The antibody recognizes β-hydroxybutyryl-lysine (Kbhb) at position 122 of histone H3.1 (UniProt ID: P68431) . Key characteristics include:
Application | Recommended Dilution | Key Limitations |
---|---|---|
WB | 1:100–1:1000 | Potential false positives under HDAC inhibition |
ICC | 1:10–1:100 | Requires corroboration with MS data |
ELISA | 1:2000–1:10000 | High sensitivity but low specificity |
Functional role: β-hydroxybutyrylation at H3K122 is implicated in metabolic-epigenetic crosstalk, potentially modulating gene promoters via readers like ENL .
Technical challenges: Antibody specificity remains a critical issue, necessitating orthogonal validation methods (e.g., peptide arrays or CRISPR-edited cell lines) .
Lysine β-hydroxybutyrylation (Kbhb) represents a relatively new histone post-translational modification identified in 2016. This modification involves the addition of a β-hydroxybutyryl group to lysine residues, creating a nuanced layer of regulation in cellular functions . Specifically for histone research, Kbhb is enriched in active gene promoters and has been found to modify numerous lysine residues across all four types of histones (H1, H2A, H2B, H3, H4) . Understanding this modification provides insight into epigenetic regulation mechanisms and potentially connects metabolic status with gene expression patterns.
The β-hydroxybutyryl-HIST1H3A (K122) polyclonal antibody has been specifically validated for several key applications in molecular biology research:
These validated applications make this antibody a versatile tool for detecting and studying β-hydroxybutyrylation at the K122 position of human Histone H3.1 across multiple experimental contexts, from protein quantification to cellular localization studies.
The β-hydroxybutyryl-HIST1H3A (K122) polyclonal antibody specifically recognizes human histone H3.1 that has been β-hydroxybutyrylated at the lysine 122 position . The antibody was generated using an immunogen consisting of a peptide sequence surrounding the β-hydroxybutyryl-Lys (122) site derived from Human Histone H3.1 (accession number P68431) . This site-specific antibody enables researchers to distinguish this particular modification from other post-translational modifications that may occur on histone H3.1.
Verifying antibody specificity is crucial for accurate interpretation of results, especially given the documented concerns about cross-reactivity with some Kbhb antibodies . A comprehensive approach to verify specificity includes:
Treatment comparison tests: Treat cells with β-hydroxybutyrate (BHB), structurally similar compounds (like butyrate), and histone deacetylase inhibitors (e.g., Trichostatin A). Properly specific antibodies should show strong signals only in BHB-treated samples .
Immunoprecipitation followed by mass spectrometry:
Peptide competition assays: Pre-incubate the antibody with β-hydroxybutyrylated peptides versus unmodified or alternatively modified peptides before immunoblotting or immunoprecipitation to test for signal blocking.
When analyzing results, consider that true Kbhb-specific antibodies should not cross-react with non-modified lysine residues, unmodified peptides, 2-hydroxyisobutyrylated peptides, or acetylated peptides .
For optimal detection of β-hydroxybutyryl-HIST1H3A (K122) modifications, follow this detailed protocol:
Cell/tissue preparation:
Histone extraction protocol:
Lyse cells in Triton Extraction Buffer (PBS containing 0.5% Triton X-100, 2 mM PMSF, 0.02% NaN₃)
Centrifuge at 6,500 x g for 10 minutes at 4°C
For histone-specific extraction, resuspend the nuclear pellet in 0.2 N HCl overnight at 4°C
Centrifuge at 6,500 x g for 10 minutes
Neutralize the supernatant with 20% NaOH
Sample preparation for Western blotting:
Determine protein concentration using the DC Protein Assay Kit
Mix sample with 4x Laemmli buffer/10% beta-mercaptoethanol
Heat at 95°C for 5 minutes
Resolve proteins by SDS-PAGE and transfer to nitrocellulose membrane
Block with 5% milk in TBS-T for 30-60 minutes
Incubate with β-hydroxybutyryl-HIST1H3A (K122) antibody at the recommended dilution
Including deacetylase inhibitors and β-hydroxybutyrylation-preserving reagents in your buffers can help maintain modification integrity throughout the extraction process.
Distinguishing true β-hydroxybutyrylation signals from cross-reactivity requires a multi-faceted approach:
Control experiments:
Mass spectrometry validation:
Cross-comparison with multiple antibodies:
Based on published research, caution is particularly warranted with H3K9bhb antibodies, which have shown cross-reactivity with acetylation and potentially other modifications .
