Target: β-hydroxybutyryl-HIST1H3A (K18)
Host Species: Rabbit
Isotype: IgG
Applications:
Immunogen: Synthetic peptide corresponding to residues surrounding β-hydroxybutyryl-K18 on human HIST1H3A .
Cross-reactivity: Confirmed in human, mouse, and rat models .
Distinct from acetylation: Unlike acetylation, β-hydroxybutyrylation introduces a larger (+86.0368 Da) and structurally distinct modification, confirmed via MS/MS .
Dose-dependent response: Antibody signals increase proportionally with β-hydroxybutyrate (BHB) concentration in cell cultures .
Cross-reactivity challenges:
Induction: Elevated during fasting, diabetes, or BHB treatment due to increased BHB-CoA, a substrate for histone acyltransferases like p300 .
Gene regulation: Associates with promoters of PGC-1α and FOXO1, enhancing oxidative metabolism .
Modification | Co-occurring Marks | Functional Outcome |
---|---|---|
H3K18bhb | H3K4me3, H3K9ac | Enhanced transcriptional elongation |
H3K18bhb | H4K8bhb | Chromatin decompaction in starvation |
Diabetes: Liver tissues from streptozotocin (STZ)-induced diabetic mice show 3.5-fold higher H3K18bhb levels vs. controls .
Cancer: Reduced H3K18bhb correlates with metabolic reprogramming in hepatocellular carcinoma (HepG2) .
ChIP-seq: Maps genome-wide H3K18bhb distribution, revealing enrichment at metabolic genes .
Multiplex imaging: Combined with pH3S10, identifies cell-cycle-dependent β-hydroxybutyrylation .
Antibody validation: Critical due to structural similarity between acylations (e.g., β-hydroxybutyryl vs. 2-hydroxyisobutyryl) . Recommendations:
Dynamic regulation: H3K18bhb levels fluctuate rapidly with nutrient status, requiring strict sample collection protocols .
Histone lysine β-hydroxybutyrylation (Kbhb) is a novel post-translational modification (PTM) derived from β-hydroxybutyrate (BHB), a primary ketone body. Unlike better-characterized modifications such as acetylation or methylation, Kbhb specifically responds to cellular metabolic states, particularly during carbohydrate restriction or elevated BHB levels. Structurally, β-hydroxybutyrylation involves the addition of a β-hydroxybutyryl group (containing a hydroxyl group at the beta position) to lysine residues on histones, creating a unique molecular signature with distinct regulatory potential .
While acetylation primarily neutralizes the positive charge of lysine residues to alter chromatin structure, β-hydroxybutyrylation introduces both charge neutralization and additional structural properties through its hydroxyl group. These distinctive biochemical characteristics may explain why Kbhb demonstrates different regulatory functions from histone acetylation and methylation, particularly in metabolic stress responses . To experimentally distinguish between these modifications, researchers must employ modification-specific antibodies and/or mass spectrometry-based proteomics approaches.
Current research has identified 44 histone Kbhb sites across the proteome, a number comparable to known histone acetylation sites . The most extensively characterized modification sites include:
Histone | Key Modification Sites | Associated Functions |
---|---|---|
H3 | K4, K9, K18, K27, K56 | Gene activation, metabolic regulation |
H4 | K8, K12 | Chromatin structure regulation |
H3K18bhb serves as a particularly important site in metabolic regulation, with documented roles in starvation-responsive gene expression . Studies have shown that H3K9bhb, H3K18bhb, H4K8bhb, and H3K4bhb levels increase in a β-hydroxybutyrate dose-dependent manner, suggesting site-specific sensitivity to metabolic fluctuations . Researchers investigating metabolic signaling through chromatin should prioritize these sites in their experimental designs.
For optimal ChIP results with β-hydroxybutyryl-HIST1H3A (K18) antibodies, researchers should implement the following methodological considerations:
Cross-linking optimization: Use 1% formaldehyde for 10 minutes at room temperature, as excessive cross-linking can mask the β-hydroxybutyrylated epitope.
