Acetyl-HIST1H3A (K115) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

Target:

  • Protein: Histone H3.1 (encoded by HIST1H3A and related paralogs)

  • Modification: Lysine 115 acetylation (K115ac)

  • Synonyms: HIST1H3A/B/C/D/E/F/G/H/I/J, H3FA, H3FL, H3FC

Host Species: Rabbit (polyclonal)

Immunogen:

  • Peptide sequence surrounding acetyl-K115 (e.g., residues 111–122)

  • KLH-conjugated synthetic acetylated peptide (exact sequence proprietary)

Species Reactivity:

  • Primary: Human (Homo sapiens)

  • Secondary: Rat (Rattus norvegicus)

Key Validation Data:

  • Western Blot: Strong signal in sodium butyrate-treated cells (e.g., HEK-293, A549) .

  • ICC: Nuclear localization confirmed in HeLa cells .

Chromatin and Transcriptional Regulation

  • Reduces DNA-histone interactions at the nucleosome dyad, facilitating chromatin remodeling .

  • Mutations (e.g., K115Q) impair transcriptional silencing at telomeres and ribosomal DNA in yeast .

DNA Repair Mechanisms

  • K115 acetylation sensitizes cells to DNA-damaging agents (e.g., Zeocin, hydroxyurea) .

  • K115A and K122A mutants exhibit defective double-strand break repair .

Epigenetic Crosstalk

  • Associated with active transcription and chromatin accessibility .

  • Modulates interactions between histone H3/H4 tetramers and DNA .

Research Limitations and Considerations

  • Specificity: Cross-reactivity with non-acetylated H3 not fully ruled out in some studies .

  • Species Restrictions: Limited reactivity beyond human and rat .

  • Functional Assays: Most data derive from in vitro or yeast models; human cell studies remain sparse .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery information.
Synonyms
Histone H3.1 (Histone H3/a) (Histone H3/b) (Histone H3/c) (Histone H3/d) (Histone H3/f) (Histone H3/h) (Histone H3/i) (Histone H3/j) (Histone H3/k) (Histone H3/l), HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, H3FA, H3FL, H3FC, H3FB, H3FD, H3FI, H3FH, H3FK, H3FF, H3FJ
Target Names
Uniprot No.

Target Background

Function
Histone H3A is a core component of the nucleosome. Nucleosomes wrap and compact DNA into chromatin, restricting DNA accessibility to cellular machinery that require DNA as a template. As such, histones play a critical role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex system of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer may occur through E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course may be useful for determining whether the tumors are heterochronous. PMID: 29482987
  3. JMJD5, a Jumonji C (JmjC) domain-containing protein, is a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that cause a DNA damage response. PMID: 28982940
  4. Data suggests that the Ki-67 antigen proliferative index has significant limitations and that phosphohistone H3 (PHH3) is an alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This series of 47 diffuse midline gliomas revealed that histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Data show that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments showed that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl) where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. For the first time, we describe the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high grade gliomas (pedHGG) including GBM and DIPG harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not rare. PMID: 28547652
  14. Data show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest whereas the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that binding of helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. On the other hand, histone 3 mutations do not seem to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis and repression of APAK. PMID: 25961932
  27. Taken together, the authors verified that histone H3 is a real substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) serves a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Data indicate that the lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays is regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Acetyl-HIST1H3A (K115) antibody and what is its significance in epigenetic research?

Acetyl-HIST1H3A (K115) antibody is a polyclonal antibody that specifically recognizes histone H3.1 acetylated at lysine 115. Histone H3.1 is a core component of nucleosomes that wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries requiring DNA as a template. The antibody plays a crucial role in epigenetic research by enabling the detection and quantification of this specific post-translational modification.

Histone acetylation represents a key regulatory mechanism in the "histone code" that influences gene expression patterns. Specifically, acetylation of lysine residues neutralizes the positive charge of histones, weakening their interaction with negatively charged DNA and potentially creating a more open chromatin structure accessible to transcription factors . The K115 acetylation site is of particular interest as it may play specific roles in chromatin remodeling distinct from the better-characterized histone tail modifications.

What are the primary experimental applications for Acetyl-HIST1H3A (K115) antibody?

