Acetyl-HIST1H2AG (K5) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

Target: Histone H2A acetylated at lysine 5 (H2AK5ac)
Synonyms: H2AC11, HIST1H2AG, H2AFP, H2A.1, Histone H2A/p

Key Features

PropertyDetails
Host SpeciesRabbit (monoclonal [EP856Y] or polyclonal)
ClonalityMonoclonal (ab45152) or Polyclonal (PACO00155, CAC15169)
ReactivityHuman, Mouse, Rat, Monkey
ApplicationsWB, IP, IHC-P, ICC/IF
Predicted Band Size14 kDa
Observed Band Size14–15 kDa (variability due to post-translational modifications)

Western Blot (WB)

  • Protocol: Detects H2AK5ac in HeLa (TSA-treated) and C6 rat glioma cell lysates at 1:1,000–1:200,000 dilution .

  • Key Findings:

    • A 14 kDa band corresponds to acetylated H2A, with an additional 34 kDa band observed in some conditions, likely due to dimerization or cross-reactivity .

    • Validated under reducing conditions with TSA (trichostatin A) treatment to enhance acetylation signals .

Immunohistochemistry (IHC)

  • Protocol: Optimized for paraffin-embedded tissues (human colon, breast cancer, liver) using heat-mediated antigen retrieval in EDTA buffer .

  • Key Findings:

    • Strong nuclear staining in cancer tissues, correlating with active transcription sites .

    • Negative controls (PBS or isotype-matched IgG) show no background signal .

Immunocytochemistry/Immunofluorescence (ICC/IF)

  • Protocol: Validated in C6 glioma cells using 4% paraformaldehyde fixation and 0.1% Triton X-100 permeabilization .

  • Key Findings:

    • Co-localization with DAPI confirms nuclear specificity .

    • TSA treatment increases signal intensity, confirming antibody specificity .

Research Implications

  • Epigenetic Regulation: H2AK5ac is associated with transcriptional activation and chromatin relaxation, making this antibody critical for studying gene expression dynamics .

  • Disease Relevance: Elevated H2AK5ac levels are observed in malignancies (e.g., breast cancer) and neurodegenerative disorders, suggesting diagnostic or therapeutic potential .

  • Technical Considerations:

    • Monoclonal antibodies (e.g., ab45152) offer higher specificity, while polyclonal versions (e.g., PACO00155) may detect broader epitopes .

    • Cross-reactivity with non-acetylated H2A is negligible, as confirmed by isotype controls .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery information.
Synonyms
H2AC11 antibody; H2AFP antibody; HIST1H2AG; antibody; H2AC13 antibody; H2AFC antibody; HIST1H2AI; antibody; H2AC15 antibody; H2AFD antibody; HIST1H2AK; antibody; H2AC16 antibody; H2AFI antibody; HIST1H2AL; antibody; H2AC17 antibody; H2AFN antibody; HIST1H2AMHistone H2A type 1 antibody; H2A.1 antibody; Histone H2A/ptl antibody
Target Names
HIST1H2AG
Uniprot No.

Target Background

Function
Histone H2A is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Consequently, histones play a crucial role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Database Links

HGNC: 4737

OMIM: 602787

KEGG: hsa:8329

STRING: 9606.ENSP00000352627

UniGene: Hs.134999

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is the significance of histone H2A lysine 5 acetylation in epigenetic regulation?

Acetylation of lysine 5 on histone H2A represents a key epigenetic modification associated with gene activation and transcriptional regulation. This post-translational modification alters chromatin structure by reducing the positive charge of histones, thereby weakening histone-DNA interactions and creating a more accessible chromatin conformation. The presence of this specific acetylation mark is particularly important for facilitating the binding of transcription factors and other regulatory proteins to DNA. Histone acetylation plays a crucial role in regulating gene expression patterns, and dysregulation of this process has been implicated in various diseases, including cancer and neurological disorders . Researchers investigating this modification can gain valuable insights into fundamental cellular processes and potentially identify new therapeutic targets for intervention.

How does an Acetyl-HIST1H2AG (K5) antibody differ from other histone modification antibodies?

Acetyl-HIST1H2AG (K5) antibodies specifically recognize histone H2A that is acetylated at the lysine 5 position, distinguishing this modification from other acetylation sites on H2A or from modifications on other histone variants. While many histone modification antibodies target common modifications across multiple histone variants, Acetyl-HIST1H2AG (K5) antibodies are highly specific to the acetylated lysine 5 residue on the HIST1H2AG variant of histone H2A. This specificity is crucial when investigating the unique functional roles of particular histone modifications in different cellular contexts. Unlike antibodies targeting other modifications such as methylation or phosphorylation, acetylation-specific antibodies like Acetyl-HIST1H2AG (K5) require careful validation to ensure they don't cross-react with unacetylated histones or other acetylated lysine residues . This specificity allows researchers to precisely map the distribution of this particular modification across the genome.

What techniques can be used to validate the specificity of an Acetyl-HIST1H2AG (K5) antibody?

