The Crotonyl-HIST1H4A (K5) Antibody specifically recognizes the crotonylation modification at lysine 5 (K5) of histone H4. Histone H4 is a core component of nucleosomes, which wrap and compact DNA into chromatin. This modification is part of the histone code that regulates DNA accessibility to cellular machinery requiring DNA as a template . The antibody binds to the peptide sequence around the site of crotonyl-lysine at position 5 derived from human histone H4 . This post-translational modification plays a crucial role in epigenetic gene regulation, DNA repair, DNA replication, and chromosomal stability .
The antibody has been validated for multiple research applications across various platforms:
| Application | Validation Status | Recommended Dilution Range |
|---|---|---|
| Western Blot (WB) | Validated | 1:500-5000 |
| Immunofluorescence (IF) | Validated | 1:50-200 |
| Immunoprecipitation (IP) | Validated | 1:200-2000 |
| Chromatin Immunoprecipitation (ChIP) | Validated | As per manufacturer protocol |
| ELISA | Validated | As per manufacturer protocol |
These applications allow researchers to investigate the presence, distribution, and relative abundance of H4K5 crotonylation in different experimental contexts .
Crotonylation is structurally distinct from acetylation, featuring a four-carbon chain with an α,β-unsaturated carbonyl group, compared to acetylation's single-carbon modification. This structural difference results in a more bulky modification that may create different binding surfaces for reader proteins. While acetylation at H4K5 is often associated with newly assembled histones (especially when K12 is also acetylated but K8 is unacetylated), crotonylation at the same position may have distinct functional implications in gene regulation . The distribution patterns of H4K5 crotonylation can be distinguished from other modifications using specific antibodies like the one discussed here, allowing researchers to investigate the unique biological roles of this modification .
For optimal Western blot results with Crotonyl-HIST1H4A (K5) Antibody:
Sample preparation: Extract histones using acid extraction methods to maximize histone yield and preservation of modifications.
Gel selection: Use 15-18% SDS-PAGE gels to properly resolve the low molecular weight histone proteins (~11-15 kDa).
Transfer conditions: Optimize transfer to PVDF membranes (preferred over nitrocellulose) using 20% methanol buffer at lower voltage (30V) for longer periods (2 hours) to ensure efficient transfer of small proteins.
Blocking: Use 5% BSA in TBST rather than milk, as milk contains enzymes that may remove some histone modifications.
Antibody dilution: Start with a 1:1000 dilution for polyclonal antibodies and 1:2000 for monoclonal versions, optimizing as needed .
Validation controls: Include positive controls (cells known to exhibit H4K5 crotonylation) and negative controls (samples treated with decrotonylase enzymes) .
Designing effective ChIP experiments with this antibody requires careful consideration of several factors:
Crosslinking optimization: Standard 1% formaldehyde for 10 minutes at room temperature works well for histone modifications, but optimization may be required for specific cell types.
Sonication parameters: Aim for chromatin fragments between 200-500 bp for optimal resolution. Verify fragment size by agarose gel electrophoresis.
Antibody amount: Use 5 μg of antibody per ChIP reaction with 4×10^6 cells as a starting point .
Controls: Always include:
Input control (pre-immunoprecipitation chromatin)
IgG negative control (same species as the primary antibody)
Positive control (antibody against abundant histone mark like H3K4me3)
Quantification: Use real-time PCR for targeted analysis or next-generation sequencing (ChIP-seq) for genome-wide profiling.
Data analysis: For ChIP-seq, compare enrichment patterns of H4K5cr with other histone marks (H4K5ac, H3K27ac) to understand the functional significance of this modification .
For optimal immunofluorescence results:
Fixation: Use 4% paraformaldehyde for 10 minutes at room temperature, followed by permeabilization with 0.2% Triton X-100.
Antigen retrieval: Perform mild heat-induced epitope retrieval using 10 mM sodium citrate buffer (pH 6.0) to improve accessibility of nuclear antigens.
Blocking: Use 5% BSA with 0.1% Triton X-100 in PBS for 1 hour at room temperature.
Primary antibody incubation: Dilute the Crotonyl-HIST1H4A (K5) Antibody 1:100 in blocking buffer and incubate overnight at 4°C.
Secondary antibody: Use fluorophore-conjugated secondary antibodies (e.g., Alexa Fluor 488) at 1:500 dilution for 1 hour at room temperature.
Counterstaining: DAPI (1 μg/mL) for nuclear visualization.