The Acetyl-HIST1H3A (K115) antibody can be utilized in multiple experimental techniques:

ApplicationRecommended DilutionPurpose
ELISANot specifiedQuantitative detection of acetylated histone H3.1
Western Blot (WB)1:100-1:1000Protein analysis of acetylated histone H3.1
Immunocytochemistry (ICC)1:1-1:100Cellular localization of acetylated histone H3.1
Immunohistochemistry (IHC)1:50-1:500Tissue localization of acetylated histone H3.1
Immunofluorescence (IF)1:30-1:200Visualization of acetylated histone H3.1

These applications allow researchers to investigate the presence, abundance, and localization of K115-acetylated histone H3.1 in various experimental systems . The optimal dilution may vary depending on the specific experimental conditions and sample types being examined.

How does the Acetyl-HIST1H3A (K115) antibody compare with antibodies targeting other histone H3 acetylation sites?

The Acetyl-HIST1H3A (K115) antibody specifically recognizes acetylation at lysine 115, which distinguishes it from other histone H3 acetylation-specific antibodies that target different lysine residues such as K9, K14, K18, K23, and K27 . While most commonly studied histone acetylation sites are located in the N-terminal tail that protrudes from the nucleosome core, K115 is positioned within the globular domain of histone H3.1, potentially making it functionally distinct.

This specificity is critical for research applications focused on understanding the unique biological roles of K115 acetylation. Unlike pan-acetyl antibodies that recognize multiple acetylated lysines on histone H3 , the K115-specific antibody allows for precise detection of this particular modification without cross-reactivity to other acetylation sites. When designing experiments to study specific histone modifications, researchers should consider whether they need information about a specific acetylation site (requiring site-specific antibodies like Acetyl-HIST1H3A (K115)) or general acetylation status (where pan-acetyl antibodies may be more appropriate).

What are the optimal sample preparation protocols for detecting acetylated HIST1H3A (K115) in different experimental systems?

For effective detection of acetylated HIST1H3A (K115), sample preparation protocols must preserve the acetylation status while ensuring accessibility of the epitope. For different applications:

For Western Blot analysis:

  • Include histone deacetylase inhibitors (e.g., sodium butyrate, trichostatin A) in lysis buffers to prevent deacetylation during extraction

  • Use acid extraction methods (e.g., 0.2N HCl) for efficient histone isolation

  • Add protease inhibitors to prevent degradation

  • Avoid excessive heat during sample processing which may alter epitope structure

For immunohistochemistry and immunocytochemistry:

  • Use appropriate fixation methods (typically 4% paraformaldehyde)

  • Perform heat-mediated antigen retrieval in EDTA buffer (pH 8.0) to expose the epitope

  • Block with 10% goat serum to reduce non-specific binding

  • Incubate with the antibody at optimal dilution (1:50 for IHC) overnight at 4°C

For chromatin immunoprecipitation (ChIP):

  • Use fresh samples when possible

  • Cross-link proteins to DNA using formaldehyde

  • Optimize sonication conditions to achieve appropriate chromatin fragment sizes

  • Include appropriate controls (input, IgG, positive control)

These protocols may require optimization depending on the specific research question, cell type, and experimental conditions.

What are the critical factors affecting antibody specificity and how can cross-reactivity with other histone modifications be minimized?

Several factors can impact the specificity of Acetyl-HIST1H3A (K115) antibody:

  • Epitope recognition: The antibody was raised against synthetic peptides derived from the region around K115 of human histone H3.1 . Variations in the amino acid sequence surrounding K115 across species may affect antibody binding.

  • Antibody production method: Polyclonal antibodies like Acetyl-HIST1H3A (K115) antibody contain a mixture of immunoglobulins that recognize different epitopes of the antigen, potentially increasing the risk of cross-reactivity compared to monoclonal antibodies .

  • Blocking efficiency: Insufficient blocking can lead to non-specific binding.

To minimize cross-reactivity:

  • Validation experiments: Perform peptide competition assays using acetylated and non-acetylated peptides to confirm specificity

  • Appropriate controls: Include samples known to be negative for K115 acetylation

  • Pre-absorption: Consider pre-absorbing the antibody with related histone peptides lacking the K115 acetylation

  • Optimized blocking: Use appropriate blocking reagents (typically 5-10% serum matched to the secondary antibody species)

  • Stringent washing: Include additional washing steps with higher salt concentrations or detergents

Validation through multiple techniques (e.g., mass spectrometry) can provide additional confirmation of antibody specificity.

What storage and handling practices maximize antibody performance and shelf life?

