Acetyl-HIST1H2BC (K116) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Overview

Target: Acetylated lysine 116 on human histone H2B type 1-C/E/F/G/I (UniProt: P62807) .
Host Species: Rabbit-derived polyclonal antibody .
Reactivity: Validated for human samples; cross-reactivity with other species is not reported .

A. Western Blot Validation

  • Detects a 14 kDa band in human 293 and A549 cell lysates treated with sodium butyrate, a histone deacetylase inhibitor .

  • Example protocol:

    1. Lysate preparation from sodium butyrate-treated cells.

    2. Primary antibody incubation at 1:100 dilution.

    3. Secondary detection using anti-rabbit IgG-HRP (1:50,000) .

B. Chromatin Immunoprecipitation (ChIP)

  • Validated in HeLa cells treated with sodium butyrate:

    • Immunoprecipitated DNA showed enrichment at the β-globin promoter via qPCR .

    • Demonstrates specificity for acetylated chromatin regions .

C. Immunofluorescence

  • Localizes acetylated HIST1H2BC (K116) to the nucleus in HeLa cells .

  • Protocol highlights:

    • Fixation with 4% formaldehyde.

    • Permeabilization using 0.2% Triton X-100.

    • Secondary antibody: Alexa Fluor 488-conjugated anti-rabbit IgG .

A. Biological Role of HIST1H2BC

  • Core component of nucleosomes, compacting DNA into chromatin .

  • Post-translational acetylation at K116 modulates:

    • Gene transcription (e.g., β-globin promoter activity) .

    • DNA repair and replication .

    • Antimicrobial responses (antibacterial/antifungal activity) .

B. Disease Relevance

  • Dysregulated histone acetylation is linked to cancer, neurodevelopmental disorders, and immune dysfunctions .

  • Sodium butyrate-induced acetylation models epigenetic drug mechanisms .

Immunogen Design

  • Peptide sequence: Acetyl-Lys (116) from human HIST1H2BC .

  • Epitope specificity confirmed via competitive binding assays .

Supplier Information

SupplierCatalog NumberPrice (50 µL)Validation Data Provided
Assay GeniePACO60506$329WB, ChIP, IF
CusabioCSB-PA010403OA116acHU$298ELISA, WB
AbbexaABX354422$315WB, IF/ICC, ChIP

Limitations and Considerations

  • Species Restriction: Reactivity limited to human samples .

  • Modification Dependency: Detects only acetylated K116; untreated cells show no signal .

  • Storage Stability: Avoid repeated freeze-thaw cycles to prevent activity loss .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, orders are dispatched within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for specific delivery information.
Synonyms
H2BC4 antibody; H2BFL antibody; HIST1H2BC; antibody; H2BC6 antibody; H2BFH antibody; HIST1H2BE; antibody; H2BC7 antibody; H2BFG antibody; HIST1H2BF; antibody; H2BC8 antibody; H2BFA antibody; HIST1H2BG; antibody; H2BC10 antibody; H2BFK antibody; HIST1H2BIHistone H2B type 1-C/E/F/G/I antibody; Histone H2B.1 A antibody; Histone H2B.a antibody; H2B/a antibody; Histone H2B.g antibody; H2B/g antibody; Histone H2B.h antibody; H2B/h antibody; Histone H2B.k antibody; H2B/k antibody; Histone H2B.l antibody; H2B/l antibody
Target Names
HIST1H2BC
Uniprot No.

Target Background

Function
Histone H2BC, acetylated at lysine 116, is a core component of the nucleosome. Nucleosomes serve as the fundamental unit of chromatin, wrapping and compacting DNA to regulate its accessibility to cellular machinery. This regulation is essential for various processes including transcription, DNA repair, replication, and chromosomal stability. Histone modifications, including acetylation, play a crucial role in modulating DNA accessibility, often referred to as the 'histone code'. Beyond its role in chromatin structure, acetylated Histone H2BC exhibits broad antibacterial activity. It may contribute to the formation of the functional antimicrobial barrier in the colonic epithelium, as well as the bactericidal activity of amniotic fluid.
Database Links

HGNC: 4757

OMIM: 602798

KEGG: hsa:3017

STRING: 9606.ENSP00000366962

UniGene: Hs.182137

Protein Families
Histone H2B family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Acetyl-HIST1H2BC (K116) Antibody and what is its role in epigenetic research?

The Acetyl-HIST1H2BC (K116) Antibody is a polyclonal antibody raised in rabbits that specifically recognizes histone H2B type 1-C/E/F/G/I acetylated at lysine 116 position. This antibody serves as a critical research tool for studying epigenetic modifications, particularly histone acetylation, which plays a fundamental role in chromatin dynamics and gene regulation. Histones are core components of nucleosomes that wrap and compact DNA into chromatin, thereby limiting DNA accessibility to cellular machinery. Acetylation of histones, including H2B at K116, can alter chromatin structure, typically leading to a more accessible conformation that facilitates transcription factor binding and gene expression. By detecting this specific modification, researchers can investigate how epigenetic patterns correlate with transcriptional states, cellular differentiation, disease progression, and responses to environmental stimuli .