Validating antibody specificity is essential before proceeding with experimental applications. For Acetyl-HIST1H2AG (K5) antibodies, multiple complementary approaches should be employed:

  • Peptide array testing: This technique involves testing the antibody against a panel of modified and unmodified histone peptides at various concentrations. As demonstrated in the validation of similar H2A acetyl K5 antibodies, the antibody should show strong affinity for the target peptide containing acetylated lysine 5 on H2A, with minimal binding to unrelated modifications .

  • Western blotting with controls: Compare samples treated with and without histone deacetylase inhibitors like Trichostatin A. As shown in validation studies, antibody reactivity should significantly increase in treated samples due to hyperacetylation of histones .

  • Immunofluorescence microscopy: Similar to Western blotting, compare samples with and without histone deacetylase inhibitor treatment to confirm increased nuclear staining in treated samples .

  • Blocking peptide competition: Pre-incubate the antibody with increasing concentrations of the specific acetylated peptide before application in your experimental procedure. Signal reduction confirms specificity.

  • Testing across multiple cell types and species: Evaluate reactivity across different cell lines and organisms to confirm conservation of the epitope and consistent binding patterns .

What are the optimal conditions for using Acetyl-HIST1H2AG (K5) antibody in Western blotting?

For optimal Western blotting results with Acetyl-HIST1H2AG (K5) antibody, consider the following methodology based on validated protocols:

  • Sample preparation: Extract histones using acid extraction methods to enrich for histone proteins. Alternatively, use whole cell lysates prepared with RIPA buffer supplemented with protease inhibitors and histone deacetylase inhibitors (e.g., sodium butyrate).

  • Gel electrophoresis: Use high percentage (15-18%) SDS-PAGE gels to resolve low molecular weight histone proteins effectively. Load 10-20 μg of whole cell lysate or 1-2 μg of purified histones per lane.

  • Transfer conditions: Employ PVDF membranes (0.2 μm pore size) and transfer at lower voltage for longer duration to ensure efficient transfer of small histone proteins.

  • Blocking: Block membranes with 5% non-fat dry milk in TBST for 1 hour at room temperature to minimize background .

  • Antibody dilution: Dilute primary antibody to 1:2000-1:4000 in blocking buffer and incubate overnight at 4°C .

  • Controls: Include both positive controls (cells treated with histone deacetylase inhibitors like Trichostatin A at 500 ng/ml for 4 hours) and negative controls (untreated cells) .

  • Expected results: Anticipate bands at approximately 14-15 kDa, corresponding to the molecular weight of histone H2A .

  • Interpretation: Quantify band intensity relative to total H2A to assess changes in acetylation levels across experimental conditions.

How can I optimize Acetyl-HIST1H2AG (K5) antibody for chromatin immunoprecipitation (ChIP) experiments?

Optimizing ChIP protocols for Acetyl-HIST1H2AG (K5) antibody requires attention to several critical parameters:

  • Crosslinking conditions: For histone modifications, use 1% formaldehyde for 10 minutes at room temperature. Excessive crosslinking can mask epitopes and reduce antibody accessibility.

  • Chromatin fragmentation: Sonicate chromatin to fragments between 200-500 bp. Monitor fragmentation efficiency by agarose gel electrophoresis before proceeding.

  • Antibody amount: Start with 2-5 μg of antibody per ChIP reaction. Titrate to determine optimal amounts for your specific experimental conditions.

  • Beads selection: Use protein A/G magnetic beads for rabbit-derived antibodies. Pre-clear chromatin with beads before adding antibody to reduce non-specific binding.

  • Washing conditions: Employ stringent washing conditions with increasing salt concentrations to minimize background while preserving specific interactions.

  • Controls: Include:

    • Input chromatin (non-immunoprecipitated sample)

    • IgG negative control (same species as the primary antibody)

    • Positive control antibody (targeting abundant histone mark)

    • Spike-in controls for quantitative analysis

  • Validation approaches: Confirm enrichment at known acetylation-rich regions (e.g., active promoters) using qPCR before proceeding to sequencing .

  • Sequential ChIP considerations: For co-occupancy studies with other histone marks, consider sequential ChIP approaches with careful epitope preservation between immunoprecipitation steps.

What is the CUT&Tag2for1 method, and how can it be applied with Acetyl-HIST1H2AG (K5) antibody?

The CUT&Tag2for1 method represents an innovative approach for simultaneously profiling multiple chromatin features in small samples or single cells:

  • Basic principle: CUT&Tag2for1 involves mixing two antibodies targeting distinct chromatin features followed by antibody-directed transposase tethering. After tagmentation, computational signal deconvolution separates the profiles based on fragment size distributions .

  • Application with Acetyl-HIST1H2AG (K5) antibody: This antibody can be paired with an antibody recognizing a contrasting chromatin state (e.g., H3K27me3 for repressive domains) in the CUT&Tag2for1 protocol. The acetylation mark typically produces smaller fragment sizes compared to repressive marks.