Multiple methodologies exist for studying β-hydroxybutyrylation patterns, each with distinct advantages:
Methodology | Advantages | Limitations | Best For |
---|---|---|---|
Western Blotting with Kbhb-specific antibodies | - Relatively simple technique - Quantitative assessment - Protein size determination | - Limited to known modifications - Possible cross-reactivity - No spatial information | - Confirming presence of modification - Relative quantification - Monitoring changes across conditions |
Immunoprecipitation + Mass Spectrometry | - Identifies modified proteins - Can discover novel Kbhb sites - Verifies antibody specificity | - Technically challenging - Expensive - Requires specialized equipment | - Validating antibody specificity - Discovery of new Kbhb sites - Comprehensive Kbhb proteomics |
Top-down Proteomics | - Analyzes intact proteins - Identifies combinatorial modifications - Resolves multiple lysine residues | - Very specialized technique - Challenging data interpretation - Limited to smaller proteins | - Studying Kbhb heterogeneity - Understanding combinatorial PTM patterns - Detailed modification stoichiometry |
Immunohistochemistry & Immunofluorescence | - Provides spatial information - In situ detection - Cell-to-cell variation visible | - Fixation may affect modifications - Limited quantification - Specificity concerns | - Subcellular localization studies - Tissue-specific modification patterns - Visualizing cellular dynamics |
For comprehensive β-hydroxybutyrylation research, integrating multiple methods provides the most robust understanding of this modification's biological significance and regulation .
Designing experiments to investigate the functional significance of β-hydroxybutyrylation at H3K122 requires a systematic approach:
Manipulating β-hydroxybutyrylation levels:
Metabolic intervention: Treat cells with β-hydroxybutyrate (4-10mM) to increase Kbhb levels
Ketogenic conditions: Use low glucose/high fat medium or serum from fasting subjects
Genetic approaches: Manipulate enzymes potentially involved in β-hydroxybutyrylation (e.g., p300, CBP) using CRISPR-Cas9 or RNAi
Functional readouts:
Transcriptional analysis: RNA-seq to identify genes affected by H3K122bhb changes
Chromatin accessibility: ATAC-seq or MNase-seq to determine if H3K122bhb affects nucleosome stability
Protein interactions: Identify readers of H3K122bhb using modified peptide pulldowns followed by mass spectrometry
Site-specific manipulation:
Generate H3K122 mutants (K122R to prevent modification; K122Q to mimic acylation)
Express these mutants in cells with endogenous H3 depletion
Assess phenotypic and molecular consequences
Integration with other histone modifications:
Perform sequential ChIP experiments (ChIP-reChIP) to determine co-occurrence with other modifications
Analyze spatial relationships between H3K122bhb and transcriptional machinery
When interpreting results, consider that H3K122 is located at the dyad axis of the nucleosome, potentially affecting DNA-histone interactions and chromatin accessibility.
Rigorous controls are essential when working with β-hydroxybutyryl-HIST1H3A (K122) antibody:
Antibody specificity controls:
Peptide competition assays: Pre-incubate antibody with β-hydroxybutyrylated and unmodified H3K122 peptides
Cross-reactivity assessment: Test against samples with known acetylation or other acylation modifications
Knockdown/knockout validation: Use H3.1-depleted cells as negative controls
Treatment controls:
Technical controls:
Loading controls: Total H3 or housekeeping proteins
Secondary antibody-only controls: To assess non-specific binding
Isotype controls: Non-specific rabbit IgG for immunoprecipitation experiments
Validation controls:
Documenting all these controls systematically enhances the reliability and reproducibility of your research findings.
Quantifying changes in β-hydroxybutyrylation levels requires appropriate techniques and analytical approaches:
Western blot quantification:
Normalize β-hydroxybutyryl-HIST1H3A (K122) signal to total H3 levels
Use densitometry software (ImageJ, Image Lab) for quantification
Apply statistical analysis across biological replicates (minimum n=3)
Present data as fold-change relative to control conditions
Mass spectrometry-based quantification:
Label-free quantification: Compare spectral counts or ion intensities of β-hydroxybutyrylated peptides
SILAC labeling: Differentially label control and treated cells for direct comparison
Calculate modification stoichiometry: Ratio of modified to unmodified peptides
Example calculation: % Kbhb = (Kbhb peptide intensity / (Kbhb peptide + unmodified peptide intensity)) × 100
ChIP-seq quantification:
Normalize H3K122bhb ChIP-seq reads to input and total H3 ChIP
Calculate differential binding scores across conditions
Correlate changes with gene expression data
Focus analysis on promoters, enhancers, and gene bodies separately
Immunofluorescence quantification:
Measure nuclear fluorescence intensity using confocal microscopy
Perform single-cell analysis across populations
Plot distribution shifts rather than just average changes
When interpreting results, consider that BHB treatment typically increases H3K122bhb levels, with observed fold-changes of 2-5× being biologically significant in published studies.