Sonication parameters: Adjust sonication conditions to generate chromatin fragments of 200-500 bp, which is optimal for histone modification ChIP.
Antibody concentration: For β-hydroxybutyryl-HIST1H3A (K18) antibodies, use a 1:50 dilution for ChIP applications, which typically corresponds to 2-4 μg of antibody per ChIP reaction .
Validation controls: Always include:
Input control (non-immunoprecipitated chromatin)
IgG negative control
Positive control targeting abundant histone marks (e.g., H3K4me3)
Spike-in controls for quantitative analyses
Washing stringency: For β-hydroxybutyrylation ChIP, include an additional high-salt wash (500 mM NaCl) to reduce background without compromising specific signal.
When performing subsequent qPCR analysis, design primers for genomic regions known to be enriched in H3K18bhb marks, particularly starvation-responsive gene promoters, to serve as positive controls .
For effective immunofluorescence detection of β-hydroxybutyryl-HIST1H3A (K18), researchers should follow this optimized protocol:
Cell preparation: Fix cells with 4% paraformaldehyde for 15 minutes at room temperature, followed by permeabilization with 0.2% Triton X-100 for 10 minutes.
Antigen retrieval: Perform mild antigen retrieval using 10 mM sodium citrate buffer (pH 6.0) at 95°C for 10 minutes to enhance epitope accessibility.
Blocking: Block with 5% BSA in PBS for 1 hour at room temperature to minimize non-specific binding.
Primary antibody incubation: Dilute β-hydroxybutyryl-HIST1H3A (K18) antibody at 1:20-1:200 in blocking buffer and incubate overnight at 4°C .
Secondary antibody: Use appropriate fluorophore-conjugated secondary antibodies (anti-rabbit IgG) at 1:500 dilution for 1 hour at room temperature.
Nuclear counterstaining: Counterstain with DAPI (1 μg/ml) for 5 minutes.
Controls: Include parallel samples with:
Primary antibody omission
Peptide competition (using the immunizing peptide)
Dual staining with other histone marks for colocalization studies
For quantitative IF analysis, acquire images using identical exposure settings and analyze signal intensity within the nuclear compartment using appropriate image analysis software.
Distinguishing between β-hydroxybutyrylation and other acyl modifications requires a multi-faceted analytical approach:
Antibody specificity validation: Verify antibody specificity through:
Dot blot assays with modified and unmodified peptides
Competition experiments with structurally related modified peptides
Western blot analysis following HDAC treatment
Mass spectrometry differentiation:
Use targeted MS/MS approaches with specific diagnostic fragment ions
Employ high-resolution MS to distinguish between isobaric modifications
Implement stable isotope labeling with isotopic β-hydroxybutyrate (e.g., [13C2]) to definitively identify β-hydroxybutyrylated peptides through mass shift detection
Chiral analysis: Consider the chirality of β-hydroxybutyrylation, as R-β-hydroxybutyrylation and S-β-hydroxybutyrylation exhibit different interactions with deacetylases like HDAC3 and SIRT3 .
Enzymatic specificity tests: Utilize the differential sensitivity of modifications to specific erasers (HDACs and SIRTs) as HDAC1-3 and SIRT1-3 have been identified to have de-β-hydroxybutyrylation activity, though with varying specificities .
This comprehensive approach enables researchers to definitively identify β-hydroxybutyrylation against the complex background of cellular acylation patterns, avoiding misattribution of biological functions.
To investigate the dynamic relationship between histone β-hydroxybutyrylation and metabolic signaling, researchers should implement these advanced approaches:
Metabolic manipulation models:
Integrated multi-omics:
Parallel ChIP-seq of H3K18bhb and other histone marks
RNA-seq to correlate histone modifications with transcriptional outputs
Metabolomics to measure BHB and related metabolite levels
Proteomics to assess global protein β-hydroxybutyrylation
Temporal resolution studies:
Time-course experiments following metabolic perturbation
Pulse-chase labeling with isotopic BHB to determine modification turnover rates
Enzymatic manipulation:
Such integrated approaches allow researchers to establish causal relationships between metabolic states, histone β-hydroxybutyrylation patterns, and downstream gene expression changes, particularly in starvation-responsive metabolic pathways where H3K18bhb has demonstrated regulatory roles .
