Histone H2B is a core component of nucleosomes, which organize DNA into chromatin. Acetylation at K20 reduces chromatin compaction, enhancing DNA accessibility for transcription, replication, and repair machinery . This modification is dynamically regulated by histone acetyltransferases (HATs) and deacetylases (HDACs), linking it to epigenetic states in health and disease .
Gene Activation: Acetyl-K20 marks transcriptionally active chromatin regions. The antibody has been used in ChIP-seq to map acetylated H2B-enriched promoters .
DNA Repair: Acetylation at K20 correlates with recruitment of repair complexes to DNA damage sites, as shown in IF studies .
Cancer: Elevated H2B-K20 acetylation is observed in breast cancer tissues (validated via IHC) , suggesting a role in oncogenic gene expression.
Metabolic Disorders: Cross-reactivity studies with β-hydroxybutyrylated H2B (K20) reveal metabolic-epigenetic crosstalk in conditions like diabetes .
Data from commercial sources highlight critical differences:
Note: "Citations" refer to peer-reviewed studies validating each product.
Dilution Range:
Cross-Reactivity: No observed reactivity with non-acetylated H2B or other histone variants (e.g., H3K27ac) .
Recent studies using this antibody have revealed:
Metabolic Regulation: Sodium butyrate treatment increases H2B-K20 acetylation in HeLa cells, detectable via WB .
Developmental Roles: Embryonic stem cells show dynamic K20 acetylation patterns during differentiation (ChIP-seq data) .
Antimicrobial Activity: Acetylated H2B exhibits enhanced bactericidal function in colonic epithelial cells .
Acetyl-HIST1H2BC (K20), commonly known as H2BK20ac, is a post-translational modification occurring at lysine 20 of histone H2B. This modification is part of a broader group of histone H2B N-terminal acetylation sites (H2BNTac) that function as significant epigenetic regulators. Biologically, H2BK20ac serves as a signature mark of active enhancers, distinguishing them from other cis-regulatory elements in the genome . Unlike some other histone modifications, H2BK20ac exhibits specificity in marking candidate active enhancers and a subset of promoters, helping to discriminate them from ubiquitously active promoters . The acetylation at this site neutralizes the positive charge of the lysine residue, altering protein conformation and contributing to chromatin structure remodeling that facilitates transcription factor access to DNA . This modification is enzymatically added by histone acetyltransferases CBP/p300 and can be removed by histone deacetylases (HDACs) 1 and 2, creating a dynamic regulation system for gene expression .
H2BK20ac differs from other histone acetylation marks in several key functional aspects:
Specificity for active enhancers: While H3K27ac marks both promoters and enhancers, H2BK20ac and other H2BNTac marks show greater specificity for active enhancers and only a subset of promoters .
Enzymatic regulation: H2BK20ac is specifically catalyzed by CBP/p300, whereas other histone acetylation marks may be regulated by multiple acetyltransferases. This specificity makes it a more precise indicator of CBP/p300 activity .
Nucleosome dynamics: H2BK20ac is found on H2A-H2B dimers, which are rapidly exchanged through transcription-induced nucleosome remodeling, unlike H3-H4 tetramers. This contributes to its distinct genomic distribution and dynamics .
Enhancer strength prediction: H2BK20ac intensity more accurately predicts enhancer strength and outperforms current models in predicting CBP/p300 target genes compared to other histone marks .
Response to transcription inhibition: Unlike some co-transcriptionally deposited modifications (such as H2BK120ub), H2BK20ac and other histone acetylation marks are not reduced by transcription inhibition but are significantly decreased by CBP/p300 inhibition .
This functional differentiation makes H2BK20ac particularly valuable for fine-grained enhancer mapping and modeling CBP/p300-dependent gene regulation in experimental settings.
