shown in the search results demonstrate that a proper H4K8ac antibody should recognize acetylated K8 but not unmodified K8 or other acetylated lysines (K5, K12, K16, K20, K31, or K91) in H4 , which is crucial for accurate ChIP results.
Thorough validation of Acetyl-HIST1H4A (K8) antibody specificity is essential for reliable experimental results:
Comprehensive Validation Strategy:
Peptide Competition Assays
Preincubate antibody with acetylated K8 peptide (specific competition)
Preincubate with unacetylated K8 peptide (should not compete)
Preincubate with peptides containing other acetylated lysines (K5ac, K12ac, K16ac) to confirm specificity
Perform Western blot or ChIP with competed and non-competed antibody
Positive and Negative Controls
Cross-Platform Validation
Compare results across multiple techniques (WB, ChIP, ICC)
Verify consistent patterns of enrichment/depletion
Antibody Performance Metrics
Signal-to-noise ratio in different applications
Concentration-dependent response curves
Batch-to-batch consistency testing
Cross-reactivity assessment with similar epitopes
Validation Test Panel:
The search results show examples of antibody validation through specificity tests that confirm the Acetyl-HIST1H4A (K8) antibody recognizes only H4 acetylated at K8 and not other acetylated lysines .
When facing weak or inconsistent H4K8 acetylation signals, multiple optimization strategies can be employed:
Signal Enhancement Approaches:
Sample Preparation Optimization
Detection Method Refinement
Western Blot: Use high-sensitivity ECL substrates, optimize transfer conditions for small proteins, consider loading more protein (2.5-5 μg)
ChIP: Increase chromatin amount (up to 25 μg) , optimize sonication, extend antibody incubation time
ICC/IF: Implement signal amplification systems, optimize fixation (test both PFA and methanol), adjust permeabilization conditions
Antibody Optimization
Biological Signal Enhancement
Troubleshooting Inconsistent Results:
The antibody dilution recommendations from the search results (0.5-2 μg/mL for Western blot, 1:100-1:1000 for ICC/IF) provide starting points, but optimization for specific experimental systems is essential for reliable detection of H4K8 acetylation.
Interpreting H4K8 acetylation changes requires nuanced analysis within the broader epigenetic context:
Interpretative Framework:
Genomic Location Analysis
Promoter-associated H4K8ac: Typically correlates with transcriptional activation
Enhancer-associated H4K8ac: May indicate enhancer activation, particularly when co-occurring with H3K27ac
Gene body H4K8ac: Can reflect active transcriptional elongation
Intergenic H4K8ac: Potential enhancers or regulatory elements
Temporal Dynamics Considerations
Rapid H4K8ac changes (minutes to hours): Often precede transcriptional changes
Stable H4K8ac patterns: May represent established epigenetic states
Sequential histone modification events: H4K8ac may be part of a cascade of modifications
Correlation with Transcription
Direct correlation: H4K8ac increases accompanied by increased transcription
Permissive state: H4K8ac present but transcription dependent on additional factors
Uncoupled patterns: Changes in H4K8ac without corresponding expression changes may indicate priming or other non-transcriptional functions
Multi-modification Context
Data Integration Approaches:
Correlate H4K8ac ChIP-seq data with RNA-seq from matched samples
Perform time-course experiments to establish causality
Integrate with transcription factor binding data to identify recruitment mechanisms
Employ genome editing to modify H4K8 residues at specific loci
The functional significance of H4K8 acetylation stems from its role in nucleosome structure and DNA accessibility. As a core component of nucleosomes, histone H4 wraps and compacts DNA, with acetylation of K8 neutralizing positive charges and potentially weakening histone-DNA interactions to facilitate transcription factor binding and gene expression .