Researchers commonly encounter several challenges when working with β-hydroxybutyryl-HIST1H3A (K122) antibodies:
Weak or absent signals:
Problem: Insufficient β-hydroxybutyrylation levels in samples
Solution: Pre-treat cells with β-hydroxybutyrate (4-10mM) for 12-24 hours
Problem: Degradation of modifications during sample preparation
Solution: Include deacetylase inhibitors and protease inhibitors in all buffers; process samples quickly at 4°C
Non-specific binding and high background:
Problem: Antibody cross-reactivity with other modifications
Solution: Increase blocking time/concentration (5% milk or BSA for 1-2 hours); optimize antibody dilution with titration experiments
Problem: Excessive secondary antibody binding
Solution: More stringent washing (additional washes with higher Tween-20 concentration)
Inconsistent results between experiments:
Problem: Variability in cell metabolic state
Solution: Standardize cell density, passage number, and serum conditions
Problem: Antibody lot-to-lot variation
Solution: Validate each new lot against previous lots; consider creating an internal standard sample
Discrepancies between detection methods:
Problem: Different results from WB vs. ChIP vs. IF
Solution: Verify antibody validation for each specific application; optimize protocols for each technique separately
Conflicting mass spectrometry data:
Optimizing ChIP protocols for β-hydroxybutyryl-HIST1H3A (K122) antibody requires several specific considerations:
Crosslinking optimization:
Test different formaldehyde concentrations (0.5-2%) and incubation times (5-15 minutes)
Consider dual crosslinking with formaldehyde followed by EGS (ethylene glycol bis-succinimidyl succinate) for better preservation of protein-protein interactions
Chromatin fragmentation:
Optimize sonication conditions to achieve fragments of 200-500bp
Verify fragment size distribution by agarose gel electrophoresis
Consider using enzymatic digestion (MNase) as an alternative to sonication for more precise fragmentation
Antibody incubation parameters:
Test different antibody amounts (2-10 μg per ChIP reaction)
Optimize incubation time (overnight at 4°C is standard, but test 4-16 hours)
Include an IgG control and a positive control antibody (e.g., H3K4me3) in parallel
Washing stringency balancing:
Implement a gradient of washing stringency to find optimal conditions
Example washing series: Low salt → High salt → LiCl → TE buffer
Monitor signal-to-noise ratio across different washing conditions
Elution and reversal of crosslinking:
Test different elution buffers (SDS-based vs. acidic elution)
Optimize reversal of crosslinking (65°C for 4-16 hours)
Include RNase A and Proteinase K treatments
Quality control metrics:
Calculate percent input recovery (should be >1% for histone marks)
Assess enrichment at known positive vs. negative genomic regions by qPCR
Measure signal-to-noise ratio compared to IgG control (should be >10-fold)
When optimizing, preserve sample aliquots at each step to troubleshoot specific stages if the final output is suboptimal.
Using β-hydroxybutyryl-HIST1H3A (K122) antibody across different cell types and tissues requires careful consideration of several factors:
Baseline β-hydroxybutyrylation variability:
Metabolically active tissues (liver, brain, heart) may have higher basal Kbhb levels
Proliferating cells often show different histone modification patterns than differentiated cells
Consider measuring β-hydroxybutyrate levels in different tissues to predict modification abundance
Cell/tissue-specific extraction protocols:
Adjust lysis conditions based on tissue hardness and extracellular matrix composition
For tissues: Consider using a Dounce homogenizer followed by filtration
For difficult tissues: Might require additional mechanical disruption or enzymatic digestion
Fixation considerations for immunohistochemistry:
Fixative choice affects epitope accessibility (4% PFA is standard, but test 1-4% range)
Fixation time should be optimized for each tissue type (10 min - 24 hours)
Antigen retrieval methods need tissue-specific optimization (citrate buffer pH 6.0 vs. Tris-EDTA pH 9.0)
Antibody concentration adjustments:
Different tissues may require different antibody dilutions
Create a dilution series (1:250 to 1:2000) for each new cell type/tissue
Monitor signal-to-noise ratio to determine optimal concentration
Confounding factors in specific tissues:
Tissues with high lipid content (brain, adipose) may have naturally elevated β-hydroxybutyrate levels
Autofluorescence in certain tissues (liver, kidney) may interfere with IF detection
Endogenous peroxidases can affect IHC staining
Physiological state considerations:
Fasting/feeding state dramatically affects β-hydroxybutyrylation levels
Consider time of collection and nutritional status
In rodent models, standardize collection time relative to feeding cycle
When publishing, thoroughly document all cell/tissue-specific protocol adaptations to ensure reproducibility.