Mounting: Use anti-fade mounting medium to preserve fluorescence signal.
Imaging: Confocal microscopy is recommended for detailed nuclear localization patterns .
Validating antibody specificity is crucial for reliable interpretation of experimental results:
Peptide competition assay: Pre-incubate the antibody with the crotonylated H4K5 peptide immunogen before application to your sample. This should abolish specific signals.
Recombinant protein testing: Test the antibody against a panel of recombinant histones with different modifications (acetylation, methylation, crotonylation) at the K5 position and other positions to confirm specificity .
Genetic validation: Use CRISPR/Cas9 to create cell lines with K5R mutation in histone H4 genes, which would prevent crotonylation at this position.
Enzyme treatment: Treat samples with decrotonylases (such as certain sirtuins) and observe signal reduction.
Cross-reactivity assessment: Test against samples containing other acylations (acetylation, butyrylation) to ensure the antibody doesn't cross-react with similar modifications .
Potential cross-reactivity concerns include:
Similar acyl modifications: Some antibody preparations may show cross-reactivity with other acyl modifications at the same position (H4K5ac, H4K5bu), particularly if the immunizing peptide design didn't adequately distinguish these modifications.
Neighboring modifications: The antibody specificity may be affected by modifications on neighboring residues. For example, some H4K5cr antibodies may have reduced binding if K8 is also modified .
Isoform specificity: While the antibody targets HIST1H4A, the high sequence conservation among H4 variants means the antibody will recognize crotonylation at position K5 in all H4 protein variants (HIST1H4A-L, HIST2H4, HIST4H4) .
Species cross-reactivity: The antibody raised against human H4K5cr typically works well with mouse and rat samples due to high sequence conservation, but validation is recommended when using with other species .
Investigating crotonylation dynamics throughout the cell cycle:
Synchronization methods: Use double thymidine block, nocodazole treatment, or mitotic shake-off to obtain cell populations at specific cell cycle stages.
Multi-parameter flow cytometry: Combine propidium iodide staining for DNA content with intracellular H4K5cr antibody staining to correlate crotonylation levels with cell cycle phases.
Time-course experiments: After synchronization release, collect cells at regular intervals (e.g., every 2 hours) and analyze H4K5cr levels by Western blot and immunofluorescence.
ChIP-seq at different cell cycle stages: Perform ChIP-seq using the Crotonyl-HIST1H4A (K5) Antibody on synchronized populations to map genome-wide changes in crotonylation patterns.
Comparison with other modifications: Parallel analysis of H4K5ac and H4K5cr can reveal whether these modifications show similar or distinct patterns during cell cycle progression, building on previous findings regarding H4 acetylation dynamics .
To investigate the functional relationship between H4K5 crotonylation and gene expression:
Integrated genomic analysis: Combine ChIP-seq data for H4K5cr with RNA-seq data from the same biological sample to correlate genomic localization with gene expression levels.
Sequential ChIP (Re-ChIP): Perform sequential immunoprecipitation with H4K5cr antibody followed by antibodies against transcriptional machinery components (RNA Pol II, transcription factors) to identify co-occupancy.
CRISPR-based approaches:
dCas9-fusion of crotonyl readers or erasers to specific genomic loci
Mutation of H4K5 to arginine to prevent crotonylation
Targeting crotonylation regulators (writers, erasers) to study effects on target genes
Global manipulation of crotonylation: Modulate cellular crotonyl-CoA levels through metabolic interventions or enzyme inhibition, then assess genome-wide H4K5cr patterns and corresponding transcriptional changes.
Single-cell approaches: Combine immunofluorescence for H4K5cr with RNA-FISH for specific transcripts to correlate modification status with transcriptional output at the single-cell level .
The genetic code expansion approach offers powerful validation strategies:
Expression system development: Utilize the evolved Mb-PylRS/Pyl-tRNA pair system that has been optimized for site-specific incorporation of ε-N-crotonyllysine (Kcr) into proteins in both E. coli and mammalian cells .
Recombinant protein production: Generate recombinant H4 proteins with:
Site-specific Kcr at position 5 only
Kcr at alternative positions (e.g., K8, K12, K16)
Combinations of Kcr and other modifications
Antibody validation: Use these defined protein standards to:
Assess binding affinity and specificity of the antibody
Determine minimum detection limits
Evaluate cross-reactivity with other acyl modifications
Functional studies: Introduce these site-specifically modified histones into nucleosome reconstitution systems or cell-free transcription assays to determine the direct functional impact of K5 crotonylation.