  • Protocol adaptation:

    • Mix equal amounts of Acetyl-HIST1H2AG (K5) and a repressive mark antibody (e.g., H3K27me3)

    • Follow standard CUT&Tag protocol with low-salt CUTAC conditions

    • Perform tagmentation and library preparation

    • Sequence to appropriate depth (minimum 5 million reads per sample)

  • Signal deconvolution: Apply a Gaussian Mixture Model to the distribution of fragment size averages using an Expectation Maximization algorithm to separate signals from different antibodies .

  • Data analysis: After deconvolution, analyze the separated profiles using standard peak-calling algorithms and comparative genomics approaches .

  • Validation: Confirm accuracy by comparing to single-antibody CUT&Tag profiles and validating with orthogonal methods like ChIP-seq.

This approach is particularly valuable for maximizing information from limited biological samples, providing comprehensive views of both active (acetylated) and repressive chromatin states from the same cells .

How do I interpret changes in Acetyl-HIST1H2AG (K5) levels in response to cellular stimuli?

Interpreting changes in Acetyl-HIST1H2AG (K5) levels requires careful consideration of both biological context and technical factors:

  • Establishing baselines: Determine normal acetylation levels across various cell types under standard conditions before assessing responses to stimuli. Histone acetylation levels can vary significantly between cell types and developmental stages.

  • Time-course considerations: Histone acetylation changes can be rapid and transient. Design time-course experiments to capture both immediate and sustained changes, typically ranging from 30 minutes to 24 hours post-stimulus.

  • Stimulus-specific patterns: Different stimuli produce distinct patterns of histone acetylation. For example:

    • Histone deacetylase inhibitors like Trichostatin A induce global hyperacetylation across multiple sites

    • Growth factors may produce more targeted acetylation at specific gene loci

    • Stress signals can lead to complex patterns of both increased and decreased acetylation

  • Integrated analysis: Correlate acetylation changes with:

    • Transcriptional changes (RNA-seq)

    • Other histone modifications (additional ChIP or CUT&Tag assays)

    • Transcription factor binding (ChIP-seq for relevant factors)

    • Chromatin accessibility (ATAC-seq)

  • Pathway analysis: Identify affected gene networks and signaling pathways by aggregating genes showing significant acetylation changes.

  • Functional validation: Confirm the causal relationship between acetylation changes and observed phenotypes through genetic manipulation or pharmacological intervention.

  • Quantification approaches: Normalize acetylation signals to total histone levels or to housekeeping genes when assessing global changes. For genomic approaches, use appropriate normalization methods such as spike-in controls.

What are the common technical artifacts when working with Acetyl-HIST1H2AG (K5) antibody, and how can they be mitigated?

Several common technical artifacts can influence experiments using Acetyl-HIST1H2AG (K5) antibody:

  • Antibody cross-reactivity:

    • Issue: Antibodies may cross-react with similar acetylated lysine residues on other histones

    • Mitigation: Validate specificity using peptide arrays or competitive binding assays

    • Control: Include specificity tests with related acetylated peptides

  • Batch-to-batch variability:

    • Issue: Different antibody lots may show varying levels of specificity and sensitivity

    • Mitigation: Validate each new lot against previous ones using consistent positive controls

    • Control: Maintain reference samples for inter-batch calibration

  • Epitope masking:

    • Issue: Neighboring modifications or protein interactions may block antibody access

    • Mitigation: Use native ChIP approaches or optimize fixation conditions

    • Control: Perform parallel experiments with antibodies recognizing adjacent modifications

  • Sample preparation artifacts:

    • Issue: Acetyl groups can be lost during sample processing due to endogenous histone deacetylase activity

    • Mitigation: Include histone deacetylase inhibitors (e.g., sodium butyrate, TSA) in all buffers

    • Control: Process samples rapidly and maintain cold temperatures

  • Quantification bias:

    • Issue: Signal intensity may not linearly correlate with acetylation levels

    • Mitigation: Establish standard curves with known quantities of acetylated histones

    • Control: Include calibration samples in each experiment

  • Background in imaging experiments:

    • Issue: Non-specific nuclear staining may obscure specific signals

    • Mitigation: Include blocking peptides and optimize antibody concentration

    • Control: Include appropriate negative controls (e.g., secondary antibody only)

  • ChIP efficiency variations:

    • Issue: Efficiency may vary between genomic regions due to chromatin context

    • Mitigation: Use spike-in controls and normalize appropriately

    • Control: Include internal control regions with stable acetylation levels

How can I integrate Acetyl-HIST1H2AG (K5) data with other epigenetic marks to understand chromatin state dynamics?

