Target Protein: HIST1H1B (Histone H1.5) is a linker histone that binds to linker DNA between nucleosomes, stabilizing higher-order chromatin structures .
Modification Site: The antibody recognizes acetylation at lysine 16, a post-translational modification linked to chromatin decondensation and transcriptional activation .
Immunogen: A peptide sequence surrounding acetylated lysine 16 derived from human Histone H1.5 .
The antibody’s high specificity for human samples makes it ideal for studying histone acetylation in cancer, development, and chromatin remodeling .
Chromatin Dynamics: Acetylation at K16 disrupts histone-DNA interactions, promoting an open chromatin state conducive to transcription .
Disease Relevance: Aberrant acetylation patterns are implicated in cancer, where chromatin remodeling disrupts gene regulation .
Therapeutic Potential: The antibody aids in identifying therapeutic targets, such as histone acetyltransferases or deacetylases (HATs/HDACs) .
Multiple vendors offer this antibody, with pricing varying by supplier and quantity:
Supplier | SKU | Size | Price (USD) |
---|---|---|---|
Assay Genie | PACO56606 | 50 µl | Not listed |
AFG Scientific | A24651 | 50 µl | $225 |
Abbexa | NA | 100 µl | Not listed |
Thomas Scientific | NA | 50 µl | Not listed |
The Acetyl-HIST1H1B (K16) Antibody is a polyclonal antibody that specifically recognizes the acetylation modification at lysine 16 of the Histone H1.5 (HIST1H1B) protein in humans. Histone H1.5 is a linker histone that binds to DNA between nucleosomes, contributing to the formation of higher-order chromatin structures. This antibody targets a specific post-translational modification that plays a crucial role in chromatin remodeling and gene expression regulation .
The antibody is typically raised in rabbits using a synthetic peptide sequence surrounding the acetylated lysine 16 residue of human Histone H1.5 as the immunogen. This highly specific design allows researchers to investigate one particular epigenetic modification among the complex landscape of histone modifications .
The Acetyl-HIST1H1B (K16) Antibody has been validated for several key research applications in epigenetics:
Chromatin Immunoprecipitation (ChIP): Used to investigate the genomic distribution of this specific histone modification and its association with regulatory elements .
Immunofluorescence (IF): Enables visualization of the subcellular localization of acetylated HIST1H1B within cell nuclei, with recommended dilutions of 1:50-1:200 .
Enzyme-Linked Immunosorbent Assay (ELISA): Allows quantitative measurement of acetylated HIST1H1B levels in biological samples, with recommended dilutions of 1:2000-1:10000 .
For immunofluorescence applications, researchers have successfully used this antibody on HepG2 cells treated with sodium butyrate (30 mM, 4h), visualized with Alexa Fluor 488-conjugated secondary antibodies .
Acetylation of lysine 16 on HIST1H1B represents a specific epigenetic mark that influences chromatin structure and gene expression. This modification is part of the "histone code" that regulates DNA accessibility to transcription machinery .
Histone H1 proteins, including HIST1H1B, are essential for the condensation of nucleosome chains into higher-order structured fibers. The acetylation of lysine 16 can alter this process by reducing the positive charge of the histone, potentially weakening its interaction with negatively charged DNA. This modification functions as a regulatory mechanism for gene transcription through:
Chromatin remodeling: Affecting the higher-order structure of chromatin
Nucleosome spacing: Influencing the positioning of nucleosomes along DNA
DNA methylation patterns: Interacting with DNA methylation machinery
Research has shown that aberrant patterns of histone acetylation, including at this specific site, have been implicated in various diseases, particularly cancer, making this antibody valuable for understanding disease mechanisms and potentially developing therapeutic interventions .
When working with rare cell populations for ChIP-seq experiments using the Acetyl-HIST1H1B (K16) Antibody, several optimization strategies should be considered:
Sample preparation optimization: For limited cell numbers, modify standard protocols by:
Implementing a micro-ChIP approach with reduced volumes
Using carrier chromatin (from another species) to maintain proper chromatin-to-surface ratios
Employing automated microfluidic devices to minimize sample loss
Antibody titration: The standard recommendation for ChIP applications should be adjusted based on cell number. For rare populations, test multiple antibody concentrations (ranging from 2-10 μg) on a small pilot experiment to determine optimal signal-to-noise ratio .
Crosslinking modification: For rare acetylation marks, consider dual crosslinking using both formaldehyde and protein-specific crosslinkers to preserve transient protein-DNA interactions.