What are the validated applications for the Acetyl-HIST1H2BC (K116) Antibody?

The Acetyl-HIST1H2BC (K116) Antibody has been validated for multiple experimental applications that are essential for epigenetic research. These applications include:

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of acetylated histone H2B

  • Western Blotting (WB): For identifying the presence and relative abundance of acetylated H2B(K116) in protein extracts

  • Immunofluorescence (IF): For visualizing the nuclear localization and distribution patterns of acetylated H2B in intact cells

  • Chromatin Immunoprecipitation (ChIP): For determining the genomic locations where this modification is present

Each application requires specific dilution ratios for optimal results, with Western Blotting typically using dilutions of 1:100-1:1000 and Immunofluorescence requiring dilutions of 1:1-1:10 . These diverse applications make the antibody versatile for investigating different aspects of histone modification biology.

What is the biological significance of histone H2B acetylation at lysine 116?

Histone H2B acetylation at lysine 116 is part of the complex "histone code" that regulates chromatin structure and function. Nucleosomes, composed of DNA wrapped around histone octamers, are the fundamental units of chromatin. By modifying specific amino acid residues on histones through acetylation, methylation, phosphorylation, and other chemical changes, cells can dynamically regulate gene expression and other DNA-templated processes. Acetylation of H2B at K116 specifically affects chromatin compaction, typically promoting a more relaxed chromatin state that is permissive for transcription. This modification is involved in various cellular processes including DNA repair, replication, and transcriptional activation. Understanding the distribution and dynamics of this modification can provide insights into how cells regulate their genomic activities in normal development and disease states .

How can I optimize ChIP protocols specifically for Acetyl-HIST1H2BC (K116) detection in different cell types?

Optimizing ChIP protocols for Acetyl-HIST1H2BC (K116) requires careful consideration of several variables depending on cell type. For standard ChIP protocols with this antibody:

  • Crosslinking optimization: Different cell types may require adjusted formaldehyde concentrations (typically 0.75-1.5%) and incubation times (5-15 minutes). More compact chromatin structures may benefit from dual crosslinking approaches using both formaldehyde and protein-protein crosslinkers like DSG or EGS.

  • Sonication parameters: Cell-type specific chromatin compaction affects optimal sonication conditions. Start with standard conditions (e.g., 15-30 seconds on/30 seconds off cycles for 10-15 cycles) and adjust based on gel electrophoresis results, aiming for fragments between 200-500bp.

  • Antibody concentration: The Acetyl-HIST1H2BC (K116) antibody performs optimally in ChIP when used at concentrations determined by titration. Start with 2-5μg of antibody per ChIP reaction and adjust based on preliminary results.

  • Beads and washing conditions: For polyclonal antibodies like Acetyl-HIST1H2BC (K116), Protein A/G beads are recommended with stringent washing steps (including high salt washes) to reduce background.

  • Positive controls: Include primers for regions known to be enriched for H2B K116 acetylation in your cell type, as well as primers for housekeeping gene promoters as general positive controls for active chromatin .

For difficult cell types or limited samples, consider using ChIP-seq protocols with amplification steps or specialized microChIP approaches that maintain sensitivity while reducing input material requirements.

What are the key technical challenges in distinguishing Acetyl-HIST1H2BC (K116) from other histone H2B acetylation sites in multiplexed assays?

Distinguishing Acetyl-HIST1H2BC (K116) from other H2B acetylation sites presents several technical challenges in multiplexed assays:

Robust controls including peptide blocking experiments, modification-specific standards, and mass spectrometry validation are recommended to ensure correct identification of specific acetylation sites.

How does the distribution of H2B K116 acetylation change during cellular differentiation and what methodologies best capture these dynamics?

The distribution of H2B K116 acetylation undergoes significant remodeling during cellular differentiation, reflecting changes in gene expression programs. Capturing these dynamics requires specialized approaches:

  • Time-course ChIP-seq analysis: Performing ChIP-seq with the Acetyl-HIST1H2BC (K116) antibody at multiple timepoints during differentiation can map the genome-wide redistribution of this mark. Analysis should focus on:

    • Promoter regions of lineage-specific genes

    • Enhancer elements that become activated or repressed

    • Global changes in acetylation patterns across the genome

  • Integrative multi-omics approaches: Combining H2B K116ac ChIP-seq with:

    • RNA-seq to correlate acetylation changes with transcriptional outputs

    • ATAC-seq to assess chromatin accessibility changes

    • ChIP-seq for other histone marks to understand the combinatorial histone code

    • DNA methylation profiling to capture complete epigenetic reprogramming

  • Single-cell epigenomic methods: Recent advances in single-cell ChIP technologies, though still challenging for histone modifications, can reveal cell-to-cell heterogeneity in acetylation patterns during differentiation. Alternative approaches include:

    • CUT&Tag at single-cell resolution

    • Single-cell ATAC-seq combined with computational inference

    • Imaging-based approaches using the Acetyl-HIST1H2BC (K116) antibody for immunofluorescence

  • Pulse-chase experiments: Using metabolic labeling of acetyl groups (e.g., with heavy isotope-labeled acetate) combined with mass spectrometry to determine turnover rates of H2B K116 acetylation during differentiation .