Integrating Acetyl-HIST1H2AG (K5) data with other epigenetic marks provides a comprehensive view of chromatin state dynamics:

  • Multi-omics experimental design:

    • Sequential ChIP: Perform sequential immunoprecipitation with Acetyl-HIST1H2AG (K5) followed by antibodies against other modifications to identify co-occurrence

    • Parallel ChIP profiles: Generate parallel ChIP profiles for multiple modifications from the same cellular population

    • CUT&Tag2for1 approach: Utilize the CUT&Tag2for1 method for simultaneous profiling of active and repressive marks from the same cells

    • Single-cell approaches: Apply single-cell versions of these techniques to capture cellular heterogeneity

  • Computational integration frameworks:

    • Correlation analysis: Calculate correlation coefficients between genome-wide distributions of different marks

    • Hidden Markov Models: Apply ChromHMM or similar algorithms to segment the genome into distinct chromatin states based on combinatorial patterns

    • Dimensionality reduction: Use techniques like UMAP to visualize relationships between different marks across genomic regions or cellular populations

    • Trajectory analysis: Construct pseudotime trajectories to track chromatin state changes during biological processes

  • Functional annotation:

    • Gene Ontology enrichment: Identify biological processes associated with regions showing specific combinations of marks

    • Motif analysis: Discover transcription factor binding motifs enriched in regions with particular chromatin signatures

    • Enhancer identification: Correlate acetylation patterns with predicted enhancer elements using databases like ENCODE or Roadmap Epigenomics

  • Visualization strategies:

    • Genome browsers: Create custom tracks displaying multiple marks aligned to genomic coordinates

    • Heatmaps: Generate heatmaps centered on features of interest (e.g., transcription start sites) showing patterns of different modifications

    • Metaplots: Create aggregate plots showing average signals across defined genomic features

  • Integration with transcriptional data:

    • Expression correlation: Correlate acetylation patterns with gene expression levels from RNA-seq

    • Transcription factor binding: Integrate with ChIP-seq data for transcription factors

    • Chromatin accessibility: Correlate with ATAC-seq or DNase-seq data

This integrated approach allows researchers to move beyond individual modifications to understand the complex language of combinatorial histone marks that define functional chromatin states.

What are effective troubleshooting strategies when Acetyl-HIST1H2AG (K5) antibody produces unexpected results?

When encountering unexpected results with Acetyl-HIST1H2AG (K5) antibody, implement the following structured troubleshooting approach:

  • No signal or weak signal in Western blotting:

    • Verify antibody activity: Test with positive control samples treated with histone deacetylase inhibitors like Trichostatin A (500 ng/ml for 4 hours)

    • Optimize protein extraction: Ensure your extraction method preserves histone modifications by including deacetylase inhibitors

    • Check protein transfer: Confirm transfer efficiency with reversible staining of membranes

    • Increase antibody concentration: Titrate antibody from 1:1000 to 1:5000 to determine optimal concentration

    • Extend exposure time: Use longer exposure times for chemiluminescent detection

  • High background in immunostaining:

    • Optimize blocking: Increase blocking time or try alternative blocking reagents (BSA vs. non-fat milk)

    • Reduce antibody concentration: Perform serial dilutions to identify minimal effective concentration

    • Increase washing steps: Extend wash duration and add additional wash steps

    • Use antigen retrieval controls: Optimize antigen retrieval methods (heat-mediated antigen retrieval with Tris/EDTA buffer pH 9.0)

  • Poor enrichment in ChIP experiments:

    • Check chromatin quality: Ensure proper fragmentation (200-500 bp) and appropriate crosslinking

    • Increase antibody amount: Test higher amounts of antibody per ChIP reaction

    • Extend incubation time: Allow longer antibody-chromatin binding time (overnight at 4°C)

    • Verify primers: Test primers on input DNA and design primers to known acetylated regions

    • Reduce stringency: Modify wash buffers to reduce stringency if specific signal is too low

  • Inconsistent results between experiments:

    • Standardize cell culture conditions: Control cell density, passage number, and serum lots

    • Use internal controls: Include consistent positive and negative controls in each experiment

    • Maintain consistent timing: Standardize sample collection and processing times

    • Document lot numbers: Track antibody lot numbers and prepare larger working aliquots

  • Unexpected band pattern in Western blots:

    • Verify sample integrity: Check for protein degradation with total H2A antibody

    • Test specificity: Perform peptide competition assays to confirm specificity of all observed bands

    • Cross-reactivity assessment: Compare band patterns with those seen using other H2A modification antibodies

  • Discrepancies between techniques:

    • Consider context-dependent accessibility: Epitope accessibility may differ between techniques

    • Verify fixation effects: Compare results between fixed and native samples where possible

    • Check for mask effects: Multiple modifications may occur at adjacent sites, affecting antibody binding

How can I design experiments to study the dynamics of Acetyl-HIST1H2AG (K5) during cell differentiation or disease progression?

