Crotonyl-HIST1H4A (K12) Antibody is a polyclonal antibody specifically designed to detect histone H4 crotonylation at lysine 12 (H4K12cr), a post-translational modification (PTM) associated with transcriptional regulation and chromatin dynamics . This antibody is generated using a synthetic peptide immunogen derived from the crotonyl-lysine 12 region of human histone H4, ensuring high specificity for this modification .
Crotonylation is a conserved histone modification that neutralizes lysine’s positive charge, destabilizing nucleosome-DNA interactions and promoting transcriptional activation . Key findings include:
Functional Parallels to Acetylation: Like acetylation, crotonylation is enriched at transcription start sites (TSS) and enhancers, but it exhibits distinct regulatory roles due to its four-carbon planar structure .
Enzymatic Regulation: The Piccolo NuA4 complex (containing Esa1) catalyzes H4K12cr in vitro, linking this modification to chromatin remodeling and gene expression .
Role in Disease: Dysregulated crotonylation is implicated in cancer, inflammation, and stem cell differentiation, with H4K12cr serving as a biomarker for active chromatin states .
Specificity: The antibody does not cross-react with acetylated, butyrylated, or propionylated histone H4 at K12, as confirmed by peptide competition assays .
Structural Insights: The crotonyl group’s planar structure is recognized via hydrophobic interactions in the antibody’s binding pocket, ensuring selective detection .
Functional Assays: Used in chromatin immunoprecipitation (ChIP) to map H4K12cr distribution, revealing enrichment at promoters of genes involved in metabolism and inflammation .
Chromatin Dynamics: Identifies H4K12cr in studies exploring nucleosome destabilization during spermatogenesis or DNA repair .
Gene Expression Profiling: Detects crotonylation-mediated transcriptional activation in macrophages exposed to lipopolysaccharides (LPS) .
Comparative Epigenetics: Distinguishes crotonylation from acetylation in histone turnover assays, aiding mechanistic studies of PTM crosstalk .
Species Restriction: Primarily validated for human samples; cross-reactivity with mouse or rat tissues requires additional verification .
Buffer Compatibility: Contains glycerol and sodium azide, which may interfere with certain downstream assays .
Custom Production: Lead times of 3–4 weeks are typical, and orders are non-refundable once in production .
Recent studies highlight the potential of H4K12cr as a therapeutic target in diseases driven by epigenetic dysregulation, such as leukemia and neurodegenerative disorders . Advances in super-resolution microscopy and single-cell sequencing are expected to enhance spatial mapping of this modification in heterogeneous cell populations.
Thorough validation of antibody specificity is essential for reliable experimental results. For Crotonyl-HIST1H4A (K12) antibodies, implement the following controls:
Positive Controls:
Use cell lines or tissues known to express high levels of crotonylated H4K12 (e.g., testis tissue for many histone crotonylation marks)
Include crotonylation-inducing treatments (e.g., exposure to crotonate or inhibition of class I HDACs)
Negative Controls:
Utilize cells where the crotonylation writer enzyme has been knocked down
Include crotonyl-specific eraser enzyme overexpression (e.g., HDAC overexpression)
Apply competing peptide blocks with crotonylated H4K12 peptides
Specificity Tests:
Perform dot blot analysis with modified peptides (crotonylated H4K12 vs. other modifications like acetylated H4K12)
Conduct western blot analysis following CRISPR/Cas9-mediated K12R mutation in H4
Use mass spectrometry validation to confirm antibody pull-down specificity
A comprehensive validation strategy increases confidence in experimental findings and helps distinguish true signals from potential cross-reactivity with similar modifications .
Distinguishing between these similar modifications requires strategic experimental design:
Sequential Immunoprecipitation: First immunoprecipitate with one antibody (e.g., acetyl-H4K12), then perform a second immunoprecipitation on the unbound fraction with the other antibody (crotonyl-H4K12)
Mass Spectrometry Analysis: Use targeted MS approaches to distinguish between the mass differences of acetyl (+42 Da) and crotonyl (+68 Da) modifications
Enzyme Sensitivity Tests: Treat samples with recombinant deacetylases or decrotonylases with known specificities, then observe differential removal patterns
Modification-Specific Reader Proteins: Employ known reader proteins that specifically bind to either crotonylated or acetylated lysines as detection tools
Metabolic Labeling: Use isotopically labeled precursors for acetyl-CoA versus crotonyl-CoA to track modification incorporation
By combining these approaches, researchers can confidently discriminate between these closely related but functionally distinct histone modifications, enabling more accurate interpretation of their biological significance .