To maintain optimal performance of Acetyl-HIST1H3A (K115) antibody:

  • Storage temperature: Store at -20°C or -80°C for long-term preservation. For frequent use over short periods, storage at 4°C is acceptable .

  • Avoid freeze-thaw cycles: Repeated freezing and thawing can damage antibody structure and reduce binding efficiency. Aliquot the antibody upon first thawing to minimize freeze-thaw cycles .

  • Buffer composition: The antibody is typically supplied in a buffer containing preservatives (0.03% Proclin 300) and stabilizers (50% Glycerol, 0.01M PBS, pH 7.4) . Maintain these conditions when diluting.

  • Working dilutions: Prepare working dilutions immediately before use rather than storing diluted antibody for extended periods.

  • Contamination prevention: Use sterile techniques when handling the antibody to prevent microbial contamination.

  • Temperature during experiments: Keep the antibody on ice during experimental procedures but avoid freezing.

  • Light exposure: Minimize exposure to light, particularly for fluorophore-conjugated secondary antibodies.

Following these practices will help maintain antibody specificity and sensitivity throughout the shelf life, typically 12-24 months from the date of receipt under optimal storage conditions.

How can Acetyl-HIST1H3A (K115) antibody be effectively employed in ChIP-seq experiments to map genome-wide distribution of this modification?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using Acetyl-HIST1H3A (K115) antibody requires careful optimization:

  • Antibody quality control: Before ChIP-seq, validate the antibody's specificity and efficiency through Western blot and ChIP-qPCR at known target regions.

  • Fixation optimization: Typically, 1% formaldehyde for 10 minutes at room temperature is used, but this may require adjustment depending on cell type.

  • Chromatin fragmentation: Optimize sonication conditions to generate DNA fragments of 200-500 bp for highest resolution mapping.

  • Immunoprecipitation conditions:

    • Use 2-5 μg of antibody per ChIP reaction

    • Include appropriate controls (input, IgG control, positive control antibody)

    • Optimize incubation time and temperature (typically overnight at 4°C)

  • Sequencing considerations:

    • Generate paired-end reads for improved mapping accuracy

    • Aim for minimum 20 million uniquely mapped reads per sample

    • Include biological replicates (minimum of 2-3)

  • Bioinformatic analysis pipeline:

    • Use specialized peak calling algorithms appropriate for histone modifications (e.g., MACS2 with broad peak settings)

    • Perform differential binding analysis between experimental conditions

    • Integrate with gene expression data to correlate modification with transcriptional outcomes

  • Validation strategies:

    • Confirm selected peaks with ChIP-qPCR

    • Validate functional significance through targeted experiments (e.g., CRISPR-mediated modification of acetylation sites)

This comprehensive approach enables genome-wide profiling of K115 acetylation patterns and their correlation with transcriptional regulation and chromatin states.

What experimental approaches can distinguish between the biological roles of K115 acetylation and other histone H3 modifications in regulating gene expression?

Distinguishing the specific biological roles of K115 acetylation from other histone modifications requires multi-faceted experimental approaches:

  • Site-specific mutagenesis:

    • Generate K115R (cannot be acetylated) or K115Q (acetylation mimetic) mutations in histone H3.1

    • Express these mutants in cells with depleted endogenous H3.1

    • Compare phenotypic and transcriptional effects to wild-type and other acetylation site mutants

  • CRISPR-based epigenome editing:

    • Use dCas9 fused to histone acetyltransferases (HATs) or deacetylases (HDACs) for site-specific manipulation

    • Target specific genomic loci to assess the direct impact of K115 acetylation changes on gene expression

    • Compare with similar manipulations of other acetylation sites

  • Combinatorial ChIP (sequential ChIP or Re-ChIP):

    • Perform sequential immunoprecipitation with Acetyl-HIST1H3A (K115) antibody followed by antibodies against other histone modifications

    • Identify genomic regions with co-occurrence or mutual exclusivity of modifications

  • Temporal dynamics analysis:

    • Analyze the temporal sequence of histone modifications during cellular processes

    • Determine whether K115 acetylation precedes or follows other modifications

    • Use synchronized cell systems or inducible gene expression models

  • Proteomic approaches:

    • Identify proteins that specifically recognize K115 acetylation using acetylated peptide pull-downs

    • Compare with proteins binding to other acetylated lysine residues

    • Perform mass spectrometry to identify modification patterns that co-occur with K115 acetylation

  • Functional genomics screen:

    • Conduct CRISPR screens targeting writers, readers, and erasers of histone modifications

    • Compare effects on K115 acetylation versus other histone marks

    • Identify enzymes specifically regulating K115 acetylation

These approaches collectively provide a comprehensive understanding of the unique functional significance of K115 acetylation in comparison to other histone modifications.