Designing experiments to study Acetyl-HIST1H2AG (K5) dynamics during biological processes requires careful planning:

  • Time-course experimental design:

    • Sampling strategy: Collect samples at critical transition points (determined by pilot studies)

    • Temporal resolution: Increase sampling frequency during periods of rapid change

    • Synchronized populations: Use cell synchronization methods for cell cycle studies

    • Single-cell approaches: Implement single-cell techniques to capture population heterogeneity

  • Cell differentiation studies:

    • Model selection: Choose appropriate differentiation models (e.g., embryonic stem cells to specific lineages)

    • Staging markers: Include established markers of differentiation stages

    • Induction methods: Use standardized induction protocols with defined factors

    • Heterogeneity analysis: Apply single-cell approaches to capture asynchronous differentiation

  • Disease progression models:

    • Patient cohort design: Stratify samples by disease stage, treatment response, and clinical parameters

    • Matched controls: Include matched healthy tissues or cells for comparison

    • Longitudinal sampling: Collect samples at multiple timepoints when possible

    • Disease-relevant cell types: Focus on cell types most relevant to pathology

  • Integrated methodological approaches:

    • Multi-omics profiling: Combine Acetyl-HIST1H2AG (K5) profiling with:

      • RNA-seq for transcriptional changes

      • Other histone modifications (H3K27me3, H3K4me3)

      • Chromatin accessibility assays (ATAC-seq)

      • DNA methylation analysis

    • Sequential sampling: Apply different methods to aliquots from the same sample

    • CUT&Tag2for1: Utilize this method for simultaneous profiling of active and repressive marks

  • Data analysis frameworks:

    • Trajectory analysis: Apply pseudotime or actual time-series analysis

    • Differential regions identification: Identify genomic regions with significant changes in acetylation

    • Co-regulation networks: Construct networks of co-regulated genes based on acetylation patterns

    • Integration with public databases: Compare with disease-relevant datasets from resources like TCGA

  • Validation strategies:

    • Orthogonal techniques: Validate key findings using alternative methods

    • Perturbation experiments: Manipulate acetylation through HDAC inhibitors or genetic approaches

    • Functional assays: Connect acetylation changes to functional outcomes

    • In vivo models: Extend findings to appropriate animal models when relevant

What are the latest advances in using Acetyl-HIST1H2AG (K5) antibodies for single-cell epigenomic analysis?

Recent advances have significantly expanded the capabilities of Acetyl-HIST1H2AG (K5) antibodies in single-cell epigenomic analysis:

  • Technical innovations in single-cell chromatin profiling:

    • CUT&Tag for single cells: Adaptations of CUT&Tag protocols allow for profiling histone modifications in single cells with improved sensitivity

    • CUT&Tag2for1 approach: This method enables simultaneous profiling of active (e.g., Acetyl-H2A-K5) and repressive marks in single cells through computational signal deconvolution based on fragment size differences

    • Microfluidic platforms: Integration with microfluidic devices allows for higher throughput and reduced reagent consumption

    • Single-cell combinatorial indexing: Barcoding strategies enable profiling thousands of single cells in a single experiment

  • Computational advancements:

    • Deconvolution algorithms: Sophisticated algorithms separate signals from different modifications based on fragment characteristics

    • Dimensionality reduction: Methods like UMAP effectively visualize relationships between cells based on epigenomic profiles

    • Integration frameworks: Tools for integrating single-cell epigenomic data with transcriptomic or proteomic data from the same cells

    • Trajectory inference: Algorithms reconstructing developmental trajectories from snapshot epigenomic data

  • Biological applications:

    • Cellular heterogeneity: Identification of epigenetically distinct cell subpopulations within seemingly homogeneous tissues

    • Regulatory network inference: Construction of gene regulatory networks based on correlations between acetylation patterns and gene expression

    • Rare cell identification: Detection of rare cell types based on their unique epigenomic signatures

    • Response to perturbation: Characterization of heterogeneous responses to drugs or genetic manipulations

  • Current limitations and solutions:

    • Low coverage per cell: Addressed through computational imputation and feature aggregation

    • Antibody specificity concerns: Mitigated with rigorous validation and spike-in controls

    • Batch effects: Controlled through experimental design and computational correction

    • Data integration challenges: Solved with advanced multi-modal data integration methods

  • Future directions:

    • Spatial epigenomics: Combining single-cell acetylation profiling with spatial information

    • Multi-modification profiling: Simultaneous measurement of multiple histone modifications in single cells

    • Live-cell dynamics: Development of sensors for tracking acetylation changes in living cells

    • Clinical applications: Implementation in diagnostic workflows for heterogeneous diseases

These advances are rapidly transforming our understanding of epigenetic heterogeneity and its role in development, disease, and cellular response to environmental factors.

How does the information obtained from Acetyl-HIST1H2AG (K5) antibodies compare with other histone acetylation markers?

Understanding the distinct properties of Acetyl-HIST1H2AG (K5) relative to other histone acetylation markers is crucial for experimental design and interpretation:

Histone Acetylation MarkPrimary AssociationGenomic LocalizationStability/DynamicsCommon Research Applications
H2A-K5ac (HIST1H2AG)Gene activation, DNA damage responsePromoters, enhancersModerate turnoverTranscriptional regulation, DNA repair studies
H3K9acActive transcriptionTSS, promotersRapid turnoverGene expression studies, enhancer mapping
H3K27acActive enhancersEnhancers, super-enhancersDynamicEnhancer activity mapping, cell-type-specific regulation
H4K5acTranscription initiationPromotersDynamicTranscription initiation studies
H4K16acChromatin decompactionBroad domainsStableChromatin structure analysis

Acetyl-HIST1H2AG (K5) provides distinct information compared to other acetylation marks:

  • Variant-specific regulation: Unlike more general H3 or H4 acetylation marks, Acetyl-HIST1H2AG (K5) can reveal regulation specific to this H2A variant, which may have unique functions in certain cellular contexts .