For detecting H2BK20ac in chromatin studies, researchers should consider multiple complementary approaches:
Chromatin Immunoprecipitation followed by sequencing (ChIP-seq): This is the gold standard for genome-wide mapping of H2BK20ac. When performing ChIP-seq for H2BK20ac, it's crucial to use highly specific antibodies and include appropriate controls such as input chromatin and IgG controls . For optimal results, crosslinking time and sonication conditions should be carefully optimized.
ELISA-based detection: Commercial ELISA kits for Acetyl-HIST1H2BC (K20) are available for quantitative assessment of this modification in chromatin extracts . These assays can provide quantitative measurements but lack the spatial resolution of ChIP-based methods.
Mass spectrometry: Quantitative mass spectrometry offers antibody-independent detection of H2BK20ac and can simultaneously measure the stoichiometry of multiple acetylation sites on the same histone protein . This approach is particularly valuable for understanding the co-occurrence of different modifications.
Immunofluorescence microscopy: For visualization of nuclear distribution patterns, immunofluorescence using specific antibodies against H2BK20ac can be employed, though this approach offers lower resolution than ChIP-seq .
CUT&RUN or CUT&Tag: These newer techniques offer higher signal-to-noise ratios than traditional ChIP and require fewer cells, making them valuable for samples with limited material.
When selecting an antibody for H2BK20ac detection, researchers should verify its specificity using peptide competition assays or knockout models to ensure it does not cross-react with other acetylated lysines on H2B or other histones.
Differentiating causative from correlative relationships between H2BK20ac and transcriptional activation requires sophisticated experimental approaches:
Temporal resolution studies: Implementing time-course experiments with high temporal resolution can establish whether H2BK20ac precedes or follows transcriptional activation. This can involve synchronized cell populations or inducible gene expression systems coupled with ChIP-seq and nascent RNA sequencing (e.g., PRO-seq or GRO-seq) .
Site-specific acetylation manipulation: CRISPR-based epigenome editing approaches using catalytically dead Cas9 (dCas9) fused to acetyltransferase domains can target specific genomic loci for H2BK20 acetylation, allowing researchers to observe if targeted acetylation leads to transcriptional activation independent of other factors .
Pharmacological inhibitor studies: Comparing the effects of transcription inhibitors (ActD, Trp, NVP-2) versus CBP/p300 inhibitors (A-485) can help dissect the relationship. Research has shown that while CBP/p300 inhibition reduces H2BK20ac, transcription inhibition does not significantly affect H2BK20ac levels, suggesting acetylation is not merely a consequence of transcription .
Mutational studies: Creating lysine-to-arginine mutations at H2BK20 (preserving charge but preventing acetylation) using CRISPR-Cas9 genome editing can test the necessity of this specific modification for transcriptional activation at target genes.
Mathematical modeling: Implementing systems biology approaches that integrate multi-omics data can help infer causal relationships. For example, structural equation modeling or Bayesian networks can be used to model the directionality between H2BK20ac, transcription factor binding, chromatin accessibility, and gene expression.
The emerging evidence from recent studies suggests that histone acetylation, including H2BK20ac, is not merely a consequence of transcription but rather plays a causative role in facilitating enhancer and promoter activity. This is evidenced by the fact that acetyltransferase recruitment and activation are uncoupled from the act of transcription, and histone acetylation is sustained even in the absence of ongoing transcription .
The relationship between H2BK20ac and other H2B N-terminal acetylation marks (H2BNTac) in determining enhancer activity represents a complex interplay of epigenetic modifications:
Co-occurrence patterns: ChIP-seq analyses have revealed extensive overlap between H2BK20ac and other H2BNTac sites (H2BK5ac, H2BK11ac, H2BK12ac, H2BK16ac), with most H2BNTac sites marking the same genomic regions . This suggests potential functional redundancy or cooperative action among these marks.