Crotonyl-HIST1H4A (K12) antibodies may exhibit cross-reactivity with:
Similar Modifications: Acetylation, butyrylation, or other acylations at the same position
Similar Epitopes: Crotonylation at other histone lysine positions with similar surrounding sequences
Non-Histone Proteins: Crotonylated lysines in non-histone proteins with sequence similarities
Strategies to Address Cross-Reactivity:
Pre-absorption: Incubate antibodies with competing peptides containing similar modifications to block cross-reactive antibodies
Sequential Dilution Testing: Perform titration experiments to identify dilution points where specific binding is maintained but cross-reactivity is minimized
Knockout Validation: Use cells/tissues where the specific histone variant is knocked out as negative controls
Modification-Specific Enzymes: Employ enzymes that selectively remove only crotonylation to confirm that signal loss correlates with modification removal
Peptide Array Screening: Test antibody specificity against arrays containing various histone modifications to quantify cross-reactivity
Careful validation using these approaches can significantly improve experimental reliability and interpretation of results involving histone crotonylation at H4K12 .
H4K12 crotonylation exhibits distinct patterns across the cell cycle, reflecting its functional role in chromatin regulation during different cellular processes:
| Cell Cycle Phase | H4K12 Crotonylation Pattern | Experimental Approach |
|---|---|---|
| G1 | Moderate levels, associated with early-response genes | Serum starvation followed by release |
| S | Increased levels at replication forks | Thymidine block and release, EdU labeling |
| G2 | Redistribution to specific genomic regions | RO-3306 (CDK1 inhibitor) treatment |
| M | Generally decreased, with enrichment at centromeres | Nocodazole or colcemid treatment |
Research Methods:
Cell Synchronization: Use chemical synchronization methods followed by ChIP-seq or immunofluorescence with Crotonyl-HIST1H4A (K12) antibodies
FACS-ChIP: Sort cells by DNA content (cell cycle stage) before performing ChIP with the antibody
Live-Cell Imaging: Use cell-permeable crotonylation probes in combination with cell cycle markers
Sequential ChIP: Combine cell cycle marker proteins with H4K12cr antibodies
Single-Cell Analysis: Perform single-cell sequencing approaches with crotonylation detection to observe cell-to-cell variation
These approaches can reveal how H4K12 crotonylation contributes to cell cycle-specific gene expression programs and chromatin organization changes .
Researchers can employ various techniques to quantify H4K12 crotonylation at different scales:
Global Quantification:
Mass Spectrometry: Provides precise quantification of modification stoichiometry
Use targeted MRM (Multiple Reaction Monitoring) for specific detection of H4K12cr
Apply SILAC or TMT labeling for comparative analysis across conditions
ELISA-Based Assays: Allow high-throughput screening of global changes
Develop sandwich ELISA with Crotonyl-HIST1H4A (K12) antibodies
Employ automated liquid handling for large-scale screens
Western Blot: Semi-quantitative approach for relative abundance measurement
Use fluorescent secondary antibodies for wider dynamic range
Apply normalization to total H4 for accurate comparison
Locus-Specific Quantification:
ChIP-qPCR: Targeted analysis of specific genomic regions
Design primers for regions of interest (promoters, enhancers)
Use spike-in controls for normalization across samples
ChIP-seq: Genome-wide profiling of H4K12cr distribution
Apply peak calling algorithms optimized for histone modifications
Consider ChIP-rx for quantitative comparisons between samples
CUT&RUN or CUT&Tag: Higher resolution mapping with lower background
Provides improved signal-to-noise ratio in low-abundance modifications
Requires fewer cells than traditional ChIP approaches
Combining global and locus-specific approaches provides comprehensive insights into the functional impact of H4K12 crotonylation changes across different experimental conditions .
Multi-omics integration creates more comprehensive understanding of H4K12 crotonylation function:
ChIP-seq + RNA-seq Integration:
Correlate H4K12cr distribution with gene expression patterns
Identify direct transcriptional targets of H4K12cr
Apply mathematical models to infer causality between modification and expression
ChIP-seq + ATAC-seq:
Associate H4K12cr with chromatin accessibility changes
Identify pioneer factor binding sites co-occurring with H4K12cr
Map the relationship between crotonylation and nucleosome positioning
Proteomics + ChIP-seq:
Identify reader proteins that specifically bind H4K12cr using SILAC pull-downs
Correlate protein complex formation with genomic distribution of H4K12cr
Map the entire "crotonylation interactome" in specific cell types
Metabolomics + ChIP-seq:
Investigate how metabolic pathways influence H4K12cr levels
Track isotopically labeled metabolites to crotonylation sites
Determine rate-limiting steps in crotonylation metabolism
Single-cell Multi-omics:
Perform joint profiling of H4K12cr, chromatin accessibility, and transcription
Identify cell state-specific relationships between crotonylation and gene expression
Map modification dynamics during cellular differentiation or response
Computational integration of these multi-omics datasets requires specialized bioinformatics pipelines, including machine learning approaches and network analysis methods, to derive meaningful biological insights from the complex interrelationships between different data types .