Signal amplification methods: Implement linear amplification methods specifically designed for ChIP-seq with limited starting material, while carefully avoiding PCR bias .
Validation controls: Always include appropriate controls specific to acetylation studies, including:
When faced with contradictory results between western blot and immunofluorescence studies using the Acetyl-HIST1H1B (K16) Antibody, a systematic troubleshooting approach is essential:
Sample preparation considerations:
For western blots: Ensure histones are properly extracted using acid extraction methods that preserve acetylation marks. Include HDAC inhibitors throughout the process
For IF: Ensure fixation methods preserve the epitope structure, as some fixatives can mask acetylation sites
Epitope accessibility assessment:
Different detection methods expose antigens differently
In western blots, proteins are denatured, potentially exposing epitopes that might be masked in fixed cells
Test alternative fixation and permeabilization protocols for IF
For western blots, consider using different detergents or denaturing conditions
Cross-validation approaches:
Perform peptide competition assays to confirm antibody specificity
Use HDAC inhibitors (sodium butyrate at 30mM for 4h has been validated) to increase acetylation signals
Compare results with another antibody targeting the same modification
Implement orthogonal techniques such as mass spectrometry to confirm acetylation status
Technical optimizations:
For IF: Test the validated dilution range of 1:50-1:200 and optimize blocking conditions
For western blots: Optimize transfer conditions specifically for histones, which can be challenging due to their small size and basic nature
Consider the buffer conditions, as the antibody is stored in 50% glycerol with 0.03% Proclin 300, which may affect performance in certain applications
The acetylation profile of HIST1H1B at lysine 16 demonstrates dynamic changes throughout the cell cycle, reflecting its role in chromatin organization during different cellular processes:
G1 Phase:
Moderate levels of K16 acetylation typically observed
Acetylation patterns show diffuse nuclear distribution
Associated with euchromatic regions and transcriptionally active domains
S Phase:
Significant reduction in K16 acetylation levels
This deacetylation appears necessary for proper DNA replication
Histone chaperones interact differentially with acetylated vs. non-acetylated H1 variants
G2 Phase:
Gradual increase in K16 acetylation
Redistribution of acetylation patterns in preparation for mitosis
Mitosis:
Sharp decrease in K16 acetylation coinciding with chromosome condensation
This deacetylation is critical for proper chromosome segregation
Specific HDACs are recruited to chromatin during this phase
These dynamic changes can be effectively monitored through immunofluorescence studies using the Acetyl-HIST1H1B (K16) Antibody (dilution 1:50-1:200) combined with cell cycle markers. Researchers can induce cell cycle synchronization through various methods (double thymidine block, nocodazole treatment, etc.) and then assess acetylation patterns at different time points using this antibody .
Chromatin immunoprecipitation experiments using this antibody on synchronized cell populations can further reveal genomic redistribution of this mark throughout the cell cycle, providing insights into its functional significance in chromosome dynamics and gene regulation.
For optimal immunofluorescence results with the Acetyl-HIST1H1B (K16) Antibody, follow this validated protocol:
Cell preparation and treatment:
Fixation and permeabilization:
Step | Reagent | Conditions | Critical Considerations |
---|---|---|---|
Fixation | 4% paraformaldehyde in PBS | 15 minutes at room temperature | Preserve acetylation marks |
Washing | PBS | 3x5 minutes | Gentle rocking |
Permeabilization | 0.2% Triton X-100 in PBS | 10 minutes at room temperature | Allows antibody access to nuclear antigens |
Blocking | 5% BSA in PBS | 1 hour at room temperature | Reduces non-specific binding |
Antibody incubation:
Primary antibody: Apply Acetyl-HIST1H1B (K16) Antibody at 1:50-1:200 dilution in blocking buffer, incubate overnight at 4°C
Washing: PBS with 0.1% Tween-20, 3x5 minutes
Secondary antibody: Apply fluorophore-conjugated anti-rabbit IgG (Alexa Fluor 488 has been validated) at 1:500 dilution, incubate for 1 hour at room temperature protected from light
Washing: PBS with 0.1% Tween-20, 3x5 minutes
Nuclear counterstaining and mounting:
Counterstain with DAPI (1 μg/mL in PBS) for 5 minutes
Mount with anti-fade mounting medium
Seal with nail polish and store at 4°C protected from light
Visualization parameters:
Excitation/emission wavelengths appropriate for the secondary antibody fluorophore
Capture Z-stack images to fully visualize nuclear distribution
Include single-stained controls for proper channel separation
This protocol has been successfully employed with HepG2 cells, demonstrating specific nuclear staining patterns consistent with the expected localization of acetylated histone H1.5 .