How can researchers optimize multiplexed detection of Acetyl-HIST1H3A (K115) alongside other histone modifications?

Multiplexed detection of histone modifications presents technical challenges but offers valuable insights into their interrelationships. Strategies for optimizing multiplexed detection include:

  • Multi-color immunofluorescence microscopy:

    • Select primary antibodies from different host species (e.g., rabbit anti-Acetyl-HIST1H3A (K115) with mouse anti-H3K27me3)

    • Use species-specific secondary antibodies with non-overlapping fluorophores

    • Implement appropriate controls to ensure no cross-reactivity between antibodies

    • Apply spectral unmixing algorithms if fluorophore emission spectra overlap

  • Sequential immunoblotting:

    • Strip and reprobe membranes sequentially with different antibodies

    • Include complete stripping controls to ensure removal of previous antibodies

    • Arrange the sequence from lowest to highest abundance modifications

    • Consider using different detection methods (chemiluminescence, fluorescence) for quantitative multiplexing

  • Flow cytometry-based approaches:

    • Use fluorophore-conjugated antibodies with distinct excitation/emission spectra

    • Include single-stain controls for compensation calculations

    • Apply hierarchical gating strategies to analyze co-occurrence patterns

  • Mass spectrometry-based approaches:

    • Develop targeted methods to quantify multiple histone modifications simultaneously

    • Implement stable isotope labeling to compare modification levels across conditions

    • Use fragmentation methods that preserve modification information

  • Multiplex ChIP-seq methods:

    • Apply ChIP-seq protocols with antibody barcoding (e.g., ChIP-STARR-seq)

    • Implement sequential ChIP for co-occurrence analysis

    • Use computational methods to integrate single-modification ChIP-seq datasets

  • Single-cell epigenomic approaches:

    • Adapt CUT&Tag or CUT&RUN protocols for single-cell analysis

    • Integrate with single-cell transcriptomics for correlation with gene expression

    • Apply dimensionality reduction and clustering to identify cell populations with distinct modification patterns

Successful multiplexing requires extensive validation of antibody specificity, optimization of staining protocols, and sophisticated data analysis approaches to interpret the complex relationships between different histone modifications.

What are the most common technical challenges when using Acetyl-HIST1H3A (K115) antibody and how can they be addressed?

Researchers may encounter several technical challenges when working with Acetyl-HIST1H3A (K115) antibody:

  • Low signal intensity:

    • Increase antibody concentration within recommended ranges

    • Optimize antigen retrieval methods (for IHC/ICC)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Use signal amplification systems (e.g., biotin-streptavidin)

    • Ensure acetylation status is preserved during sample preparation by including HDAC inhibitors

  • High background signal:

    • Increase blocking time or concentration (use 5-10% goat serum)

    • Add additional washing steps with higher stringency

    • Reduce primary and secondary antibody concentrations

    • Pre-absorb antibody with non-specific proteins

    • Use more specific detection systems

  • Inconsistent results between experiments:

    • Standardize sample preparation protocols

    • Use positive and negative controls in each experiment

    • Implement quantitative analysis methods

    • Consider batch effects in experimental design

    • Use the same lot of antibody when possible

  • Cross-reactivity with other acetylation sites:

    • Validate specificity using peptide competition assays

    • Include appropriate controls (e.g., samples with K115R mutation)

    • Perform parallel experiments with antibodies against other acetylation sites

    • Confirm critical findings with orthogonal techniques (e.g., mass spectrometry)

  • Poor reproducibility in ChIP experiments:

    • Optimize chromatin fragmentation

    • Increase antibody amount or affinity purify the antibody

    • Include spike-in controls for normalization

    • Standardize bioinformatic analysis pipelines

Addressing these challenges requires systematic optimization and careful experimental design with appropriate controls.

How can researchers validate the specificity of their acetylation signal detected using this antibody?