For optimal detection, researchers should:
Use fresh histones extracted with acidic extraction methods
Run 15-18% SDS-PAGE gels for better resolution of histone proteins
Transfer to PVDF membranes (rather than nitrocellulose) at 30V overnight at 4°C
Use recommended dilutions (1:100-1:1000) for Western blot applications
Include parallel blots for total H3 as loading controls
For accurate quantification of histone β-hydroxybutyrylation changes, researchers should implement these methodological approaches:
Western blot quantification:
Use infrared fluorescence-based detection systems rather than chemiluminescence
Normalize H3K18bhb signal to total H3 signal from the same membrane (after stripping) or parallel membrane
Include standard curves with known quantities of modified peptides
Apply appropriate statistical analyses (minimum of three biological replicates)
Mass spectrometry-based quantification:
Implement label-free quantification of modified peptides with appropriate normalization
Use SILAC or TMT labeling for more accurate comparative analysis
Monitor multiple β-hydroxybutyrylated peptides, not just H3K18bhb
Calculate modification stoichiometry (percentage of a site carrying the modification)
ChIP-seq quantification:
Use spike-in normalization with exogenous chromatin
Apply appropriate normalization methods for between-sample comparisons
Calculate differential binding metrics rather than raw peaks
Correlate with gene expression changes for functional significance
Experimental design considerations:
Include time-course measurements to capture dynamic changes
Use appropriate metabolic controls (e.g., acetate treatment, glucose manipulation)
Include both technical and biological replicates
Implement multiple independent methods for cross-validation
Proper quantification is essential as histone β-hydroxybutyrylation levels change in response to β-hydroxybutyrate concentration in a dose-dependent manner, with H3K9bhb, H3K18bhb, H4K8bhb, and H3K4bhb all showing differential sensitivity to metabolic fluctuations .
To explore the connections between histone β-hydroxybutyrylation and disease states, researchers should consider these advanced investigative approaches:
Clinical sample analysis:
Compare β-hydroxybutyrylation patterns in patient-derived tissues and matched controls
Correlate H3K18bhb levels with clinical parameters and disease progression
Develop tissue microarray analyses with β-hydroxybutyrylation antibodies
Disease model integration:
Examine β-hydroxybutyrylation in established disease models of metabolic disorders, cardiovascular diseases, kidney diseases, and cancer
Create genetic models with altered β-hydroxybutyrylation machinery (p300 mutants, HDAC mutants)
Implement diet and pharmacological interventions that modulate BHB levels
Therapeutic targeting strategies:
Screen for specific inhibitors/activators of histone β-hydroxybutyrylation
Investigate BHB precursors as potential epigenetic therapeutics
Explore combination approaches targeting multiple epigenetic marks
Mechanistic investigations:
Identify disease-specific β-hydroxybutyrylation regulatory networks
Determine the interplay between β-hydroxybutyrylation and inflammation pathways
Explore β-hydroxybutyrylation in cellular stress responses relevant to disease
This research is particularly promising given that histone Kbhb has been associated with the pathogenesis of metabolic cardiovascular diseases, kidney diseases, tumors, neuropsychiatric disorders, and metabolic diseases with functions distinct from histone acetylation and methylation .