H4K8 acetylation has been implicated in various biological processes and pathological conditions:
Physiological Processes:
Transcriptional Regulation
Cell Cycle Regulation
Fluctuations in H4K8ac levels during different cell cycle phases
Potential role in DNA replication and chromosome segregation
Development and Differentiation
Dynamic H4K8ac patterns during cellular differentiation
Tissue-specific H4K8ac profiles reflecting developmental programs
Stress Response
Rapid changes in H4K8ac in response to environmental stressors
Potential role in activating stress-response genes
Pathological Associations:
Cancer
Neurodegenerative Disorders
Dysregulated histone acetylation including H4K8ac in conditions like Alzheimer's and Parkinson's
Therapeutic potential of restoring normal acetylation patterns
Inflammatory Conditions
H4K8ac changes at inflammatory gene loci
Potential involvement in cytokine gene regulation
Metabolic Disorders
Links to metabolic gene regulation
Crosstalk with metabolic sensors and signaling pathways
Research Applications:
Using H4K8ac antibodies to identify disease-specific epigenetic signatures
Monitoring treatment responses to epigenetic therapies
Developing targeted approaches to modulate specific genes through H4K8 acetylation
Understanding these connections helps researchers design meaningful experiments to investigate H4K8ac in their specific biological contexts. The availability of highly specific antibodies that distinguish H4K8ac from other acetylation sites is crucial for accurately mapping these relationships.
Distinguishing causality from correlation in H4K8 acetylation studies requires sophisticated experimental designs:
Causal Relationship Determination Methods:
Temporal Analysis
Time-course experiments with fine resolution sampling
Track H4K8ac changes relative to transcriptional changes
Example approach: ChIP-seq and RNA-seq at multiple timepoints after stimulus
Direct Manipulation Strategies
HAT/HDAC Modulation: Targeted recruitment of HATs/HDACs to specific loci
CRISPR-dCas9 fused to HATs (p300, CBP) or HDACs to modify H4K8ac at specific loci
Monitor consequent transcriptional changes
Histone Mutation: K→R or K→Q mutations to prevent or mimic acetylation
Requires careful experimental design in model systems
Domain-Specific Inhibitors: Target specific HAT/HDAC complexes that modify H4K8
Reader Protein Identification
Identify proteins that specifically bind H4K8ac
Disrupt these interactions to determine functional consequences
Techniques: CRISPR screens, protein-protein interaction studies, bromodomain inhibitors
Multi-omics Integration
Correlate H4K8ac changes with:
Transcription factor binding (ChIP-seq)
Chromatin accessibility (ATAC-seq)
Transcriptional output (RNA-seq)
Other histone modifications
Use causal inference statistical approaches
Experimental Design Considerations:
| Experimental Approach | Strengths | Limitations | Key Controls |
|---|---|---|---|
| Time-course analysis | Establishes temporal order | Correlation ≠ causation | Parallel profiling of multiple marks |
| CRISPR-dCas9-HAT/HDAC | Direct manipulation | Off-target effects | dCas9 only controls, multiple guide RNAs |
| Histone mutants | Direct test of function | System-wide effects | Wild-type controls, rescue experiments |
| Pharmacological intervention | Relatively simple | Low specificity | Dose-response, multiple inhibitors |
To confidently establish causality, researchers should employ multiple complementary approaches and carefully control for confounding factors. The availability of highly specific H4K8ac antibodies enables precise monitoring of acetylation changes following experimental manipulations, though researchers should remain cognizant of potential cross-reactivity with other acetylation sites when interpreting results.