Validation of antibody specificity is critical for confidence in experimental results. Recommended approaches include:

  • Peptide competition assays:

    • Pre-incubate the antibody with excess acetylated peptide (specific to K115)

    • Pre-incubate with unmodified peptide as control

    • Compare signal reduction between conditions

    • Signal should be eliminated with acetylated peptide but not with unmodified peptide

  • Genetic validation:

    • Use cells expressing K115R mutant histone H3.1 (cannot be acetylated)

    • Signal should be absent or significantly reduced in these cells

    • Compare with K115Q (acetylation mimetic) mutants as positive controls

  • Enzyme treatment controls:

    • Treat samples with histone deacetylases (HDACs) to remove acetylation

    • Compare signal with untreated samples

    • Signal should decrease in HDAC-treated samples

  • Mass spectrometry validation:

    • Perform parallel MS analysis to confirm K115 acetylation status

    • Quantify relative abundance of the modification

    • Correlate MS data with antibody-based detection results

  • Multiple antibody validation:

    • Compare results using alternative antibodies against the same modification

    • Consistent results across different antibodies increase confidence

  • Induction experiments:

    • Treat cells with HDAC inhibitors to increase acetylation levels

    • Compare signal before and after treatment

    • Signal should increase with HDAC inhibitor treatment

  • Reproducibility across techniques:

    • Confirm findings using multiple detection methods (WB, IHC, ChIP)

    • Consistent results across techniques support specificity

What experimental controls are essential when using Acetyl-HIST1H3A (K115) antibody for quantitative analysis of histone acetylation levels?

Robust quantitative analysis of K115 acetylation requires comprehensive controls:

  • Positive controls:

    • Samples with known high levels of K115 acetylation (e.g., cells treated with HDAC inhibitors)

    • Recombinant acetylated histones or synthetic acetylated peptides

    • Previously validated positive samples

  • Negative controls:

    • Samples lacking the target (e.g., K115R mutant-expressing cells)

    • Samples from specific tissues/cell types known to lack the modification

    • Secondary antibody-only controls to assess non-specific binding

  • Normalization controls:

    • Total histone H3 levels (using pan-H3 antibodies)

    • Housekeeping proteins for loading control in Western blots

    • Spike-in standards for ChIP experiments (e.g., Drosophila chromatin)

  • Antibody controls:

    • IgG control matching the host species of the primary antibody

    • Isotype controls to assess non-specific binding

    • Antibody titration series to establish linear detection range

  • Technical controls:

    • Standard curves using defined quantities of acetylated peptides

    • Replicate samples to assess technical variability

    • Dilution series to confirm proportional signal relationships

  • Treatment controls:

    • HDAC inhibitor treatment (positive control)

    • HAT inhibitor treatment (negative control)

    • Time course experiments to establish modification dynamics

  • Validation standards:

    • Include established laboratories' validated positive samples when possible

    • Participate in antibody validation initiatives or ring trials

Incorporation of these controls, along with appropriate statistical analysis, enables reliable quantification of K115 acetylation levels across experimental conditions.

How might single-cell epigenomic techniques be adapted to study HIST1H3A K115 acetylation patterns across heterogeneous cell populations?

Adapting single-cell technologies to study K115 acetylation offers exciting opportunities to uncover cell-type-specific regulation:

  • Single-cell CUT&Tag/CUT&RUN approaches:

    • Modify protein A-Tn5 fusion protocols to use Acetyl-HIST1H3A (K115) antibody

    • Implement microfluidic platforms for cell isolation and processing

    • Incorporate cell barcoding strategies for multiplexed analysis

    • Optimize tagmentation conditions for histone modifications

  • scChIC-seq (single-cell Chromatin Immunocleavage sequencing):

    • Adapt for K115 acetylation using specific antibodies

    • Implement droplet-based or plate-based workflows

    • Integrate with transcriptomic readouts for multi-omic analysis

  • Mass cytometry (CyTOF) adaptation:

    • Develop metal-conjugated Acetyl-HIST1H3A (K115) antibodies

    • Establish multiparameter panels including other histone modifications

    • Apply dimensionality reduction and clustering algorithms to identify cell populations with distinct modification patterns

  • Single-cell combinatorial indexing:

    • Apply combinatorial indexing methods for high-throughput single-cell analysis

    • Optimize fixation and permeabilization to preserve K115 acetylation

    • Integrate with RNA readouts for correlation with gene expression

  • Computational challenges and solutions:

    • Develop specialized computational pipelines for sparse data analysis

    • Implement imputation methods appropriate for epigenomic data

    • Apply trajectory inference to model dynamics of K115 acetylation

    • Integrate with other single-cell data types through multi-modal analysis frameworks

  • Validation strategies:

    • Confirm cell type-specific patterns using sorted cell populations

    • Validate key findings with orthogonal methods like imaging

    • Integrate with spatial information using techniques like spatial transcriptomics

These approaches enable characterization of K115 acetylation heterogeneity at unprecedented resolution, potentially revealing cell state-specific roles in diverse biological processes.