β-hydroxybutyrylation at H3K122 presents distinct characteristics compared to other histone modifications:
Structural and functional significance:
H3K122 is located at the dyad axis of the nucleosome, where DNA makes strong contacts with the histone octamer
Modifications at this site potentially disrupt histone-DNA interactions more directly than modifications on histone tails
This disruption may facilitate DNA accessibility to transcription factors and the transcriptional machinery
Genomic localization patterns:
Comparison with other modifications at H3K122:
H3K122 can also undergo acetylation (H3K122ac), which similarly associates with active chromatin
The chemical differences between β-hydroxybutyrylation and acetylation (additional hydroxyl group and longer carbon chain) may recruit different reader proteins
Metabolic connections:
As a β-hydroxybutyrylation site, H3K122bhb likely responds to metabolic fluctuations, particularly ketone body metabolism
This creates a potential direct link between cellular metabolic state and gene regulation at specific genomic regions
Relationship with other histone modifications:
May work cooperatively with activating marks like H3K4me3 or H3K27ac
Could have antagonistic relationships with repressive marks like H3K27me3
The combinatorial pattern with other modifications determines the ultimate functional outcome
Understanding these distinctive characteristics helps researchers interpret H3K122bhb data in the broader context of chromatin regulation and gene expression.
The enzymatic regulation of β-hydroxybutyrylation at H3K122 remains an evolving area of research:
Putative "writers" (acetyltransferases with potential β-hydroxybutyrylation activity):
HATs like p300 and CBP may have promiscuous acyltransferase activity, potentially catalyzing β-hydroxybutyrylation
These enzymes typically use acyl-CoA as donors, suggesting β-hydroxybutyryl-CoA may serve as a substrate
Definitive identification of specific enzymes for H3K122bhb remains an active research question
Regulation mechanisms:
Metabolic regulation: β-hydroxybutyryl-CoA levels, derived from β-hydroxybutyrate metabolism, likely influence modification rates
Enzymatic competition: Various acyl-CoA species (acetyl-CoA, butyryl-CoA, β-hydroxybutyryl-CoA) may compete for the same enzyme active sites
Spatial regulation: Nuclear localization of metabolic enzymes generating β-hydroxybutyryl-CoA affects local substrate availability
Potential "erasers" (deacylases):
HDACs (particularly class I and II) and sirtuins may remove β-hydroxybutyryl groups
SIRT3 has been implicated in removing various acylations, potentially including β-hydroxybutyrylation
Enzymatic specificity for different acylations varies, potentially creating differential regulation
"Readers" of β-hydroxybutyrylation:
Bromodomain-containing proteins that recognize acetylated lysines might also interact with β-hydroxybutyrylated residues
YEATS domain proteins, which read various acylations, are potential candidates
Specific readers for H3K122bhb have not been definitively characterized
This incomplete understanding highlights opportunities for researchers to make significant contributions to this field through targeted experiments identifying and characterizing the enzymatic machinery governing H3K122 β-hydroxybutyrylation.
Integrating β-hydroxybutyrylation data with other omics approaches provides a comprehensive understanding of its biological significance:
Multi-omics integration strategies:
Correlate H3K122bhb ChIP-seq data with:
Transcriptomics (RNA-seq) to associate modification with gene expression
Other histone modification ChIP-seq to identify co-occurrence patterns
Chromatin accessibility (ATAC-seq, DNase-seq) to assess functional impact
Proteomics to connect histone modifications with protein expression
Metabolomics integration:
Measure β-hydroxybutyrate levels and related metabolites alongside Kbhb profiling
Correlate β-hydroxybutyrate/β-hydroxybutyryl-CoA concentrations with modification abundance
Create metabolic perturbation time series to track dynamic responses
Bioinformatic approaches:
Implement supervised machine learning to identify predictive relationships between Kbhb and other datasets
Use network analysis to place H3K122bhb in broader regulatory networks
Apply integrative genomics approaches like WGCNA (Weighted Gene Co-expression Network Analysis)
Biological context integration:
Connect β-hydroxybutyrylation changes to:
Cell cycle progression
Differentiation trajectories
Stress responses
Metabolic adaptations (fasting, ketogenic conditions)
Visualization and analysis tools:
Generate multi-dimensional visualizations integrating various omics layers
Use genome browsers with multiple tracks to visualize spatial relationships
Implement pathway enrichment analyses incorporating multiple data types
The integration of these diverse data types helps position H3K122bhb within the broader cellular regulatory landscape, revealing its functional roles in various biological processes and potentially identifying new therapeutic targets or biomarkers.