  • Damage response correlation: H2A.X, which shares the K5 acetylation site, has established roles in DNA damage response when phosphorylated at S139. The acetylation at K5 may provide complementary information about chromatin states during repair processes .

  • Cross-talk with other modifications: Acetylation at H2A-K5 can influence or be influenced by nearby modifications, creating complex regulatory patterns distinct from those seen with other acetylation marks.

  • Temporal dynamics: Studies comparing various acetylation marks have shown differing temporal dynamics in response to stimuli, with H2A-K5ac showing unique patterns in certain cell types and conditions.

  • Technical considerations: Each acetylation mark presents distinct challenges for detection, with some being more sensitive to fixation conditions or antibody accessibility issues than others.

Understanding these distinctions allows researchers to select the most appropriate markers for specific research questions and to interpret results in the context of the broader epigenetic landscape.

What emerging technologies are improving the application of Acetyl-HIST1H2AG (K5) antibodies in epigenetic research?

Several emerging technologies are enhancing the utility of Acetyl-HIST1H2AG (K5) antibodies in epigenetic research:

  • Advanced ChIP methodologies:

    • CUT&Tag and CUT&RUN: These techniques offer improved signal-to-noise ratio and require fewer cells than traditional ChIP, making them valuable for precious samples

    • CUT&Tag2for1: This approach enables simultaneous profiling of acetylation marks and other chromatin features through computational deconvolution, maximizing information from limited samples

    • Micro-ChIP: Miniaturized ChIP protocols allow for profiling from extremely small cell numbers (<1000 cells)

    • ChIPmentation: Integration of tagmentation into ChIP workflows streamlines library preparation

  • Single-cell technologies:

    • Single-cell CUT&Tag: Adaptations for single-cell analysis reveal cell-to-cell variability in acetylation patterns

    • Single-cell multi-omics: Methods for parallel profiling of histone acetylation and transcriptomes or other epigenetic features in the same cells

    • Microfluidic platforms: Devices enabling high-throughput single-cell epigenomic profiling with reduced reagent consumption

  • Imaging innovations:

    • Super-resolution microscopy: Techniques like STORM and PALM allow visualization of acetylation marks at unprecedented resolution

    • Live-cell acetylation sensors: Genetically encoded sensors for tracking acetylation dynamics in living cells

    • Multiplexed imaging: Methods for simultaneously visualizing multiple histone modifications in the same sample

  • Computational advances:

    • Deep learning approaches: Neural networks for pattern recognition in acetylation data across the genome

    • Integration frameworks: Tools for integrating acetylation data with other omics datasets

    • Signal deconvolution algorithms: Methods for separating signals from different modifications in multiplexed assays

  • Synthetic biology approaches:

    • CRISPR-based epigenome editing: Targeted manipulation of acetylation at specific genomic loci

    • Engineered reader domains: Modified proteins that recognize specific acetylation patterns with enhanced specificity

    • Orthogonal acetylation systems: Introduction of non-native acetylation machinery for controlled studies

  • Clinical applications:

    • Liquid biopsy adaptations: Methods for detecting histone acetylation patterns in circulating nucleosomes

    • Diagnostic implementations: Development of acetylation-based biomarkers for disease diagnosis and prognosis

    • Therapeutic monitoring: Use of acetylation profiles to track responses to epigenetic therapies

These technological advances are rapidly expanding the scope and impact of research using Acetyl-HIST1H2AG (K5) antibodies, enabling more comprehensive understanding of epigenetic regulation in health and disease.

What are the critical quality control steps for maintaining reproducibility when working with Acetyl-HIST1H2AG (K5) antibody?

Implementing rigorous quality control measures ensures reliable and reproducible results when working with Acetyl-HIST1H2AG (K5) antibody:

  • Antibody validation:

    • Lot testing: Validate each new antibody lot against previous lots using consistent positive controls

    • Specificity confirmation: Regularly perform peptide array or competition assays to confirm specificity

    • Cross-reactivity assessment: Test against related acetylated histones to ensure specificity

    • Publication of validation data: Document and share validation results to support data reproducibility

  • Sample preparation standardization:

    • Consistent fixation: Standardize fixation conditions for all experiments (time, temperature, reagent concentrations)

    • Preservation of modifications: Include deacetylase inhibitors in all buffers to prevent loss of acetylation

    • Chromatin preparation: Establish consistent protocols for chromatin isolation and fragmentation

    • Sample storage: Implement standardized storage conditions to maintain modification integrity

  • Experimental controls:

    • Positive controls: Include samples treated with histone deacetylase inhibitors like Trichostatin A