What technological advances in antibody development might enhance the specificity and sensitivity of detecting acetylated HIST1H3A (K115)?

Several emerging technologies hold promise for improving antibody performance:

  • Recombinant antibody engineering:

    • Development of recombinant monoclonal antibodies with defined sequences

    • Affinity maturation through directed evolution

    • Humanization of antibodies for reduced background in human samples

    • Creation of smaller antibody formats (e.g., single-chain variable fragments) for improved tissue penetration

  • Synthetic antibody alternatives:

    • Aptamer development specific to acetylated K115

    • Engineered binding proteins based on alternative scaffolds

    • Nanobodies (single-domain antibodies) with enhanced specificity

    • Peptide-based affinity reagents designed through computational approaches

  • Modification-specific enhancement strategies:

    • Dual-recognition antibodies requiring binding to both the histone backbone and the acetyl modification

    • Proximity-based detection systems that amplify signal only when specific epitope configurations are recognized

    • Conformation-sensitive antibodies that detect structural changes induced by acetylation

  • Production improvements:

    • Cell-free expression systems for antibody production

    • Glycoengineering to optimize antibody properties

    • Advanced purification methods to isolate only the highest-affinity antibody populations

  • Validation technologies:

    • High-throughput epitope mapping

    • Structural analysis of antibody-antigen complexes

    • Comprehensive cross-reactivity profiling against related modifications

    • Standardized validation pipelines across laboratories

  • Detection enhancements:

    • Signal amplification technologies (e.g., tyramide signal amplification)

    • Photoswitchable antibodies for super-resolution imaging

    • Multiplexed detection through DNA-barcoded antibodies

These technological advances could significantly improve the reliability and utility of Acetyl-HIST1H3A (K115) antibodies for research applications, potentially enabling detection of modifications present at lower abundance or in challenging sample types.

How can multi-omics approaches integrate Acetyl-HIST1H3A (K115) data with other epigenetic and transcriptomic datasets to elucidate functional relationships?

Integrative multi-omics approaches provide comprehensive insights into the functional significance of K115 acetylation:

  • Sequential multi-omics from the same samples:

    • Perform ChIP-seq for K115 acetylation followed by RNA-seq

    • Integrate with other histone modification ChIP-seq datasets

    • Add DNA methylation profiling (WGBS or RRBS)

    • Include chromatin accessibility data (ATAC-seq or DNase-seq)

  • Advanced computational integration frameworks:

    • Apply machine learning models to predict functional relationships

    • Implement network analysis to identify regulatory modules

    • Use Bayesian approaches to infer causal relationships

    • Develop multidimensional visualization tools for complex data exploration

  • Single-cell multi-omics adaptations:

    • Implement scM&T-seq (simultaneous transcriptome and methylome)

    • Develop methods to combine CUT&Tag with scRNA-seq

    • Apply trajectory inference to model temporal relationships between modifications and expression

  • Spatial multi-omics:

    • Integrate spatial transcriptomics with imaging-based histone modification detection

    • Develop multiplexed in situ hybridization and immunofluorescence protocols

    • Apply computational methods to align data from different modalities in spatial contexts

  • Functional validation strategies:

    • Targeted epigenome editing to manipulate K115 acetylation at specific loci

    • Perturbation experiments targeting writers/erasers/readers of K115 acetylation

    • Time-resolved studies to establish causality between epigenetic changes and transcriptional outcomes

  • Data integration challenges and solutions:

    • Standardize data preprocessing across modalities

    • Implement batch correction methods for multi-omic datasets

    • Develop specialized statistical approaches for integrated hypothesis testing

    • Establish data sharing standards and repositories for multi-omic datasets

By implementing these integrative approaches, researchers can comprehensively map the functional relationships between K115 acetylation and other molecular features, potentially revealing novel regulatory mechanisms and therapeutic targets.

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.