Recent research has expanded our understanding of H3K122 β-hydroxybutyrylation's role in gene regulation:
Functional genomics insights:
H3K122bhb, like other Kbhb marks, appears enriched at active gene promoters
The strategic position of K122 at the nucleosome dyad axis suggests direct impacts on DNA accessibility
Recent studies indicate potential roles in regulating specific gene sets responsive to metabolic fluctuations
The modification may serve as a direct link between cellular metabolic state and transcriptional regulation
Biological context discoveries:
Emerging evidence connects H3K122bhb to cellular adaptations during fasting/feeding cycles
Studies suggest potential roles in metabolic diseases, particularly those involving ketone body metabolism
Investigation into developmental contexts reveals dynamic regulation during cellular differentiation
Potential regulatory functions during stress responses that alter metabolic states
Technological advancements:
Improved antibody specificity has enabled more reliable detection of H3K122bhb
Advanced mass spectrometry techniques allow better quantification of modification stoichiometry
Single-cell approaches are beginning to reveal cell-to-cell variation in H3K122bhb patterns
CRISPR-based approaches enable site-specific manipulation of H3K122 to assess functional consequences
Mechanistic understanding:
Recent investigations into the "readers" of H3K122bhb have identified potential protein interactors
Structural studies provide insight into how β-hydroxybutyrylation may alter histone-DNA interactions
Emerging models integrate H3K122bhb into broader epigenetic regulatory networks
These developments collectively point to H3K122bhb as an important epigenetic regulator that connects cellular metabolism with gene expression programs.
Comparing antibody-based and mass spectrometry-based approaches for studying β-hydroxybutyryl-HIST1H3A (K122) reveals important complementarities and discrepancies:
Detection sensitivity comparison:
Antibody-based methods generally offer higher sensitivity for detecting low-abundance modifications
Mass spectrometry provides definitive chemical identification but may miss low-stoichiometry modifications
Studies using both approaches show antibodies can detect H3K122bhb in conditions where MS approaches may yield negative results
Specificity considerations:
Quantification capabilities:
MS approaches provide more accurate absolute quantification of modification stoichiometry
Antibody methods offer better relative quantification across different samples/conditions
Studies combining both approaches show that antibody signal intensity doesn't always linearly correlate with modification abundance
Coverage differences:
Antibody methods enable genome-wide localization studies (ChIP-seq)
MS approaches better identify co-occurring modifications on the same histone molecule
Integrated studies reveal unique insights not obtainable from either method alone
Consensus and discrepancies:
Both methods confirm H3K122bhb increases upon BHB treatment
Potential discrepancies in baseline detection levels should be interpreted carefully
Researchers should be aware that MS-identified bhb sites may not always be detectable by available antibodies
The field is increasingly moving toward integrated approaches, using antibodies for high-sensitivity detection and localization, complemented by MS for definitive identification and accurate quantification.
Several promising research directions will advance our understanding of β-hydroxybutyrylation function:
Molecular mechanism exploration:
Identifying and characterizing specific "writers," "erasers," and "readers" of H3K122bhb
Determining the structural consequences of β-hydroxybutyrylation at the nucleosome level
Developing specific inhibitors or activators of enzymes regulating β-hydroxybutyrylation
Physiological and disease relevance:
Investigating β-hydroxybutyrylation dynamics during:
Aging and longevity
Metabolic diseases (diabetes, obesity)
Neurodegenerative disorders
Cancer metabolism reprogramming
Exploring therapeutic potential of manipulating β-hydroxybutyrylation through diet or pharmacological approaches
Technological developments:
Creating more specific antibodies against various Kbhb sites
Developing live-cell imaging tools for tracking dynamic β-hydroxybutyrylation changes
Implementing CRISPR-based technologies for site-specific manipulation of lysine residues
Enhancing computational approaches to integrate Kbhb data with other omics datasets
Evolutionary perspectives:
Comparative analysis of β-hydroxybutyrylation across species
Investigating conservation of regulatory mechanisms
Exploring species-specific adaptations in β-hydroxybutyrylation pathways
Single-cell and spatial technologies:
Mapping β-hydroxybutyrylation heterogeneity at the single-cell level
Spatial transcriptomics integrated with histone modification data
In situ detection of β-hydroxybutyrylation in tissue contexts
These research directions collectively will provide a comprehensive understanding of β-hydroxybutyrylation's role in cellular physiology and potentially reveal new therapeutic opportunities for manipulating this modification in disease contexts.