These methodologies collectively provide insights into both the spatial redistribution and temporal dynamics of H2B K116 acetylation during cell fate transitions.

What are the optimal sample preparation protocols for detecting Acetyl-HIST1H2BC (K116) in Western blot applications?

Optimal sample preparation for detecting Acetyl-HIST1H2BC (K116) in Western blot applications requires special consideration due to the nature of nuclear proteins and potential epitope sensitivity:

  • Histone extraction protocol:

    • Use a specialized histone extraction protocol involving acid extraction (typically with 0.2N HCl)

    • Alternative approach: use commercial histone extraction kits that preserve post-translational modifications

    • Include histone deacetylase inhibitors (e.g., sodium butyrate at 5-10mM, TSA at 0.5-1μM) in all buffers

    • Include protease inhibitors to prevent degradation

  • Quantification and loading:

    • Accurately quantify histone concentration using Bradford or BCA assays calibrated for basic proteins

    • Load 10-20μg of total histone extract per lane

    • Use Ponceau S staining to confirm equal loading after transfer

  • Gel electrophoresis considerations:

    • Use high percentage (15-18%) SDS-PAGE gels for optimal histone separation

    • Consider specialized gel systems like Triton-Acid-Urea gels for separation of differently modified histones

    • Run at lower voltage (80-100V) to improve resolution of the 17kDa histone H2B band

  • Transfer and detection optimization:

    • Use PVDF membranes (rather than nitrocellulose) for better protein retention

    • Transfer at lower voltage for longer time (25V overnight) in Tris-glycine buffer with 20% methanol

    • Block with 5% BSA (not milk) to prevent phosphatase activity affecting modifications

    • Dilute primary Acetyl-HIST1H2BC (K116) antibody in range of 1:100-1:1000 in TBST with 3% BSA

    • Incubate with gentle agitation overnight at 4°C for maximum sensitivity

Control samples should include unmodified recombinant H2B and, if possible, samples treated with histone deacetylase inhibitors to increase acetylation signal as positive controls.

How can I troubleshoot specificity issues when using Acetyl-HIST1H2BC (K116) antibody in immunofluorescence experiments?

When troubleshooting specificity issues with Acetyl-HIST1H2BC (K116) antibody in immunofluorescence experiments, consider the following methodological approaches:

  • Validation controls:

    • Peptide competition assay: Pre-incubate the antibody with excess acetylated peptide vs. unmodified peptide

    • Knock-down verification: Use cells with siRNA/CRISPR against H2B or HATs/HDACs affecting K116 acetylation

    • Pharmacological controls: Compare signals in cells treated with HDAC inhibitors (should increase signal) versus HDAC activators (should decrease signal)

  • Fixation optimization:

    • Test multiple fixation methods: 4% paraformaldehyde (10 min), methanol (-20°C, 10 min), or combination fixation

    • Acetylation marks can be sensitive to overfixation; validate optimal fixation times

    • Include post-fixation permeabilization with 0.1-0.5% Triton X-100 to improve nuclear antigen accessibility

  • Antibody incubation conditions:

    • Use recommended dilution (1:1-1:10) in antibody diluent containing 1% BSA

    • Extend primary antibody incubation to overnight at 4°C to improve signal-to-noise ratio

    • Test different blocking solutions (3% BSA, 10% normal serum, commercial blockers)

  • Reducing background:

    • Include 0.1-0.3% Triton X-100 in blocking and antibody diluents

    • Increase washing steps (at least 3x10 minutes with agitation)

    • Use Sudan Black B (0.1% in 70% ethanol) to reduce autofluorescence

    • Consider signal amplification methods like tyramide signal amplification if specific signal is weak

  • Co-staining verification:

    • Co-stain with antibodies against other histone marks with known relationships to H2B K116ac

    • Use DAPI as nuclear counterstain to verify nuclear localization

    • Consider super-resolution microscopy techniques for more precise localization

Document all optimization steps systematically, maintaining all other variables constant while changing one parameter at a time.

What are the critical parameters for achieving reproducible results in ChIP-seq experiments using Acetyl-HIST1H2BC (K116) antibody?