Emerging technologies are revolutionizing how researchers study H4K8 acetylation:
Advanced Detection Technologies:
CUT&RUN/CUT&Tag
Higher signal-to-noise ratio than traditional ChIP
Requires less starting material (thousands vs. millions of cells)
Compatible with H4K8ac antibodies for improved genome-wide mapping
Provides higher resolution of acetylation boundaries
Single-Cell Epigenomics
scCUT&Tag, scATAC-seq with targeted antibody enrichment
Reveals cell-to-cell variation in H4K8ac patterns
Can correlate with single-cell transcriptomics for direct function assessment
Live-Cell Imaging
Antibody-derived H4K8ac sensors for real-time monitoring
FRET-based approaches to detect dynamic changes
Super-resolution microscopy for subnuclear localization
Mass Spectrometry Advances
Improved sensitivity for detecting low-abundance modifications
Ability to identify combinatorial modifications on the same histone tail
Quantitative approaches for measuring absolute acetylation levels
Functional Analysis Innovations:
CRISPR Epigenome Editing
Site-specific modulation of H4K8ac using dCas9-HAT/HDAC fusions
Allows direct testing of functional consequences at specific loci
Combinatorial editing of multiple modifications simultaneously
Acetylation-Specific Readers and Degrons
Tools to selectively bind or degrade proteins based on H4K8ac status
Enables temporal control of acetylation-dependent processes
Spatial Technologies
Coupling H4K8ac detection with spatial transcriptomics
Visualizing nuclear organization of H4K8ac domains
Multi-modal Single-molecule Approaches
Direct observation of H4K8ac dynamics on individual nucleosomes
Real-time correlation with transcriptional machinery recruitment
These technologies will depend on highly specific antibodies like those described in the search results , with recombinant monoclonal antibodies offering advantages in reproducibility and specificity . As these methods evolve, they promise to reveal new insights into the dynamic regulation and functional consequences of H4K8 acetylation in diverse biological contexts.
Several promising research directions are emerging in the field of H4K8 acetylation biology:
Identification of specific HATs and HDACs controlling H4K8 acetylation
Elucidation of recruitment mechanisms to specific genomic loci
Understanding the kinetics of H4K8 acetylation/deacetylation cycles
Mapping the integration of signaling pathways that regulate these enzymes
Comprehensive mapping of H4K8ac co-occurrence with other modifications
Understanding functional differences between isolated H4K8ac and combined patterns with other acetylation sites (K5, K12, K16)
Developing predictive models for transcriptional outcomes based on modification patterns
Identifying reader proteins that recognize specific combinations
Comparative analysis of H4K8ac landscapes across diverse cell types
Identifying tissue-specific regulatory roles and targets
Understanding context-dependent functions in different cellular environments
Single-cell analysis of H4K8ac heterogeneity within tissues
Relationship between H4K8ac and higher-order chromatin structure
Role in enhancer-promoter interactions and topologically associating domains
Contribution to phase separation and biomolecular condensate formation
Integration with chromosome conformation capture technologies
Potential roles beyond transcriptional regulation
Investigation of H4K8ac in DNA repair processes
Functions in non-coding RNA regulation
Potential non-histone protein targets of the same acetylation machinery
Development of small molecules to specifically modulate H4K8ac
Targeting writer/eraser enzymes with improved specificity
Biomarker applications in disease diagnosis and treatment response
H4K8ac-based patient stratification for precision medicine approaches
The availability of highly specific antibodies that can distinguish H4K8ac from other acetylation sites will be crucial for these research directions, particularly as techniques become more sensitive and require greater epitope specificity. Recombinant monoclonal antibodies with validated specificity offer advantages for reproducible findings across different research groups .
When developing new assays for H4K8 acetylation, researchers must implement rigorous controls and considerations:
Essential Antibody Validation Controls:
Specificity Verification
Cross-Platform Validation
Confirm consistent results across multiple applications
Compare results from different antibody clones targeting the same epitope
Validate key findings with orthogonal non-antibody methods when possible
Assay Development Considerations:
Sample Preparation Standardization
Consistent fixation protocols for cross-linking assays
Standardized extraction methods for histone proteins
Inclusion of deacetylase inhibitors throughout processing
Careful consideration of buffer compositions
Technical Parameters
Signal-to-noise optimization for each assay format
Linear dynamic range determination
Limit of detection and quantification establishment
Reproducibility assessment through technical and biological replicates
Biological Controls
Novel Assay Calibration Approach:
For any new technological approach, comprehensive comparison with established methods is essential. The search results provide examples of validated applications for H4K8ac antibodies , which can serve as benchmarks for novel assay development and validation.