    • Negative controls: Include appropriate negative controls (IgG, untreated samples)

    • Internal standards: Use spike-in controls for quantitative experiments

    • Reference genes/regions: Monitor consistently acetylated regions as internal controls

  • Technical replication:

    • Multiple biological replicates: Analyze at least three independent biological samples

    • Technical duplicates: Perform technical duplicates for critical experiments

    • Independent validation: Confirm key findings using orthogonal methods

    • Blinding: Implement blinded analysis for subjective assessments

  • Data analysis standardization:

    • Consistent normalization: Apply uniform normalization methods across experiments

    • Batch correction: Account for batch effects in data analysis

    • Statistical thresholds: Establish and maintain consistent statistical criteria

    • Analysis pipelines: Document and version-control computational workflows

  • Documentation and reporting:

    • Detailed methods: Record all experimental parameters, including reagent sources and lot numbers

    • Protocol sharing: Make detailed protocols available to the scientific community

    • Raw data availability: Deposit raw data in appropriate repositories

    • Metadata reporting: Include all relevant metadata according to community standards

  • Cross-laboratory validation:

    • Ring trials: Participate in multi-laboratory validation studies when possible

    • Independent replication: Collaborate with independent labs for critical findings

    • Standard reference materials: Use community standard samples when available

    • Protocol harmonization: Align protocols with community standards

Adhering to these quality control measures significantly enhances the reproducibility and reliability of research using Acetyl-HIST1H2AG (K5) antibody, contributing to more robust and translatable findings in epigenetic research.

How should researchers integrate Acetyl-HIST1H2AG (K5) antibody data into broader epigenetic regulatory networks?

Integrating Acetyl-HIST1H2AG (K5) data into comprehensive epigenetic regulatory networks requires systematic approaches spanning experimental design through computational analysis:

  • Experimental design considerations:

    • Coordinate experimental timing: Collect samples for different assays at matched timepoints

    • Paired analyses: When possible, perform multiple assays on aliquots from the same biological samples

    • Multi-omics strategies: Plan for integrated analyses across epigenetic, transcriptomic, and proteomic levels

    • CUT&Tag2for1 implementation: Consider using this approach to simultaneously profile active and repressive marks from the same cells

  • Data collection across modalities:

    • Histone modifications: Profile complementary modifications (H3K27me3, H3K4me3, H3K27ac)

    • DNA methylation: Perform whole-genome bisulfite sequencing or reduced representation approaches

    • Chromatin accessibility: Include ATAC-seq or DNase-seq data

    • Transcription factor binding: ChIP-seq for relevant transcription factors

    • RNA expression: RNA-seq for steady-state expression and nascent RNA methods for transcription rates

  • Computational integration frameworks:

    • Correlation analyses: Compute correlation matrices across different data types

    • Network construction: Build regulatory networks incorporating different epigenetic layers

    • Module identification: Identify co-regulated modules across data types

    • Causal modeling: Apply causal inference methods to establish regulatory relationships

    • Visualization approaches: Develop multi-modal visualization strategies for integrated data

  • Integration with public resources:

    • Reference epigenomes: Compare with reference datasets from ENCODE or Roadmap Epigenomics

    • Disease databases: Integrate with disease-relevant datasets from sources like TCGA

    • Gene regulatory databases: Incorporate knowledge from established regulatory databases

    • Evolutionary conservation: Consider conservation patterns across species

  • Functional validation strategies:

    • Perturbation experiments: Manipulate acetylation at key loci using CRISPR-based epigenome editing

    • Pathway modulation: Alter relevant signaling pathways and monitor effects on acetylation networks

    • Mutational analyses: Examine effects of genetic variants on acetylation patterns

    • Phenotypic correlations: Connect network patterns to cellular or organismal phenotypes

  • Translational considerations:

    • Biomarker development: Identify acetylation signatures with diagnostic or prognostic value

    • Therapeutic targeting: Pinpoint network nodes amenable to therapeutic intervention

    • Patient stratification: Use integrated networks for patient classification

    • Drug response prediction: Develop predictive models for response to epigenetic therapies

By systematically integrating Acetyl-HIST1H2AG (K5) data with other epigenetic and transcriptional information, researchers can construct comprehensive regulatory networks that provide deeper insights into biological processes and disease mechanisms than any single data type alone.

What are the most valuable resources for researchers working with Acetyl-HIST1H2AG (K5) antibody?