Achieving reproducible ChIP-seq results with Acetyl-HIST1H2BC (K116) antibody requires careful attention to several critical parameters throughout the experimental workflow:

  • Experimental design and sample preparation:

    • Include biological replicates (minimum n=3) for statistical robustness

    • Maintain consistent cell culture conditions (density, passage number, treatments)

    • Standardize crosslinking conditions (1% formaldehyde, 10 minutes at room temperature)

    • Optimize chromatin shearing to achieve 200-500bp fragments consistently

    • Include input controls, IgG controls, and spike-in normalization controls

  • Immunoprecipitation optimization:

    • Determine optimal antibody amount through titration experiments (typically 2-5μg)

    • Use consistent antibody lots whenever possible

    • Maintain consistent IP conditions (buffer composition, incubation time, temperature)

    • Include HDAC inhibitors in all buffers to preserve acetylation status

    • Implement rigorous washing protocols to reduce background

  • Library preparation considerations:

    • Quantify ChIP DNA accurately using qPCR or fluorometric methods

    • Use consistent amounts of starting material for library preparation

    • Minimize PCR cycles to reduce amplification bias

    • Include library preparation controls

    • Use unique molecular identifiers (UMIs) to identify PCR duplicates

  • Data analysis and quality control metrics:

    • Implement standardized computational pipeline for all samples

    • Assess quality metrics: fragment size distribution, library complexity, mapping rates

    • Evaluate enrichment using metrics like fraction of reads in peaks (FRiP)

    • Use appropriate peak calling algorithms (e.g., MACS2 with parameters optimized for histone modifications)

    • Implement batch correction if processing multiple sample sets

  • Validation strategies:

    • Validate key peaks by ChIP-qPCR in independent samples

    • Compare with published datasets for similar cell types/conditions

    • Correlate acetylation patterns with gene expression data

    • Validate biological findings using orthogonal approaches

A detailed protocol with all critical parameters should be established and strictly followed for all experiments to ensure reproducibility between different operators and over time.

How can I effectively use Acetyl-HIST1H2BC (K116) antibody in combination with other histone modification antibodies for multi-parameter epigenetic analysis?

Implementing multi-parameter epigenetic analysis with Acetyl-HIST1H2BC (K116) antibody alongside other histone modification antibodies requires careful methodological planning:

  • Sequential ChIP (Re-ChIP) approach:

    • Perform initial ChIP with Acetyl-HIST1H2BC (K116) antibody

    • Elute chromatin complexes under mild conditions (glycine buffer pH 2.5, 10mM DTT, or competing peptides)

    • Perform second round of ChIP with antibodies against other modifications

    • Use optimized elution buffers that release the first antibody-chromatin complexes without denaturing histones

    • Include controls where the same antibody is used in both rounds to establish baseline re-ChIP efficiency

  • Co-staining in immunofluorescence microscopy:

    • Select primary antibodies raised in different host species (Acetyl-HIST1H2BC (K116) antibody is rabbit-derived)

    • Use directly conjugated secondary antibodies with minimal spectral overlap

    • Implement appropriate blocking steps to prevent cross-reactivity

    • Include single-stained controls for establishing compensation settings

    • Consider advanced imaging techniques like STORM or STED for high-resolution co-localization studies

  • Multi-omic data integration approaches:

    • Perform parallel ChIP-seq experiments for different modifications

    • Use consistent chromatin preparation and bioinformatic pipelines

    • Apply correlation analyses and machine learning approaches to identify combinatorial patterns

    • Implement visualization tools that allow simultaneous viewing of multiple epigenetic marks

    • Correlate with expression data (RNA-seq) and chromatin accessibility (ATAC-seq)

  • Mass spectrometry-based approaches:

    • Couple immunoprecipitation with mass spectrometry (IP-MS)

    • Enrich for modified histones using Acetyl-HIST1H2BC (K116) antibody

    • Analyze co-occurring modifications by MS/MS

    • Quantify relative abundances of different modification patterns

    • Implement top-down proteomics to preserve intact histone molecules for combinatorial analysis

Document all optimization steps systematically to establish robust protocols that can be shared across research groups and contribute to reproducible epigenetic research.

What are the recommended controls for validating Acetyl-HIST1H2BC (K116) antibody specificity in different experimental contexts?

Rigorous validation of Acetyl-HIST1H2BC (K116) antibody specificity requires implementing various controls tailored to each experimental application:

  • Universal controls applicable across methods:

    • Peptide competition assay: Pre-incubate antibody with acetylated K116 peptide versus unmodified peptide

    • Genetic manipulation controls:

      • CRISPR/Cas9 K116R mutation (non-acetylatable lysine to arginine)

      • Knockdown/knockout of relevant histone acetyltransferases

    • Pharmacological controls:

      • HDAC inhibitors (sodium butyrate, TSA, etc.) should increase signal

      • HAT inhibitors should decrease signal

  • Western blotting specific controls:

    • Recombinant protein controls: Use unmodified and in vitro acetylated H2B

    • Molecular weight verification: H2B runs at approximately 17 kDa

    • Alternative antibody comparison: Use commercially available alternative antibodies against the same modification

  • ChIP and ChIP-seq specific controls:

    • Input DNA control: 5-10% of starting chromatin material

    • IgG negative control: Non-specific IgG from same species as primary antibody

    • Spike-in normalization control: Add chromatin from alternative species

    • Positive genomic locus controls: Regions known to contain H2B K116ac

    • Negative genomic locus controls: Regions known to lack H2B K116ac

  • Immunofluorescence specific controls:

    • Secondary antibody-only control: Omit primary antibody

    • Pre-immune serum control: If available from antibody production

    • Acetylation site mutagenesis: Transfect cells with K116R mutant constructs

    • Subcellular localization verification: Nuclear localization consistent with histone distribution

  • Flow cytometry specific controls:

    • Fluorescence-minus-one (FMO) controls

    • Isotype controls: Matched concentration of irrelevant rabbit IgG

    • Permeabilization controls: Compare different nuclear permeabilization methods

These controls should be systematically implemented and documented with each new antibody lot and experimental system to ensure data reliability.

How can I quantitatively analyze changes in H2B K116 acetylation levels in response to epigenetic drug treatments?

Quantitative analysis of H2B K116 acetylation changes in response to epigenetic drugs requires multi-faceted analytical approaches:

  • Western blot quantification method:

    • Use increasing protein loading concentrations to establish linear detection range

    • Normalize Acetyl-HIST1H2BC (K116) signal to total H2B or loading controls (e.g., GAPDH)

    • Implement triplicate biological samples with technical replicates

    • Use digital imaging systems rather than film for wider dynamic range

    • Apply appropriate statistical tests comparing treatment vs. control conditions

  • Flow cytometry approach:

    • Optimize cell fixation and permeabilization for nuclear antigens

    • Use Acetyl-HIST1H2BC (K116) antibody at optimal dilution (typically 1:100)

    • Include appropriate fluorophore-conjugated secondary antibody

    • Calculate median fluorescence intensity (MFI) and perform statistical analysis

    • Consider dual staining with cell cycle markers to assess cell cycle-dependent effects

  • ChIP-qPCR quantification:

    • Select genomic regions of interest based on preliminary data or literature

    • Design qPCR primers for these regions (amplicons 80-150bp)

    • Calculate enrichment as percentage of input or fold enrichment over IgG control

    • Use multiple reference regions including positive and negative controls

    • Apply appropriate statistical tests for comparing treatment conditions

  • Global ChIP-seq analysis pipeline:

    • Normalize read counts appropriately (spike-in normalization recommended)

    • Identify differential binding sites between treatment conditions

    • Quantify changes in peak intensity, width, and genomic distribution

    • Correlate with changes in gene expression

    • Perform pathway enrichment analysis on genes associated with altered peaks

  • Immunofluorescence microscopy quantification:

    • Capture images with consistent exposure settings

    • Measure nuclear fluorescence intensity using image analysis software

    • Analyze population distributions rather than just mean values

    • Consider single-cell heterogeneity in drug response

    • Plot results as cumulative frequency distributions or violin plots

Time-course experiments are particularly valuable for understanding the dynamics of acetylation changes in response to epigenetic drugs, with optimal timepoints established through pilot studies.

What bioinformatics pipelines are most appropriate for analyzing ChIP-seq data generated with Acetyl-HIST1H2BC (K116) antibody?

Analyzing ChIP-seq data generated with the Acetyl-HIST1H2BC (K116) antibody requires specialized bioinformatics pipelines tailored to histone modification profiles:

  • Data pre-processing and quality control:

    • FastQC for initial quality assessment of raw reads

    • Trimmomatic or Cutadapt for adapter and quality trimming

    • Alignment using Bowtie2 or BWA with appropriate parameters for short reads

    • Remove PCR duplicates using Picard or samtools markdup

    • Filter for uniquely mapped reads with MAPQ score ≥30

    • Generate normalized bigWig files for visualization (use spike-in normalization if available)

  • Peak calling approaches:

    • Use MACS2 with histone-specific parameters (--broad flag)

    • Alternative specialized algorithms: SICER2 or epic2 for broad histone marks

    • Implement IDR (Irreproducible Discovery Rate) analysis for replicate consistency

    • Consider differential binding analysis using DiffBind or MAnorm packages

    • Filter peaks based on fold enrichment (typically >2-fold over input)

  • Genomic feature association and annotation:

    • Annotate peaks relative to genomic features using HOMER, ChIPseeker, or GREAT

    • Generate aggregate profiles and heatmaps around transcription start sites using deepTools

    • Calculate enrichment at promoters, gene bodies, and enhancers

    • Integrate with chromatin state annotations (if available for your cell type)

    • Perform motif enrichment analysis to identify associated transcription factors

  • Integrative analysis with other data types:

    • Correlate with RNA-seq data to assess functional impact on gene expression

    • Integrate with other histone modification ChIP-seq datasets

    • Compare with chromatin accessibility data (ATAC-seq, DNase-seq)

    • Use ChromHMM or other segmentation tools for chromatin state analysis

    • Visualize in genome browsers (UCSC, IGV) alongside other epigenomic tracks

  • Specialized analyses for histone acetylation patterns:

    • Analyze distribution relative to known enhancers and super-enhancers

    • Assess correlation with transcriptional activity levels

    • Compare patterns with other acetylation marks like H3K27ac

    • Quantify changes in acetylation breadth and intensity after treatments

    • Implement machine learning approaches to identify combinatorial patterns

Document all analysis parameters and software versions to ensure reproducibility, and validate key findings with alternative analytical approaches.