Differential enrichment at enhancers versus promoters: While H2BK16ac, H2BK20ac, and H3K27ac mark most MED1-positive intergenic regions (candidate enhancers), H2BK12ac and certain H2BK5ac marks show more limited coverage . The table below summarizes the relative enrichment of different acetylation marks at enhancers:
Histone Mark | Enrichment at MED1+ Intergenic Regions | Enrichment at Promoters |
---|---|---|
H2BK16ac | High | Moderate |
H2BK20ac | High | Low |
H3K27ac | High | High |
H2BK12ac | Moderate | Low |
H2BK5ac | Variable | Low |
Functional hierarchy: Evidence suggests a functional hierarchy among H2BNTac marks, with some showing stronger predictive power for enhancer activity. H2BK20ac intensity, along with H2BK16ac, more accurately predicts enhancer strength compared to H3K27ac or other H2BNTac marks .
Differential regulation by HATs and HDACs: While all H2BNTac marks are catalyzed by CBP/p300, they show differential sensitivity to CBP/p300 inhibition. A-485 (CBP/p300 inhibitor) more strongly reduces H2BNTac in promoters and gene bodies than in distal regions , suggesting context-dependent regulation of these marks.
Combinatorial effects: The presence of multiple H2BNTac marks appears to have an additive effect on enhancer activity. Regions with multiple H2BNTac marks show higher transcriptional output from the target genes compared to regions with single marks .
This complex relationship underscores the importance of considering the entire profile of H2B acetylation marks rather than individual modifications when studying enhancer activity and gene regulation.
H2BK20ac contributes significantly to prognostic assessment in hepatocellular carcinoma (HCC) and potentially other cancers through multiple mechanisms:
The prognostic value of H2BK20ac extends beyond HCC, with emerging evidence suggesting similar roles in other cancer types including colorectal, breast, and prostate cancers. Importantly, as a CBP/p300-specific modification, H2BK20ac may serve as a more precise biomarker of CBP/p300 dysregulation in cancer compared to the more broadly regulated H3K27ac .
When conducting ChIP-seq experiments with Acetyl-HIST1H2BC (K20) antibody, implementing comprehensive controls is essential for accurate data interpretation:
Input chromatin control: Always process a portion of the same chromatin preparation without immunoprecipitation to account for biases in chromatin fragmentation, DNA isolation, and sequencing. Input normalization is crucial for accurate peak calling and quantitative analysis of H2BK20ac enrichment .
IgG negative control: Include a non-specific IgG control from the same species as the H2BK20ac antibody to identify non-specific binding events and background signal levels.
Peptide competition control: Pre-incubate the H2BK20ac antibody with synthetic peptides containing acetylated H2BK20 to demonstrate binding specificity. Include both acetylated and unacetylated peptides to confirm acetylation-specific recognition.
Spike-in normalization: Add a small amount of chromatin from a different species (e.g., Drosophila) as an internal control to enable quantitative comparisons between samples, especially when global changes in H2BK20ac levels are expected.
CBP/p300 inhibition control: Include samples treated with CBP/p300 inhibitors like A-485 as positive controls for reduction in H2BK20ac. This control validates both antibody specificity and the experimental system .
Transcription inhibition control: Include samples treated with transcription inhibitors (ActD, NVP-2, or Trp) to demonstrate that H2BK20ac is maintained independent of active transcription .
Biological validation: Confirm key findings using orthogonal techniques such as CUT&RUN, CUT&Tag, or quantitative mass spectrometry to validate the ChIP-seq results .
The inclusion of these controls addresses potential artifacts, validates antibody specificity, and enables accurate quantitative analysis of H2BK20ac distribution across the genome. Additionally, performing parallel ChIP-seq for H3K27ac and/or H2BK16ac can provide valuable comparative data, as these marks frequently co-occur with H2BK20ac at enhancers but may show distinct patterns at promoters .
Resolving discrepancies between H2BK20ac and other enhancer marks in genome-wide studies requires systematic analytical approaches:
Integrative peak analysis: For regions showing discordance between H2BK20ac and other marks (e.g., H3K27ac, H3K4me1), perform detailed integrative analysis using tools like bedtools, deepTools, or the R/Bioconductor package DiffBind to characterize the genomic features of concordant versus discordant regions .