Rigorous experimental design for ChIP studies with the Acetyl-HIST1H1B (K16) Antibody requires comprehensive controls:
Technical Controls:
Input sample: Retain 5-10% of chromatin before immunoprecipitation to normalize for differences in chromatin quantity and quality
No antibody control: Perform IP procedure without adding antibody to assess non-specific binding to beads
IgG negative control: Use normal rabbit IgG at the same concentration as the specific antibody to measure background signal
Positive control antibody: Include an antibody against a well-characterized histone mark (H3K4me3 at active promoters) to verify ChIP procedure success
Biological Controls:
Treatment validation: Use HDAC inhibitors (validated approach: treat HeLa cells with 30mM sodium butyrate) to increase global acetylation levels
Cell-type specificity: Compare ChIP results across different cell types to identify cell-type-specific patterns
Gene region controls: Include primers for genomic regions known to be enriched for H1 variants (e.g., certain heterochromatic regions) and regions typically depleted of H1
Quantification Controls:
Standard curve: For qPCR analysis, include a standard curve using input DNA
Multiple primer sets: Analyze both positive regions (expected to show enrichment) and negative regions (not expected to show enrichment)
Technical replicates: Perform qPCR in triplicate
Biological replicates: Perform at least three independent ChIP experiments
Validation Strategies:
Sequential ChIP (Re-ChIP): Perform sequential immunoprecipitation with antibodies against known interacting factors to verify co-occupancy
Peptide competition: Pre-incubate antibody with acetylated and non-acetylated peptides to confirm specificity
Orthogonal validation: Confirm key findings with alternative methods such as CUT&RUN or CUT&Tag
Published protocols have demonstrated successful ChIP applications with this antibody at concentrations of approximately 5μg per 4×10^6 HeLa cells treated with sodium butyrate .
Proper storage and handling of the Acetyl-HIST1H1B (K16) Antibody is critical for maintaining its performance across different applications:
Long-term storage considerations:
Working solution preparation:
Application | Recommended Dilution | Diluent |
---|---|---|
Immunofluorescence | 1:50-1:200 | Blocking buffer (5% BSA in PBS) |
ChIP | 5μg per 4×10^6 cells | ChIP dilution buffer |
ELISA | 1:2000-1:10000 | Blocking buffer |
Critical handling factors:
Avoid repeated freeze-thaw cycles which significantly reduce antibody activity
Prepare small aliquots for single use when dividing the stock
Centrifuge briefly before opening vials to collect liquid at the bottom
Maintain cold chain during all handling steps
Avoid contamination by using sterile technique
Application-specific considerations:
For IF: Make fresh dilutions for each experiment; do not store diluted antibody
For ChIP: Pre-clear chromatin samples thoroughly to reduce non-specific binding
For all applications: Validate each new lot with positive controls
Performance monitoring over time:
Periodically test the antibody against positive controls
Document results to track any decline in performance
Consider adding BSA (0.1-1%) as a carrier protein for very dilute solutions
Following these guidelines ensures optimal antibody performance across experimental applications and maximizes the usable lifespan of this research reagent .
The Acetyl-HIST1H1B (K16) Antibody provides a powerful tool for investigating the complex crosstalk between different epigenetic modifications:
Sequential ChIP (Re-ChIP) approaches:
First immunoprecipitate with Acetyl-HIST1H1B (K16) Antibody
Then perform a second IP with antibodies against other modifications:
Other histone marks (H3K27me3, H3K4me3, etc.)
DNA methylation-associated proteins (MeCP2, DNMT1)
Chromatin remodelers (BRG1, SNF2H)
This reveals genomic regions where multiple modifications co-exist
Combination with advanced genomic technologies:
ChIP-seq with this antibody followed by bioinformatic correlation with:
DNA methylation data (WGBS, RRBS)
Chromatin accessibility profiles (ATAC-seq, DNase-seq)
Other histone modification maps
This allows comprehensive epigenetic landscape visualization
Perturbation experiments:
Protein interaction studies:
IP with Acetyl-HIST1H1B (K16) Antibody followed by mass spectrometry
Identify proteins that specifically interact with this acetylated form
Compare with interactome of non-acetylated HIST1H1B
This reveals readers of this specific modification
These integrated approaches have already begun to reveal how acetylation at K16 of HIST1H1B functions within the broader context of epigenetic regulation, particularly in relation to gene expression patterns in cancer and development .