How can I optimize the signal-to-noise ratio when using Acetyl-HIST1H2BC (K116) antibody in challenging cell types or limited samples?

Optimizing signal-to-noise ratio for Acetyl-HIST1H2BC (K116) antibody in challenging contexts requires implementing targeted technical adjustments:

  • Sample preparation optimization:

    • For limited cell numbers: Scale down protocols while maintaining reagent ratios

    • For difficult-to-lyse cells: Implement dual fixation with DSG/formaldehyde

    • For tissues: Optimize tissue disaggregation and fixation protocols

    • For all samples: Add protease and HDAC inhibitors early in processing

    • Minimize steps: Reduce handling to prevent epitope degradation

  • Antibody incubation conditions:

    • Extended incubation: 16-24 hours at 4°C with gentle rotation

    • Optimized buffer composition: Include 0.1% Triton X-100 and 100mM NaCl

    • Sequential antibody approach: Use indirect detection methods

    • Increase antibody concentration: For challenging samples, use higher concentration (1:50)

    • Reduce buffer volume: Maintain effective antibody concentration in minimal volumes

  • Background reduction strategies:

    • Extensive blocking: Use 5% BSA or commercial blockers for 2+ hours

    • Pre-clear lysates: With beads alone before adding antibody

    • Extensive washing: Increase wash number and stringency

    • Use monovalent blocking reagents: Fab fragments to block Fc receptors

    • Filter secondary antibodies: Pre-adsorb against cellular components

  • Detection sensitivity enhancement:

    • Signal amplification: TSA (tyramide signal amplification) for IF

    • Enhanced chemiluminescence: Super-signal reagents for Western blots

    • Optimized imaging parameters: Extended exposure times, Z-stack acquisition

    • Alternative detection systems: Near-IR fluorescent secondaries

    • Microfluidic approaches: For ultra-low cell numbers

  • Combined approaches for ultra-low input samples:

    • CUT&Tag protocol adaptation: Modified for acetylation marks

    • Low-input ChIP-seq protocols: Using carrier DNA/chromatin

    • Microfluidic devices: For single-cell epigenomics

    • Linear amplification methods: To maintain representation

    • Super-resolution microscopy: For direct visualization

Document all optimization steps systematically to establish robust protocols that can be applied consistently across diverse experimental conditions.

What are the common technical artifacts in ChIP-seq experiments with Acetyl-HIST1H2BC (K116) antibody and how can they be identified and mitigated?

ChIP-seq experiments with Acetyl-HIST1H2BC (K116) antibody can produce several technical artifacts that require systematic identification and mitigation:

  • Antibody cross-reactivity artifacts:

    • Identification: Unexpected peaks in regions lacking other active marks; signals in knockout/knockdown controls

    • Mitigation: Validate antibody with peptide competition assays; use multiple antibody lots; implement strict cutoffs for peak calling

  • Chromatin preparation issues:

    • Identification: Inconsistent fragment size distribution; over-represented regions in input samples

    • Mitigation: Optimize sonication parameters; filter out samples with improper fragmentation; use enzymatic fragmentation alternatives

  • PCR amplification bias:

    • Identification: GC content bias in peak distribution; excessive duplicate reads

    • Mitigation: Minimize PCR cycles; use UMIs to identify duplicates; employ GC-bias correction in analysis

  • Batch effects between experiments:

    • Identification: Principal component analysis shows separation by batch rather than condition

    • Mitigation: Process experimental and control samples together; implement spike-in normalization; use batch correction algorithms

  • Sequencing depth artifacts:

    • Identification: Correlation between peak numbers and sequencing depth

    • Mitigation: Standardize sequencing depth; implement subsampling analysis; use saturation analysis to determine optimal depth

  • False positives from highly accessible regions:

    • Identification: Enrichment at promoters of highly expressed genes regardless of treatment

    • Mitigation: Compare with ATAC-seq or DNase-seq data; normalize to input and IgG controls

  • Broad versus narrow peak calling issues:

    • Identification: Fragmented peaks where continuous domains are expected

    • Mitigation: Use histone-specific peak callers (SICER, epic2); implement appropriate merging parameters

  • Cell cycle heterogeneity effects:

    • Identification: Bimodal distributions in replicate experiments

    • Mitigation: Synchronize cells when possible; analyze cell cycle subpopulations separately

  • Epitope masking artifacts:

    • Identification: Inconsistent results between antibody lots; regions lacking signal despite known activity

    • Mitigation: Use alternative antibodies or approaches (e.g., mass spectrometry) for validation

Systematic quality control metrics, including FRiP (Fraction of Reads in Peaks), NSC/RSC (Normalized/Relative Strand Cross-correlation), IDR (Irreproducible Discovery Rate), and PBC (PCR Bottleneck Coefficient) should be calculated and reported for all datasets.