Chromatin state segmentation: Apply unsupervised learning algorithms like ChromHMM with and without including H2BK20ac to identify how this mark influences chromatin state definitions. Previous studies have shown that including H2BNTac in ChromHMM analysis can merge H3K4me1-enriched transcriptional start site states with enhancer states containing both H3K27ac and H2BNTac marks .
Functional validation of discordant regions: For regions marked by H2BK20ac but not other established enhancer marks (or vice versa), perform targeted validation using enhancer reporter assays, CRISPR interference, or activation to assess their functional significance .
Cell type-specific analysis: When discrepancies arise between different studies, consider cell type-specific effects. H2BNTac marks may show greater tissue specificity than H3K27ac in certain contexts .
Technical bias assessment: Systematically evaluate technical factors that might contribute to discrepancies:
Antibody specificity and batch effects
ChIP-seq protocol variations (crosslinking time, sonication conditions)
Bioinformatic pipeline differences (peak calling algorithms, threshold settings)
Sequential ChIP (re-ChIP): For directly assessing co-occurrence of marks at the same nucleosomes, perform sequential ChIP experiments where chromatin is first immunoprecipitated with one antibody (e.g., H2BK20ac) and then with another (e.g., H3K27ac) .
Transcription factor co-occupancy analysis: Analyze the co-occurrence of transcription factors at regions with concordant versus discordant histone marks. Regions with H2BK20ac but not H3K27ac may show distinct transcription factor binding patterns .
When resolving such discrepancies, researchers should remember that some enhancers might be in different activity states, with H2BK20ac potentially marking a specific subset of active enhancers or marking enhancers at different stages of activation compared to other histone modifications .
Optimizing Acetyl-HIST1H2BC (K20) antibody-based assays requires careful attention to several critical parameters that vary across experimental contexts:
Antibody selection and validation:
Confirm specificity through peptide arrays testing reactivity against other acetylated H2B lysines (K5, K11, K12, K16)
Verify performance in the intended application (ChIP-seq, immunofluorescence, Western blot)
For quantitative applications, determine the linear detection range of the antibody
When possible, validate results with multiple antibody clones to rule out clone-specific artifacts
ChIP-seq optimization:
Crosslinking: Test multiple formaldehyde concentrations (0.5-2%) and incubation times (5-20 minutes)
Sonication: Optimize conditions to achieve 200-500 bp fragments for standard ChIP-seq
Antibody concentration: Typically 2-5 μg per ChIP reaction, but should be titrated
Washing stringency: Balanced to minimize background while preserving specific signal
Library preparation: Select methods appropriate for the typically limited material recovered from ChIP
Immunofluorescence parameters:
Fixation: 4% paraformaldehyde (10 minutes) works well for most histone modifications
Permeabilization: Triton X-100 (0.2-0.5%) is typically sufficient
Blocking: BSA (3-5%) with normal serum matching secondary antibody species
Antibody dilution: Start with 1:200-1:1000 and optimize
Controls: Include CBP/p300 inhibitor-treated samples as negative controls
Western blot considerations:
Sample preparation: Use histone extraction protocols that preserve acetylation (e.g., acid extraction)
Blocking: Milk can contain deacetylases; BSA or commercial blockers are preferred
Antibody concentration: Typically 1:500-1:2000 dilution
Loading controls: Total H2B or H3 are appropriate
Signal detection: Enhanced chemiluminescence or fluorescent secondary antibodies for quantification
ELISA optimization:
Mass spectrometry considerations:
Sample preparation: Optimize histone extraction and digestion (typically using propionylation and trypsin)
Peptide enrichment: Consider using antibody-based enrichment for acetylated peptides
Analytical parameters: Optimize collision energy and resolution for acetylated peptides
Data analysis: Use specialized software for histone PTM quantification
The optimal parameters vary significantly by experimental context, sample type, and specific research question. Pilot experiments to determine ideal conditions for each unique experimental setup are strongly recommended.