Histone H4 (encoded by HIST1H4A) is a 103-amino acid protein that, alongside other histones, forms the nucleosome core around which DNA wraps. It plays roles in transcription regulation, DNA repair, and chromosomal stability . The HIST1H4A antibody is raised against specific epitopes of Histone H4 to enable its detection in experimental assays.
Two primary formats are available:
| Catalog Number | Type | Host | Reactivity | Applications |
|---|---|---|---|---|
| ABIN7181576 | Polyclonal | Rabbit | Human | ELISA, ICC |
| A14495-2 | Polyclonal | Rabbit | Human, Mouse, Rat | WB, ICC, IF, IHC |
| MAB20165 | Monoclonal | Rabbit | Human, Mouse, Rat | ChIP, Flow Cytometry, IP, ICC, IHC |
| A14495 | Polyclonal | Rabbit | Human | ELISA, WB |
Histone H4 antibodies are critical for studying chromatin remodeling, gene expression, and epigenetic regulation. Post-translational modifications (e.g., acetylation, methylation) of Histone H4 influence chromatin accessibility and transcriptional outcomes .
HIST1H4A (Histone Cluster 1, H4a) is a core component of nucleosomes, the basic structural units of chromatin. This histone protein plays a critical role in DNA packaging and gene regulation through post-translational modifications. As a fundamental element in the "histone code," HIST1H4A and its modifications influence transcription regulation, DNA repair, DNA replication, and chromosomal stability .
Histone H4 modifications are particularly important in epigenetic research because they serve as markers for chromatin states, with specific modifications (acetylation, methylation, phosphorylation) correlating with gene activation or repression. The ability to detect these modifications using specific antibodies enables researchers to map epigenetic landscapes across the genome.
Distinguishing between HIST1H4A antibodies requires careful consideration of their specific binding properties:
| Modification Target | Common Applications | Typical Host | Clonality | Example Catalog Numbers |
|---|---|---|---|---|
| Acetyl-Lys5 (K5) | WB, ChIP, IF, IHC | Rabbit | Monoclonal/Polyclonal | MAB9549, orb418625 |
| Acetyl-Lys8 (K8) | ELISA, IF, ChIP, ICC | Rabbit | Polyclonal | Listed in search result 1 |
| Acetyl-Lys12 (K12) | ELISA, IF, ChIP, ICC | Rabbit | Polyclonal | ABIN7139153 |
| Acetyl-Lys16 (K16) | WB, IF, ChIP, ICC | Rabbit | Polyclonal/Monoclonal | ABIN7139167, M14495-3 |
| Pan-H4 (no specific modification) | WB, IHC, ChIP, IP | Rabbit | Polyclonal | ab10158, ab7311 |
When selecting an antibody, consider:
The specific modification you're targeting
Required applications (ChIP vs. WB vs. IHC)
Species reactivity needed (human, mouse, rat)
Whether you need a monoclonal (higher specificity) or polyclonal (potentially higher sensitivity)
For successful Chromatin Immunoprecipitation with HIST1H4A antibodies:
Crosslinking optimization: For histone modifications, use 1% formaldehyde for 10 minutes at room temperature for efficient DNA-protein crosslinking .
Sonication parameters: Aim for chromatin fragments between 200-500bp for optimal resolution. This typically requires 10-15 cycles (30 seconds ON/30 seconds OFF) with modern sonicators.
Antibody quantity: Use 2-5μg of HIST1H4A antibody per ChIP reaction with 25μg of chromatin . Validation experiments show that antibodies like ab10158 perform effectively at this ratio.
Controls: Always include:
Input control (non-immunoprecipitated chromatin)
IgG negative control (non-specific antibody)
Positive control (known target region)
Washing stringency: For histone modifications, perform four washes with increasing salt concentration to reduce background while maintaining specific binding.
Quantification method: Real-time PCR (Taqman or Sybr green approaches) is recommended for quantifying immunoprecipitated DNA, with primers located in the first kb of the transcribed region .
Optimizing western blot protocols for HIST1H4A detection requires specific adjustments:
Sample preparation:
Gel selection:
Use 15-18% SDS-PAGE gels to resolve the low molecular weight (11-12 kDa) histone proteins
Consider specialized Triton-Acid-Urea (TAU) gels for separating differentially modified histones
Transfer conditions:
Use PVDF membrane (0.2μm pore size) for better retention of small proteins
Transfer at lower voltage (30V) overnight at 4°C for efficient transfer
Antibody dilution:
Detection optimization:
Validation: Test specificity using histone deacetylase inhibitors (e.g., sodium butyrate, TSA) to increase acetylation levels as positive controls .
Multiple bands or unexpected patterns with HIST1H4A antibodies could result from:
Post-translational modifications: Histone H4 undergoes numerous modifications (acetylation, methylation, phosphorylation) that can alter migration patterns. For example, hyperacetylation can cause slower migration .
Cross-reactivity: Some antibodies may cross-react with other histone variants due to high sequence homology. The search results indicate that many HIST1H4A antibodies recognize multiple histone H4 family members (HIST1H4A through HIST1H4L) .
Proteolytic degradation: Incomplete protease inhibition during sample preparation can result in degradation products appearing as lower molecular weight bands.
Antibody specificity issues: Especially for modification-specific antibodies, the specificity for a particular modification may be compromised by similar epitopes on other histones or proteins.
Ubiquitination or other large modifications: Larger modifications can cause significant mobility shifts, resulting in higher molecular weight bands.