How can I integrate Acetyl-HIST1H2BC (K116) ChIP-seq data with other epigenomic datasets to gain comprehensive insights into gene regulation?

Integrating Acetyl-HIST1H2BC (K116) ChIP-seq data with other epigenomic datasets requires sophisticated computational approaches:

  • Multi-omic data integration framework:

    • Correlation analysis: Calculate pairwise correlations between H2B K116ac and other histone modifications

    • Co-occurrence patterns: Identify genomic regions with specific combinations of marks

    • Chromatin state segmentation: Use tools like ChromHMM or Segway to define chromatin states

    • Trajectory analysis: For developmental or treatment time-course data

    • Network-based approaches: Construct gene regulatory networks incorporating epigenetic data

  • Integration with transcriptomic data:

    • Expression correlation: Associate H2B K116ac patterns with gene expression levels

    • Differential analysis: Compare changes in acetylation with changes in expression

    • Regulatory element assignment: Use correlation patterns to link enhancers to target genes

    • Splicing analysis: Investigate relationship between gene body acetylation and splicing patterns

    • Transcription factor binding correlation: Integrate with TF ChIP-seq data

  • Chromatin accessibility integration:

    • Peak overlap analysis: Compare with ATAC-seq or DNase-seq peaks

    • Nucleosome positioning: Correlate with MNase-seq data

    • Footprinting analysis: Identify TF binding within accessible regions

    • Enhancer prediction: Identify active enhancers using H2B K116ac and accessibility data

    • Insulator elements: Analyze boundaries between acetylation domains

  • 3D genome organization context:

    • TAD (Topologically Associated Domain) analysis: Examine distribution relative to TAD boundaries

    • Chromatin interaction data: Integrate with Hi-C or ChIA-PET data

    • Enhancer-promoter interactions: Correlate with chromatin conformation capture data

    • Nuclear compartmentalization: Analyze relationship with A/B compartments

    • Phase separation domains: Correlate with markers of biomolecular condensates

  • Visualization and interpretation tools:

    • Genome browsers: UCSC, IGV, WashU Epigenome Browser for multiple track visualization

    • Heat maps and aggregate plots: deepTools, EnrichedHeatmap for pattern visualization

    • 3D visualization: Juicebox, HiGlass for chromatin conformation data

    • Network visualization: Cytoscape for gene regulatory networks

    • Interactive dashboards: Create R Shiny apps for data exploration

This integrative approach enables identification of novel regulatory principles beyond what can be observed with any single epigenomic dataset.

What statistical approaches are most appropriate for quantifying differential Acetyl-HIST1H2BC (K116) patterns between experimental conditions?

Appropriate statistical approaches for analyzing differential Acetyl-HIST1H2BC (K116) patterns require careful consideration of the experimental design and data characteristics:

  • Differential binding analysis frameworks:

    • DiffBind/edgeR approach: Uses negative binomial models for count data

    • DESeq2 adaptation: Alternative method using variance stabilization

    • MACS2 with bdgdiff: For directly comparing treatment conditions

    • ChIPComp method: Specifically designed for histone modification comparisons

    • MMDiff approach: For analyzing shape changes in modification patterns

  • Normalization considerations:

    • Spike-in normalization: Essential when global changes are expected

    • Input normalization: Corrects for genomic biases

    • Quantile normalization: When comparing datasets with similar distributions

    • TMM (Trimmed Mean of M-values): Robust for ChIP-seq comparisons

    • CSAW approach: Uses sliding windows for flexible region definition

  • Peak-based versus bin-based analyses:

    • Peak-centric approaches: Define consensus peaksets across conditions

    • Bin-based approaches: Divide genome into bins for unbiased analysis

    • Dynamic window approaches: SICER or MACS2 with variable window sizes

    • Signal profile analysis: Compare shape and intensity of signals

    • AUC (Area Under Curve) methods: For quantifying total enrichment

  • Statistical testing frameworks:

    • Multiple hypothesis testing correction: Benjamini-Hochberg FDR control

    • Empirical Bayes methods: For improved variance estimation

    • Permutation testing: For distribution-free hypothesis testing

    • Bayesian approaches: For integrating prior information

    • Meta-analysis methods: For combining results across replicates or studies

  • Accounting for biological confounders:

    • Batch effect correction: Using ComBat or RUV methods

    • Cell cycle correction: Regressing out cell cycle effects

    • Controlling for DNA accessibility: Normalizing to ATAC-seq signal

    • GC content correction: Accounting for sequencing biases

    • Covariates in statistical models: Incorporating known biological variables

For all approaches, implement adequate quality control steps, including assessment of replicate consistency (using IDR or correlation analysis) and sensitivity analysis to ensure robustness of findings.