The Acetyl-HIST1H1B (K16) Antibody is finding increasingly important applications in cancer research and precision medicine:
Biomarker development:
Analysis of acetylation patterns across cancer types reveals tumor-specific signatures
Correlation of HIST1H1B K16 acetylation with:
Clinical outcomes
Treatment responses
Cancer subtypes
Development of diagnostic and prognostic panels incorporating this mark
Therapeutic target identification:
Screening for compounds that modulate this specific acetylation
Determining whether cancer cells with altered HIST1H1B acetylation show differential sensitivities to:
HDAC inhibitors
Bromodomain inhibitors
Other epigenetic therapies
Mechanistic understanding of oncogenesis:
ChIP-seq mapping of this modification in:
Primary tumors vs. normal tissues
Treatment-responsive vs. resistant tumors
Different stages of cancer progression
Correlation with gene expression changes to identify acetylation-responsive cancer genes
Precision medicine applications:
Patient stratification based on acetylation profiles
Selection of epigenetic therapies based on acetylation status
Development of companion diagnostics for epigenetic drugs
Liquid biopsy development:
Detection of acetylated histone fragments in circulation
Monitoring treatment response through changes in acetylation patterns
These applications leverage the ability of the antibody to specifically detect a modification that appears to be dysregulated in various cancers, potentially offering new avenues for cancer diagnosis, monitoring, and treatment .
Despite its utility, the Acetyl-HIST1H1B (K16) Antibody technology faces several limitations that ongoing research aims to address:
Current technical limitations:
Specificity challenges: Potential cross-reactivity with similar acetylated lysines on other histone variants
Sensitivity issues: Detection of low-abundance modifications requires significant cell input
Batch-to-batch variability: Polyclonal nature leads to heterogeneity between productions
Application restrictions: Better validated for some applications (IF, ChIP) than others
Species limitations: Current antibodies primarily validated for human samples
Emerging technological improvements:
Recombinant antibody development:
Single-chain variable fragments with improved specificity
Consistent production without batch variation
Engineered for enhanced affinity
Advanced detection methods:
Super-resolution microscopy compatible antibody conjugates
Mass cytometry (CyTOF) compatible metal-conjugated antibodies
Single-cell epigenomic applications
Multiplexing capacity:
Antibody conjugation with barcoded oligos for simultaneous detection
Integration with spatial transcriptomics technologies
Future research directions:
Development of dual-recognition antibodies that detect both the histone variant and its modification
Creation of synthetic biology tools (e.g., nanobodies, aptamers) as alternatives to traditional antibodies
Engineering of reader domain fusion proteins for specific acetylation recognition
Integration with CRISPR-based technologies for functional interrogation
Expansion of species reactivity to enable comparative studies
Validation standardization:
Establishment of community standards for antibody validation
Development of reference materials with defined acetylation states
Automated analysis pipelines to reduce subjective interpretation
These advancements will address current limitations and expand the utility of Acetyl-HIST1H1B (K16) detection technologies in both basic research and clinical applications .
When troubleshooting weak or absent signals in immunofluorescence experiments with the Acetyl-HIST1H1B (K16) Antibody, consider this systematic approach:
Epitope accessibility issues:
Problem: The acetyl-lysine epitope may be masked due to fixation or chromatin structure
Solution: Implement epitope retrieval by:
Heat-induced retrieval (10mM citrate buffer, pH 6.0)
Extending permeabilization time with 0.2% Triton X-100
Testing alternative fixation methods (methanol vs. paraformaldehyde)
Acetylation level optimization:
Antibody concentration optimization:
Problem: Suboptimal antibody concentration leads to weak signal
Solution: Perform titration experiments:
Test the full recommended range (1:50-1:200)
Include both higher (1:25) and lower (1:400) concentrations
Extend primary antibody incubation to overnight at 4°C
Detection system enhancement:
Problem: Insufficient amplification of signal
Solution: Implement signal amplification methods:
Use tyramide signal amplification (TSA)
Apply biotin-streptavidin amplification systems
Switch to a more sensitive secondary antibody or fluorophore
Technical verification steps:
Confirm antibody viability with dot blot of acetylated peptide
Verify secondary antibody functionality with a different primary antibody
Include positive control samples (HepG2 cells treated with sodium butyrate)
Check microscope settings and fluorophore compatibility
Following this structured approach has resolved weak signal issues in multiple studies using this antibody for immunofluorescence applications .