To address these issues:
Include positive and negative controls for the specific modification
Use purified histones as reference standards
Test with different antibody lots or sources
Perform peptide competition assays to confirm specificity
When facing contradictory results between different HIST1H4A antibody-based assays:
Consider epitope accessibility differences: Different assays (ChIP, IF, WB) have different sample preparation methods that may affect epitope accessibility. For example, an antibody might work well in ChIP but poorly in IHC due to fixation effects on epitope structure .
Evaluate antibody specificity:
Modification-specific antibodies (e.g., acLys16) may have different cross-reactivity profiles
Some antibodies may recognize multiple modifications or be sensitive to neighboring modifications
Analyze experimental conditions:
Different buffers, pH, and salt concentrations between assays can affect antibody binding
Fixation methods in IHC/IF can mask epitopes
Validation approach:
Use orthogonal techniques (mass spectrometry, ELISA) to validate findings
Test with cells treated with HDAC inhibitors or HAT inhibitors to modulate acetylation levels
Use genetic models (knockout/knockdown of histone modifying enzymes) to confirm specificity
Quantitative considerations:
ChIP provides relative enrichment data while IF/IHC provides spatial information
WB can be more quantitative but loses spatial information
A systematic approach comparing results across techniques with appropriate controls is essential for accurate interpretation.
Investigating chromatin dynamics during cell cycle progression using HIST1H4A antibodies requires a multi-faceted approach:
Cell synchronization techniques:
Double thymidine block for G1/S boundary
Nocodazole treatment for M phase
Serum starvation/reintroduction for G0/G1
Temporal analysis strategy:
Multimodal analysis:
ChIP-seq with phase-specific cells to map genome-wide distributions
Immunofluorescence microscopy for spatial organization in nucleus
Flow cytometry with cell cycle markers and H4 modification antibodies for quantitative assessment
Functional validation:
Inhibit specific modifying enzymes at defined cell cycle stages
Measure consequences on cell cycle progression
Correlate with transcriptional and replication timing data
This approach allows for comprehensive mapping of dynamic H4 modification patterns throughout the cell cycle and their functional implications.
Validating modification-specific HIST1H4A antibodies requires multiple complementary approaches:
Peptide competition assays:
Genetic manipulation approaches:
CRISPR/Cas9 to create point mutations at specific modification sites (e.g., K16R to prevent acetylation)
Knockdown/knockout of specific histone modifying enzymes (e.g., HDAC inhibitors for acetylation sites)
Overexpression of modifying enzymes to increase target modification
Pharmacological manipulation:
Orthogonal technical validation:
Mass spectrometry to confirm modification status
Multiplex ChIP using different antibodies targeting the same modification
Sequential ChIP (re-ChIP) to confirm co-occurrence of modifications
Compare results across multiple antibody sources/clones
Cross-reactivity assessment:
Test against synthetic peptide arrays containing various histone modifications
Evaluate signal with modified vs. unmodified recombinant histones
Assess reactivity across species to leverage evolutionary conservation
These comprehensive validation strategies ensure the reliability of experimental results when using modification-specific HIST1H4A antibodies.
Incorporating HIST1H4A antibodies into single-cell epigenomics requires adaptation of traditional methods:
Single-cell CUT&Tag/CUT&RUN modifications:
Immobilize single cells on concanavalin A-coated magnetic beads
Use HIST1H4A antibodies (particularly modification-specific ones) as primary antibodies
Employ pA-Tn5 fusion proteins for tagmentation
Incorporate cell barcoding strategies during library preparation
This approach provides genome-wide profiles of H4 modifications at single-cell resolution
Single-cell immunofluorescence quantification:
Use high-content imaging platforms with HIST1H4A antibodies
Employ computational image analysis for quantification
Correlate with other single-cell markers (transcription factors, RNA)
Provides spatial context missing from sequencing approaches
Mass cytometry (CyTOF) integration:
Conjugate HIST1H4A antibodies with heavy metal isotopes
Combine with cell cycle markers and other epigenetic modifications
Enables high-dimensional analysis of histone modification patterns
Allows correlation with cellular phenotypes in heterogeneous populations
Single-cell multi-omics strategies:
Combine scCUT&Tag using HIST1H4A antibodies with scRNA-seq
Integrate with chromatin accessibility data (scATAC-seq)
Develop computational methods to align epigenetic and transcriptomic data
Creates comprehensive single-cell epigenetic landscapes
These approaches enable researchers to resolve cellular heterogeneity in histone modification patterns that would be masked in bulk analyses.
Using HIST1H4A antibodies for live-cell imaging presents unique challenges and considerations:
Antibody delivery strategies:
Cell-penetrating peptide conjugation to antibodies
Microinjection of fluorescently labeled antibodies
Electroporation for temporary membrane permeabilization
Specialized protein transfection reagents
Alternative approaches:
Use modification-specific nanobodies instead of conventional antibodies
Consider recombinant modification-specific binding domains (e.g., bromodomains for acetylation)
Express fluorescently tagged readers of histone modifications rather than direct antibody labeling
Technical optimizations:
Test different fluorophore conjugations for optimal signal-to-noise ratio
Titrate antibody concentration to minimize interference with normal cellular functions
Employ rapid image acquisition to capture dynamic changes
Use spinning disk or light sheet microscopy for reduced phototoxicity
Controls and validation:
Compare live imaging results with fixed-cell immunofluorescence
Validate with cells treated with modification-altering compounds (HDAC inhibitors)
Use non-binding antibody controls to assess background
Include genetic controls (modification site mutants)
Limitations to consider:
Antibodies may interfere with normal chromatin dynamics
Access to densely packed heterochromatin may be limited
Resolution constraints for visualizing individual nucleosomes
Potential functional interference with histone-protein interactions