What are the emerging applications of Acetyl-HIST1H2BC (K116) antibody in single-cell epigenomic studies?

Emerging applications of Acetyl-HIST1H2BC (K116) antibody in single-cell epigenomics represent cutting-edge developments in the field:

  • Single-cell CUT&Tag adaptations:

    • scCUT&Tag protocol: Modified to detect H2B K116ac in individual cells

    • Microfluidic implementations: For increased throughput and reduced reagent usage

    • Combinatorial indexing approaches: For massively parallel single-cell profiling

    • Computational deconvolution: Methods to address sparsity in single-cell data

    • Integration with scRNA-seq: Multi-omic profiling of the same cells

  • Imaging-based single-cell epigenomics:

    • Immunofluorescence with super-resolution microscopy: For spatial distribution analysis

    • Mass cytometry (CyTOF) adaptations: For high-parameter protein-level analysis

    • Imaging mass cytometry: For tissue section analysis with spatial resolution

    • CODEX multiplexed imaging: For highly multiplexed epitope detection

    • Live-cell imaging approaches: Using Fab fragments for dynamics studies

  • Advanced computational frameworks:

    • Trajectory inference methods: Adapted for epigenomic data

    • Transfer learning approaches: Leverage RNA-seq data to interpret sparse epigenetic data

    • Imputation strategies: Addressing technical dropout in single-cell data

    • Multi-modal data integration: Combining with other single-cell assays

    • Spatial reconstruction methods: For tissue-level epigenetic patterns

  • Novel biological applications:

    • Heterogeneity in cancer: Identifying epigenetically distinct subpopulations

    • Developmental epigenetics: Mapping epigenetic changes during differentiation

    • Response to therapy: Single-cell drug response monitoring

    • Cellular reprogramming studies: Tracking epigenetic remodeling

    • Aging research: Analyzing epigenetic drift at single-cell resolution

  • Technical innovations for limited samples:

    • Nano-ChIP approaches: Scaled-down protocols for minimal input

    • Barcode-enabled antibody detection: For multiplexed epitope detection

    • Split-pool barcoding strategies: For massively parallel processing

    • Microfluidic droplet systems: For ultra-low-input processing

    • Enzymatic amplification methods: For signal enhancement with minimal bias

These emerging applications are rapidly evolving and require careful validation, including comparisons to bulk techniques and complementary approaches for comprehensive characterization of epigenetic states at single-cell resolution.

How can machine learning approaches be applied to analyze complex patterns of H2B K116 acetylation in large-scale epigenomic datasets?

Machine learning approaches offer powerful frameworks for analyzing complex patterns of H2B K116 acetylation in large-scale epigenomic datasets:

  • Supervised learning for pattern recognition:

    • Classification algorithms: Identify genomic regions with distinct acetylation signatures

    • Regression models: Predict gene expression levels from H2B K116ac patterns

    • Feature importance analysis: Identify most informative genomic features

    • Ensemble methods: Combine multiple prediction algorithms for improved accuracy

    • Deep learning CNN approaches: Capture complex spatial patterns in ChIP-seq signal

  • Unsupervised learning for pattern discovery:

    • Clustering algorithms: Identify distinct acetylation patterns across the genome

    • Dimensionality reduction: t-SNE, UMAP for visualizing complex relationships

    • Self-organizing maps: For chromatin state identification

    • Latent variable models: Capture underlying structure in epigenetic data

    • Autoencoders: For feature extraction and noise reduction

  • Integration of multi-omic datasets:

    • Multi-modal deep learning: Jointly model RNA-seq, ChIP-seq, ATAC-seq

    • Transfer learning: Apply knowledge from one dataset to another

    • Domain adaptation: Adjust models between different cell types or conditions

    • Attention mechanisms: Focus on most relevant features across datasets

    • Graph neural networks: Model relationships between genomic elements

  • Interpretability approaches:

    • Feature attribution methods: SHAP, integrated gradients for model interpretation

    • Rule extraction techniques: Derive biological rules from complex models

    • Visualization of learned representations: Understanding latent spaces

    • Benchmark against known biology: Validate findings with established knowledge

    • Ablation studies: Test importance of specific features

  • Advanced applications:

    • Generative models: Predict effects of perturbations on acetylation patterns

    • Time-series modeling: Capture dynamics of acetylation changes

    • Causal inference: Identify drivers of acetylation changes

    • Domain-specific models: Tailored to specific biological contexts

    • Federated learning: Combine datasets across multiple studies while preserving privacy

Implementation requires careful cross-validation approaches, external validation datasets, and integration with biological domain knowledge to ensure that machine learning insights are biologically meaningful and not artifacts of technical biases.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.