High background or non-specific binding in ChIP experiments with the Acetyl-HIST1H1B (K16) Antibody can significantly impact data quality. These strategies can help resolve such issues:
Chromatin preparation optimization:
Problem: Inadequate chromatin fragmentation or quality
Solution: Optimize sonication conditions:
Verify fragment size (aim for 200-500bp)
Increase sonication cycles for dense heterochromatin
Add SDS (0.1%) to improve solubilization
Pre-clear chromatin more extensively with protein A/G beads
Blocking and washing stringency:
Problem: Insufficient blocking or washing
Solution: Enhance blocking and washing steps:
Add salmon sperm DNA (100μg/ml) to blocking buffer
Increase BSA concentration in blocking buffer to 5%
Include more stringent wash steps with higher salt concentrations
Extend wash times and increase number of washes
Antibody specificity verification:
Problem: Antibody binding to non-target acetylation sites
Solution: Validate specificity:
Perform peptide competition assays with acetylated and non-acetylated peptides
Compare ChIP-seq profiles with published datasets for similar marks
Validate key findings with alternative antibodies if available
Protocol modifications for high signal-to-noise ratio:
Step | Standard Protocol | Modified Protocol for Reducing Background |
---|---|---|
Chromatin amount | Standard IP | Reduce input chromatin by 25-50% |
Antibody concentration | 5μg per IP | Titrate down to 2-3μg |
Bead volume | Standard | Reduce bead volume by 25% |
Pre-clearing | Once | Double pre-clearing with fresh beads |
Washes | Standard buffers | Include lithium chloride wash step |
Data analysis approaches:
Implement spike-in normalization with foreign DNA
Use IgG control for background subtraction
Apply more stringent peak calling parameters
Compare enrichment to input at known negative regions
These optimizations have been shown to significantly improve signal-to-noise ratios in ChIP experiments with acetylation-specific antibodies like the Acetyl-HIST1H1B (K16) Antibody .
When faced with contradictory results between different lots or sources of the Acetyl-HIST1H1B (K16) Antibody, validation is essential for research reproducibility:
Direct comparison experiments:
Approach: Perform side-by-side experiments with both antibody lots:
Western blot with acetylated and non-acetylated controls
ChIP-qPCR targeting known regions enriched for this mark
Immunofluorescence on the same cell preparations
Analysis: Quantify signals and compare signal-to-noise ratios
Epitope verification:
Approach: Conduct peptide competition assays:
Pre-incubate each antibody lot with:
Acetylated HIST1H1B K16 peptide (specific)
Non-acetylated HIST1H1B peptide (control)
Acetylated peptides from other histone variants (specificity)
Compare binding inhibition patterns
Outcome: Specific antibodies will show inhibition only with the acetylated target peptide
Cross-validation with orthogonal methods:
Approach: Verify key findings with alternative techniques:
Mass spectrometry to directly measure acetylation levels
CUT&RUN or CUT&Tag as alternatives to ChIP
Proximity ligation assays to verify colocalization with readers of acetylated histones
Analysis: Compare results from multiple methodologies
Detailed antibody characterization:
Validation Parameter | Experimental Approach | Expected Outcome |
---|---|---|
Sensitivity | Titration series with known amounts of acetylated protein | Minimum detection threshold |
Specificity | Panel testing against related modifications | Cross-reactivity profile |
Reproducibility | Multiple experiments with different batches | Consistency measures |
Cell type variability | Testing across multiple cell lines | Range of signal patterns |
Documentation and reporting standards:
Record complete antibody information (catalog number, lot number, production date)
Document detailed experimental conditions
Share validation data with the scientific community
Consider validating with reference laboratories
This structured validation approach ensures experimental reproducibility and helps identify the source of contradictory results between different antibody lots or sources .
The integration of single-cell technologies with Acetyl-HIST1H1B (K16) Antibody research represents a frontier in epigenetic analysis:
Single-cell CUT&Tag applications:
Methodology: Adapt the antibody for CUT&Tag protocols at single-cell level:
Optimize antibody concentration for limited cellular material
Test with pA-Tn5 fusion proteins for direct tagmentation
Validate on mixed cell populations with known acetylation differences
Outcome: Maps of K16 acetylation patterns with single-cell resolution
Integration with multi-omics platforms:
Approach: Combine acetylation profiling with other single-cell measurements:
CITE-seq-like approaches (cellular indexing of transcriptomes and epitopes)
Concurrent measurement of acetylation and transcription
Integration with chromatin accessibility data (scATAC-seq)
Analysis: Correlation between acetylation states and cellular phenotypes
Spatial epigenomics applications:
Methodology: Adapt the antibody for spatial detection technologies:
Optimization for Imaging Mass Cytometry
Application in advanced microscopy with DNA-barcoded antibodies
Integration with spatial transcriptomics platforms
Outcome: Spatial maps of acetylation patterns in tissue contexts
Microfluidic implementations:
Technical development: Miniaturize immunoprecipitation protocols:
Droplet-based microfluidic ChIP
Reduction of antibody consumption through microchannels
Automated processing of single cells or small cell numbers
Advantages: Increased throughput, reduced reagent consumption
Computational integration challenges:
Development of analytical pipelines specific to sparse single-cell epigenetic data
Methods for integrating multiple epigenetic marks at single-cell resolution
Trajectory analysis of acetylation changes during cellular processes
These emerging applications leverage the specificity of the Acetyl-HIST1H1B (K16) Antibody while addressing the technical challenges of single-cell analysis, opening new avenues for understanding epigenetic heterogeneity in complex biological systems .
The acetylation of HIST1H1B at lysine 16 has emerging significance in cellular identity changes and developmental processes:
Dynamics during cellular reprogramming:
Observation pattern: HIST1H1B K16 acetylation undergoes dramatic remodeling during:
Induced pluripotent stem cell (iPSC) generation
Direct lineage conversion between differentiated states
Transition from primed to naive pluripotency
Functional significance: Changes in this mark precede and potentially regulate gene expression changes necessary for cell fate transitions
Role in developmental processes:
Tissue-specific patterns: Differential acetylation profiles between:
Embryonic vs. adult tissues
Stem cells vs. committed progenitors
Various differentiated cell types within the same organ
Developmental regulation: Dynamic changes during embryogenesis and organogenesis
Mechanistic investigations:
Writer/eraser enzymes: Identification of specific:
HATs (histone acetyltransferases) responsible for K16 acetylation
HDACs (histone deacetylases) that remove this mark
Reader proteins: Characterization of factors that specifically recognize this modification and mediate downstream effects
Experimental approaches:
ChIP-seq with the Acetyl-HIST1H1B (K16) Antibody at multiple timepoints during differentiation
Manipulation of acetylation levels through:
HDAC inhibitors (sodium butyrate at 30mM has been validated)
Overexpression or depletion of specific HATs/HDACs
Targeted editing of the K16 residue
Correlation with changes in chromatin accessibility and gene expression
Disease relevance:
Alterations in HIST1H1B K16 acetylation patterns in:
Developmental disorders
Regenerative processes after injury
Age-related cellular dysfunction
Understanding these dynamics requires time-course studies with the Acetyl-HIST1H1B (K16) Antibody across different cellular transitions, providing insights into how this specific epigenetic mark contributes to cell fate decisions .
Computational approaches offer powerful tools for understanding the structural and functional consequences of HIST1H1B K16 acetylation:
Molecular dynamics simulations:
Approach: Compare acetylated vs. non-acetylated HIST1H1B:
Simulations of histone tail flexibility and interactions
Calculation of binding energies with DNA and other proteins
Prediction of conformational changes induced by acetylation
Insight: How acetylation alters the biophysical properties of histone-DNA interactions
Integrative structural biology:
Methodology: Combine computational models with experimental data:
Cryo-EM structures of nucleosomes with linker histones
NMR studies of histone tail dynamics
Crosslinking mass spectrometry data
Outcome: Multi-scale models of chromatin with acetylated HIST1H1B
Prediction of reader protein interactions:
Approach: Virtual screening and molecular docking:
Identification of potential bromodomain proteins that recognize K16ac
Simulation of protein-protein interaction networks
Prediction of structural consequences for higher-order chromatin
Application: Design of experiments to validate predicted interactions
Genome-wide modeling of acetylation impact:
Integration with experimental data: Combine ChIP-seq data using the Acetyl-HIST1H1B (K16) Antibody with:
Hi-C chromatin conformation data
DNA methylation profiles
Other histone mark distributions
Outcome: Predictive models of chromatin domain organization
Machine learning applications:
Approach: Train algorithms on:
ChIP-seq data from the Acetyl-HIST1H1B (K16) Antibody
DNA sequence features
Other epigenetic marks
Prediction: Genomic regions likely to contain this modification
Validation: Test predictions with experimental ChIP data