Researchers working with Acetyl-HIST1H2AG (K5) antibody can benefit from numerous resources spanning reagents, protocols, databases, and analytical tools:

  • Antibody resources:

    • Antibody validation databases: Resources like Antibodypedia and the Antibody Registry provide validation data and user reviews

    • Alternative antibody sources: Compare antibodies from different vendors with detailed validation data

    • Recombinant antibody options: Consider recombinant monoclonal antibodies for enhanced reproducibility

    • Custom validation services: Services for independent antibody validation

  • Protocol repositories:

    • Protocol Exchange and protocols.io: Platforms with detailed, version-controlled experimental protocols

    • CUT&Tag protocols: Resources for implementing CUT&Tag with acetylation antibodies

    • Single-cell adaptation guides: Protocols for adapting antibody-based methods to single-cell applications

    • Troubleshooting guides: Community resources addressing common technical challenges

  • Reference datasets:

    • ENCODE and Roadmap Epigenomics: Comprehensive reference epigenomes across cell types and tissues

    • Gene Expression Omnibus (GEO): Repository containing relevant ChIP-seq and related datasets

    • 4D Nucleome: Resources on 3D chromatin organization and its relationship to histone modifications

    • Cancer epigenome atlases: Collections of cancer-specific epigenomic data including histone acetylation

  • Computational tools:

    • Peak calling software: Tools optimized for histone modification data (MACS2, SICER)

    • Integration frameworks: Software for multi-omics data integration

    • Visualization platforms: Genome browsers and specialized visualization tools

    • Analysis pipelines: Standardized workflows for epigenomic data processing

  • Cell and tissue resources:

    • Reference cell lines: Well-characterized cell lines with established acetylation profiles

    • Epigenetically relevant disease models: Cell and animal models with epigenetic dysregulation

    • Organoid systems: Advanced 3D culture systems for more physiologically relevant studies

    • Primary cell biobanks: Sources of primary cells with diverse genetic backgrounds

  • Training and networking:

    • Specialized workshops: Training in epigenomic techniques and data analysis

    • Research consortia: Collaborative networks focused on epigenomic research

    • Online communities: Forums for troubleshooting and method discussion

    • Specialized conferences: Meetings focused on histone modifications and chromatin biology

  • Emerging technology access:

    • Technology development grants: Funding opportunities for implementing cutting-edge methods

    • Core facilities: Specialized facilities offering advanced epigenomic profiling services

    • Commercial services: Companies providing specialized epigenomic profiling services

    • Collaborative networks: Partnerships for accessing emerging technologies

These resources collectively support researchers in effectively utilizing Acetyl-HIST1H2AG (K5) antibodies across diverse research applications, from basic mechanistic studies to translational research.

What are the future directions and unanswered questions in research involving Acetyl-HIST1H2AG (K5)?

The study of Acetyl-HIST1H2AG (K5) continues to evolve, with several promising future directions and critical unanswered questions:

  • Mechanistic understanding:

    • Writers and erasers: Which specific histone acetyltransferases and deacetylases regulate H2A-K5 acetylation in different contexts?

    • Readers: What proteins specifically recognize and bind to Acetyl-HIST1H2AG (K5) to mediate downstream effects?

    • Crosstalk: How does H2A-K5 acetylation interact with other histone modifications, particularly on the same nucleosome?

    • Variant-specific functions: How does acetylation at K5 differ functionally between canonical H2A and variant forms like H2A.X?

  • Biological roles:

    • Cell type-specific functions: Does Acetyl-HIST1H2AG (K5) play distinct roles in different cell types and developmental stages?

    • DNA damage response: What is the relationship between H2A-K5 acetylation and the DNA damage response, particularly given H2A.X's known role in this process?

    • Transcriptional regulation: How does this modification specifically contribute to gene activation or repression in different genomic contexts?

    • Nuclear architecture: Does H2A-K5 acetylation influence higher-order chromatin structure and nuclear organization?

  • Disease relevance:

    • Cancer biomarkers: Can patterns of H2A-K5 acetylation serve as diagnostic or prognostic biomarkers in specific cancer types?

    • Neurodegenerative diseases: Is there altered H2A-K5 acetylation in conditions like Alzheimer's and Parkinson's diseases?

    • Inflammatory disorders: How does this modification change during inflammatory responses and in chronic inflammatory conditions?

    • Therapeutic targeting: Can modulation of H2A-K5 acetylation be exploited for therapeutic interventions?

  • Technological advances:

    • Live-cell dynamics: Development of tools to monitor H2A-K5 acetylation in living cells in real-time

    • Single-molecule approaches: Methods to study this modification at the single-nucleosome level

    • Spatial organization: Technologies to map the 3D distribution of Acetyl-HIST1H2AG (K5) in the nucleus

    • Targeted modulation: Precise tools to manipulate this modification at specific genomic loci

  • Evolutionary perspectives:

    • Conservation patterns: How conserved is the regulation and function of H2A-K5 acetylation across species?

    • Evolutionary adaptations: Does this modification play roles in species-specific adaptations?

    • Comparative epigenomics: How do patterns of H2A-K5 acetylation compare across evolutionary diverse organisms?

    • Ancient origins: When did regulatory mechanisms for this specific modification emerge during evolution?

  • Translational potential:

    • Clinical applications: Can Acetyl-HIST1H2AG (K5) patterns serve as biomarkers for disease diagnosis or treatment response?

    • Drug development: Is this modification a viable target for epigenetic therapies?

    • Personalized medicine: Can individual variations in H2A-K5 acetylation patterns inform personalized treatment strategies?

    • Environmental influences: How do environmental factors influence this modification, and what are the implications for human health?

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.