To identify and characterize the enzymatic machinery regulating histone β-hydroxybutyrylation, researchers should implement these specialized approaches:
Enzyme activity profiling:
In vitro assays with recombinant enzymes and isotope-labeled BHB-CoA
Test known acyltransferases (particularly p300) for β-hydroxybutyrylation activity
Screen HDAC and SIRT family members for de-β-hydroxybutyrylation activity
Investigate enzyme kinetics and substrate preferences
Protein-protein interaction studies:
Perform BioID or proximity labeling with potential writer/eraser enzymes
Use co-immunoprecipitation to identify protein complexes involved in β-hydroxybutyrylation
Implement FRET-based approaches to detect dynamic enzyme-substrate interactions
Genetic screening:
Conduct CRISPR screens targeting epigenetic regulators and assess β-hydroxybutyrylation levels
Generate knockout/knockdown models of candidate enzymes
Perform rescue experiments with wild-type and catalytically dead enzyme variants
Structural biology approaches:
Determine crystal structures of enzymes in complex with β-hydroxybutyrylated peptides
Investigate binding pockets and catalytic mechanisms
Explore the structural basis for chiral preferences (R- vs. S-β-hydroxybutyrylation)
Current research has identified p300 as a histone Kbhb "writer" that catalyzes the addition of BHB to lysine residues, with particular activity at H3K9, H3K18, H3K27, and H4K8 sites . For erasers, HDAC1-3 and SIRT1-3 have demonstrated de-β-hydroxybutyrylation activity in vitro, with HDAC1 and HDAC2 functioning as primary deacylases in cellular contexts . The differential activity of these enzymes on R- versus S-β-hydroxybutyrylation highlights the complexity of this regulatory system and presents opportunities for targeted manipulation.
For high-quality ChIP-seq data with β-hydroxybutyryl-HIST1H3A (K18) antibodies, researchers should address these critical experimental parameters:
Antibody validation for ChIP-seq:
Confirm antibody specificity via Western blot and peptide competition
Perform preliminary ChIP-qPCR at known target regions before sequencing
Validate antibody lot-to-lot consistency with standard samples
Chromatin preparation optimization:
Use dual cross-linking (formaldehyde followed by EGS) for enhanced capture
Optimize sonication for consistent fragment size distribution (200-400 bp)
Implement stringent quality control of chromatin (fragment analysis, DNA concentration)
Library preparation considerations:
Use ChIP-seq optimized library preparation kits with minimal PCR cycles
Include appropriate controls (input, IgG, spike-in)
Implement unique molecular identifiers (UMIs) to control for PCR duplication
Bioinformatic analysis pipeline:
Use peak callers optimized for histone modifications (e.g., MACS2 with broad peak settings)
Implement robust normalization methods
Perform integrative analysis with RNA-seq and other epigenomic data
Consider differential binding analysis rather than binary peak calling
ChIP-seq studies have demonstrated that histone Kbhb is enriched in active gene promoters, particularly those associated with starvation-responsive metabolic pathways . When designing ChIP-seq experiments, researchers should consider metabolic perturbations that elevate cellular β-hydroxybutyrate levels to enhance the detection of differential binding patterns.
For multiplexed flow cytometry or mass cytometry (CyTOF) applications with β-hydroxybutyryl-HIST1H3A (K18) antibodies, implement these specialized protocols:
Sample preparation optimization:
Use gentle fixation (1% formaldehyde, 10 minutes) followed by methanol permeabilization
Implement specialized nuclear preparation kits for consistent results
Include RNase treatment to reduce background
Antibody labeling strategies:
For flow cytometry: Use bright fluorophores (Alexa Fluor 488 or PE) for histone modifications
For CyTOF: Label with rare earth metals with minimal signal overlap
Titrate antibody concentrations carefully to optimize signal-to-noise ratio
Panel design considerations:
Include markers for cell cycle phases (e.g., Ki-67, PCNA)
Add other histone modifications for correlation analysis
Include metabolic state markers when possible
Analysis approaches:
Implement dimensionality reduction techniques (tSNE, UMAP)
Use advanced clustering algorithms to identify cell populations with distinct modification patterns
Consider trajectory analysis for dynamic processes
Validation requirements:
Confirm patterns with imaging flow cytometry
Validate with orthogonal techniques (Western blot, ChIP)
Include appropriate biological controls (metabolic perturbations)
This approach enables single-cell resolution analysis of β-hydroxybutyrylation dynamics and heterogeneity within complex populations, revealing cell state-specific regulation that may be obscured in bulk analyses.