Western blot standards: Use defined amounts of these recombinant proteins as quantitative standards for estimating relative H4K5cr levels in biological samples .
When encountering signal issues with H4K5cr antibody detection:
Sample preparation optimization:
Ensure complete cell lysis and histone extraction
Add HDAC/decrotonylase inhibitors (sodium butyrate, TSA, nicotinamide) to buffers
Avoid freeze-thaw cycles of extracted histones
Antibody-specific adjustments:
Titrate antibody concentration (try 1:500, 1:1000, 1:2000)
Extend primary antibody incubation time (overnight at 4°C)
Try different blocking agents (BSA vs. casein)
Signal enhancement strategies:
Increase sample loading (up to 30 μg for total cell lysate)
Use high-sensitivity ECL substrates for Western blot
Try biotin-streptavidin amplification for immunostaining
Biological considerations:
When facing inconsistent results across different techniques:
Technique-specific considerations:
Western blot detects denatured proteins, while IF/IP work with native conformations
ChIP efficiency depends on chromatin accessibility and crosslinking efficiency
Different applications have distinct sensitivity thresholds
Systematic validation approach:
Use positive control samples known to contain H4K5cr for all techniques
Compare multiple antibody sources/lots across techniques
Perform peptide competition assays in each technique separately
Technical refinement:
Optimize fixation/extraction protocols for each technique
Consider epitope masking in different contexts
Evaluate buffer compatibility with maintaining the crotonyl modification
Orthogonal validation:
To maintain antibody performance over time:
Storage conditions:
Working solution handling:
Keep on ice during experiments
Return to 4°C promptly after use
Do not leave at room temperature for extended periods
Stability considerations:
Buffer compatibility:
Adapting H4K5cr antibodies for single-cell applications:
Single-cell CUT&Tag/CUT&RUN:
Protocol optimization for using Crotonyl-HIST1H4A (K5) Antibody in microfluidic-based single-cell platforms
Integration with single-cell RNA-seq to correlate H4K5cr patterns with gene expression at single-cell resolution
Computational approaches for handling sparse data typical of single-cell epigenomic profiles
Mass cytometry (CyTOF) applications:
Metal-conjugated H4K5cr antibodies for high-parameter analysis
Multiplexing with other histone modification antibodies and cellular markers
Clustering algorithms to identify cell subpopulations with distinct H4K5cr signatures
In situ visualization:
Combining H4K5cr immunofluorescence with RNA-FISH for spatial context
Development of proximity ligation assays (PLA) to detect H4K5cr co-occurrence with reader proteins
Super-resolution microscopy techniques for detailed nuclear localization of H4K5cr domains
Technical challenges:
Understanding H4K5cr in the complex landscape of histone modifications:
Modification crosstalk patterns:
H4K5cr may have antagonistic relationships with H4K5 methylation
Potential synergistic or sequential relationships with other nearby modifications (H4K8ac, H4K12ac)
Undetermined relationships with modifications on other histones (H3K27ac, H3K4me3)
Research approaches:
Sequential ChIP to identify co-occurrence with other modifications
Mass spectrometry of histone peptides to quantify combinatorial modification patterns
Reconstituted designer nucleosomes with specific modification combinations
Reader protein interactions:
Identify proteins that specifically recognize H4K5cr versus H4K5ac
Determine how neighboring modifications affect reader protein binding
Investigate whether H4K5cr creates unique protein interaction surfaces
Functional implications:
The intersection of metabolism and histone crotonylation:
Crotonyl-CoA metabolism regulation:
Fatty acid oxidation produces crotonyl-CoA as an intermediate
Manipulation of β-oxidation pathways can alter cellular crotonyl-CoA pools
Short-chain fatty acid supplementation (particularly crotonate) can increase histone crotonylation
Experimental approaches:
Metabolic interventions (glucose deprivation, fatty acid supplementation) followed by H4K5cr ChIP-seq
Isotope tracing to track the incorporation of labeled precursors into histone crotonylation
Genetic manipulation of key metabolic enzymes (ACADS, HADH) to alter crotonyl-CoA levels
Physiological contexts:
Changes in crotonylation during cellular stress responses
Tissue-specific patterns of crotonylation reflecting metabolic specialization
Altered crotonylation in metabolic diseases (obesity, diabetes, cancer)
Technical considerations: