Acetyl-HIST1H3A (K37) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery details.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
Histone H3 is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, thus limiting DNA accessibility to cellular machinery that utilizes DNA as a template. Histones, therefore, play a critical role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is modulated through a complex array of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies suggest that epigenetic regulation in cancer may be influenced by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course could be helpful in determining whether the tumors are heterochronous. PMID: 29482987
  3. Research indicates that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that induce a DNA damage response. PMID: 28982940
  4. Data suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) may serve as 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 research indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, histone H3-K27M mutation was found to be 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. Experiments revealed 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. This research describes, for the first time, 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 uncommon. PMID: 28547652
  14. Research indicates 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. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that binding of the 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 the 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 the 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. Research indicates that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) plays 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 lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Research indicates that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the 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

How does Acetyl-HIST1H3A (K37) differ from other histone acetylation sites?

Acetyl-HIST1H3A (K37) differs from other histone acetylation sites such as K14 and K23 in several important ways. While all histone acetylations generally promote a more open chromatin structure, each site has unique biological functions and interacting partners. Acetylation at K37 has distinct recognition patterns by reader proteins and potentially different roles in transcriptional regulation compared to well-studied sites like K14 . The K37 site is located in a different region of the histone H3 protein compared to K14 (which is found in the N-terminal tail) and K23, potentially affecting different aspects of nucleosome structure and function. Understanding these differences is critical for accurate interpretation of experimental results when comparing multiple histone modifications. Unlike K14 acetylation, which has been extensively characterized in various tissues and cell types, K37 acetylation may have more specialized functions that are still being elucidated in current research.

What are the recommended applications for Acetyl-HIST1H3A (K37) antibodies in epigenetic research?

Acetyl-HIST1H3A (K37) antibodies are versatile tools suitable for multiple experimental applications in epigenetic research. Based on the available data, these antibodies have been validated for Western blotting (WB), immunofluorescence (IF), chromatin immunoprecipitation (ChIP), and enzyme-linked immunosorbent assay (ELISA) . For optimal results in Western blotting, a dilution range of 1:100-1:1000 is recommended, while immunofluorescence typically requires a more concentrated dilution of 1:1-1:10 . In ChIP experiments, these antibodies can effectively precipitate acetylated histones to identify genomic regions associated with this specific modification. Additionally, they can be employed in immunohistochemistry (IHC) to visualize the distribution of this histone mark in tissue sections, offering insights into tissue-specific epigenetic patterns. Each application requires specific optimization for buffer conditions, incubation times, and antibody concentrations to achieve optimal signal-to-noise ratios.

What is the optimal sample preparation protocol for detecting Acetyl-HIST1H3A (K37) in different experimental contexts?

The optimal sample preparation protocol for detecting Acetyl-HIST1H3A (K37) varies depending on the experimental application and sample type. For Western blotting and ELISA, acid extraction is the preferred method for isolating histones from cells or tissues. This typically involves cell lysis followed by extraction with dilute acid (typically 0.2N HCl) to selectively solubilize histones while leaving most other cellular proteins behind . For immunofluorescence and immunohistochemistry applications, formaldehyde fixation (typically 4% paraformaldehyde) followed by permeabilization is recommended to preserve nuclear structure while allowing antibody access .

For ChIP experiments, crosslinking with formaldehyde (typically 1% for 10 minutes at room temperature) followed by sonication to fragment chromatin into 200-500bp pieces yields optimal results. When working with tissue samples for immunohistochemistry, heat-mediated antigen retrieval in EDTA buffer (pH 8.0) has proven effective for exposing the K37 epitope, as demonstrated with related histone acetylation antibodies . Regardless of the application, inclusion of histone deacetylase inhibitors (such as sodium butyrate or trichostatin A) in all buffers is crucial to prevent artificial loss of acetylation during sample processing.

How should researchers design proper controls for experiments using Acetyl-HIST1H3A (K37) antibodies?

Proper experimental controls are essential when working with Acetyl-HIST1H3A (K37) antibodies to ensure reliable and interpretable results. The following control strategy is recommended:

  • Positive Controls: Include samples known to express high levels of the acetylation mark, such as cells treated with histone deacetylase inhibitors like trichostatin A or sodium butyrate. These treatments increase global histone acetylation levels and serve as excellent positive controls .

  • Negative Controls: Utilize one or more of the following:

    • Samples treated with histone acetyltransferase inhibitors

    • Immunoprecipitation with isotype-matched IgG (for ChIP experiments)

    • Peptide competition assays to demonstrate antibody specificity

    • Samples where the target epitope has been enzymatically removed

  • Normalization Controls: For quantitative applications, always measure total Histone H3 levels in parallel to normalize for variations in histone content between samples .

  • Cross-reactivity Controls: Test the antibody against recombinant histones with different acetylation patterns to confirm specificity for the K37 site versus other acetylation sites.

  • Technical Controls: Include no-primary-antibody controls in immunostaining experiments and loading controls in Western blots to account for technical variations.

This comprehensive control strategy helps distinguish true biological signals from artifacts and ensures that any observed changes in Acetyl-HIST1H3A (K37) levels reflect genuine biological phenomena rather than technical variations.

What are the critical parameters for optimizing Western blot detection of Acetyl-HIST1H3A (K37)?

Optimizing Western blot detection of Acetyl-HIST1H3A (K37) requires careful attention to several critical parameters:

  • Sample Preparation: Utilize acid extraction methods (0.2N HCl) to isolate histones, and include histone deacetylase inhibitors (e.g., 5-10mM sodium butyrate) in all buffers to prevent loss of acetylation during processing .

  • Gel Selection: Use 12-15% SDS-PAGE gels to achieve optimal separation of histones, which have low molecular weights (approximately 15-17 kDa for histone H3) .

  • Transfer Conditions: Employ shorter transfer times (50-90 minutes) at lower currents (150 mA) to prevent small proteins like histones from transferring through the membrane . Nitrocellulose membranes with 0.22μm pore size are recommended for better retention of small proteins.

  • Blocking Conditions: Block with 5% non-fat milk in TBS for 1.5 hours at room temperature to minimize background while preserving epitope accessibility .

  • Antibody Dilution: Start with a 1:500 dilution of primary antibody in blocking buffer and incubate overnight at 4°C. This concentration can be adjusted based on signal strength in preliminary experiments .

  • Detection System: Use enhanced chemiluminescence (ECL) systems for sensitive detection. For Acetyl-HIST1H3A (K37), the expected band should appear at approximately 15-17 kDa .

  • Loading Controls: Include a total histone H3 antibody blot as a loading control to normalize acetylation levels across samples.

By carefully optimizing these parameters, researchers can achieve consistent and specific detection of Acetyl-HIST1H3A (K37) in Western blot experiments.

What is the recommended protocol for ChIP experiments targeting Acetyl-HIST1H3A (K37)?

The recommended protocol for ChIP experiments targeting Acetyl-HIST1H3A (K37) involves several critical steps to ensure high specificity and sensitivity:

  • Crosslinking: Fix cells with 1% formaldehyde for 10 minutes at room temperature to preserve protein-DNA interactions. Quench with 125mM glycine for 5 minutes.

  • Cell Lysis and Chromatin Isolation: Lyse cells in appropriate buffers containing protease inhibitors and histone deacetylase inhibitors (5-10mM sodium butyrate) to preserve acetylation marks .

  • Chromatin Shearing: Sonicate chromatin to generate fragments of 200-500bp. Optimize sonication conditions for each cell type to avoid over or under-shearing.

  • Immunoprecipitation:

    • Pre-clear chromatin with protein A/G beads

    • Incubate 2-5μg of Acetyl-HIST1H3A (K37) antibody with chromatin overnight at 4°C

    • Add protein A/G beads and incubate for 2-4 hours

    • Include a parallel IP with non-specific IgG as a negative control

    • Include an input sample (5-10% of starting chromatin) as a reference

  • Washing and Elution: Perform stringent washes to remove non-specific binding. Elute chromatin-antibody complexes with elution buffer containing SDS.

  • Reverse Crosslinking and DNA Purification: Reverse crosslinks at 65°C overnight, treat with proteinase K, and purify DNA using column-based methods.

  • Quantification: Analyze enrichment by qPCR, focusing on regions of interest and including control regions known to lack the modification. Alternatively, the samples can be processed for ChIP-seq analysis for genome-wide profiling.

For accurate results, it's essential to normalize ChIP data to input controls and to include positive control regions where acetylation is expected and negative control regions where it should be absent.

How can researchers accurately quantify changes in Acetyl-HIST1H3A (K37) levels across experimental conditions?

Accurate quantification of changes in Acetyl-HIST1H3A (K37) levels requires a multi-faceted approach that accounts for technical variability and biological context:

  • Western Blot Quantification:

    • Always normalize acetylation signals to total histone H3 levels

    • Use digital image analysis software to measure band intensities

    • Ensure images are captured in the linear range of detection

    • Include a standard curve of recombinant acetylated histones when possible

    • Present data as the ratio of acetylated H3K37 to total H3

  • ELISA-Based Quantification:

    • Utilize standard curves with recombinant acetylated histones

    • Perform technical triplicates for each sample

    • Calculate acetylation levels as absolute values or as a percentage of total H3

    • A sandwich ELISA approach with a capture antibody for total H3 and detection antibody for acetyl-K37 provides the most reliable results

  • ChIP-qPCR Quantification:

    • Express enrichment as percent of input or as enrichment over IgG control

    • Normalize to a housekeeping gene region with stable acetylation

    • For comparative analysis across conditions, use the ΔΔCt method

    • Include multiple genomic regions as internal controls

  • Mass Spectrometry Approaches:

    • For absolute quantification, use isotope-labeled synthetic peptides as internal standards

    • Calculate the stoichiometry of acetylation at K37 relative to unmodified peptides

    • Integrate peaks corresponding to the acetylated and unmodified peptides

This multi-method approach provides robust quantification of Acetyl-HIST1H3A (K37) levels, enabling confident interpretation of biological changes across experimental conditions.

What are common challenges in Acetyl-HIST1H3A (K37) antibody experiments and how can they be overcome?

Researchers working with Acetyl-HIST1H3A (K37) antibodies frequently encounter several challenges that can be addressed with specific troubleshooting strategies:

  • Cross-reactivity with Other Acetylation Sites:

    • Challenge: Antibodies may recognize acetylation marks at similar sites on H3

    • Solution: Validate specificity using peptide competition assays with acetylated peptides covering different sites

    • Solution: Use recombinant histones with defined acetylation patterns as controls

  • Weak or Variable Signal Intensity:

    • Challenge: Inconsistent detection of the acetylation mark

    • Solution: Include histone deacetylase inhibitors in all buffers (5-10mM sodium butyrate)

    • Solution: Optimize fixation conditions (for IF/IHC) to preserve epitope accessibility

    • Solution: Test different antibody concentrations and incubation times

  • Background in Western Blots and Immunostaining:

    • Challenge: High non-specific binding obscuring specific signals

    • Solution: Increase washing steps and stringency

    • Solution: Use higher dilutions of primary antibody (e.g., 1:1000 instead of 1:100)

    • Solution: Pre-absorb antibodies with acetylated peptides from other sites

  • Variability in ChIP Experiments:

    • Challenge: Inconsistent enrichment patterns

    • Solution: Standardize chromatin preparation methods, particularly sonication conditions

    • Solution: Optimize antibody-to-chromatin ratios

    • Solution: Include spike-in controls to normalize for technical variations

  • Limited Sample Material:

    • Challenge: Insufficient material from rare cell populations

    • Solution: Adapt protocols for low-input ChIP or use carrier proteins

    • Solution: Consider more sensitive detection methods like ELISA instead of Western blot

By implementing these troubleshooting strategies, researchers can overcome common challenges and generate reliable data using Acetyl-HIST1H3A (K37) antibodies across various experimental applications.

How should researchers interpret contradictory results between different detection methods for Acetyl-HIST1H3A (K37)?

When faced with contradictory results between different detection methods for Acetyl-HIST1H3A (K37), researchers should conduct a systematic analysis to resolve these discrepancies:

  • Methodological Considerations:

    • Different methods have varying sensitivities and dynamic ranges

    • Western blots provide semi-quantitative measure of bulk acetylation

    • ChIP measures genomic localization rather than absolute levels

    • ELISA offers quantitative measurement but lacks spatial information

    • Immunofluorescence provides spatial information but is less quantitative

  • Reconciliation Strategy:

    • Create a comparison table of results from each method

    • Identify patterns in the contradictions (e.g., do discrepancies occur only in certain cell types?)

    • Assess whether discrepancies reflect technical limitations or biological complexity

    • Consider whether different methods are measuring different populations of the modification

  • Validation Approaches:

    • Use orthogonal techniques such as mass spectrometry to provide unbiased verification

    • Test multiple antibodies targeting the same modification

    • Employ genetic approaches (e.g., histone mutants) to validate antibody specificity

    • Use cells with known modulation of the mark (e.g., HAT/HDAC inhibition) as controls

  • Biological Context:

    • Consider whether contradictions reflect biologically relevant dynamics

    • Acetylation marks may differ between nuclear compartments or chromatin states

    • Temporal dynamics of acetylation may explain differences between methods

    • Cell-cycle dependent changes may affect results from asynchronous populations

The table below summarizes how to interpret and reconcile contradictory results:

Detection MethodStrengthLimitationReconciliation Approach
Western BlotBulk quantificationLacks spatial informationCompare to total H3 levels; validate with multiple antibodies
ChIPGenomic localizationLimited to accessible regionsCorrelate with gene expression changes; validate with sequential ChIP
ELISAQuantitativeLacks spatial informationUse as complement to other methods for precise quantification
ImmunofluorescenceSpatial distributionLess quantitativeCorrelate patterns with other nuclear markers; quantify with digital image analysis
Mass SpectrometryDirect peptide identificationRequires specialized equipmentUse to validate antibody-based methods

By systematically analyzing contradictions between methods, researchers can develop a more complete understanding of Acetyl-HIST1H3A (K37) dynamics and function.

How can Acetyl-HIST1H3A (K37) antibodies be used in genome-wide studies of chromatin regulation?

Acetyl-HIST1H3A (K37) antibodies can be leveraged in several sophisticated approaches for genome-wide chromatin regulation studies:

  • ChIP-Sequencing (ChIP-seq):

    • Use Acetyl-HIST1H3A (K37) antibodies for immunoprecipitation followed by next-generation sequencing

    • This approach maps genome-wide distribution of K37 acetylation

    • Integrate with transcriptome data to correlate acetylation with gene expression

    • Compare with other histone marks to identify co-occurring modifications

  • CUT&RUN and CUT&Tag:

    • These newer techniques offer higher signal-to-noise ratios than traditional ChIP

    • They require fewer cells and less antibody

    • For Acetyl-HIST1H3A (K37), they can provide higher resolution mapping of acetylation sites

    • Particularly useful for rare cell populations or limited clinical samples

  • Sequential ChIP (Re-ChIP):

    • Perform successive immunoprecipitations with Acetyl-HIST1H3A (K37) antibody and antibodies to other modifications

    • This identifies genomic regions with co-occurrence of multiple modifications

    • Helps establish the histone code combinations associated with specific regulatory states

  • Chromosome Conformation Capture with ChIP (HiChIP):

    • Combines ChIP with Hi-C to identify long-range chromatin interactions associated with K37 acetylation

    • Links this specific acetylation to three-dimensional genome organization

    • Reveals how K37 acetylation may influence enhancer-promoter interactions

  • Single-Cell Approaches:

    • Adapt ChIP protocols for single-cell analysis (scChIP-seq)

    • Examine cell-to-cell variability in K37 acetylation patterns

    • Correlate with single-cell transcriptomics to link epigenetic heterogeneity to gene expression variability

These genome-wide approaches provide comprehensive insights into the regulatory functions of Acetyl-HIST1H3A (K37) across the entire genome, facilitating discoveries about its role in transcriptional regulation, chromatin organization, and cellular identity.

What is known about the role of Acetyl-HIST1H3A (K37) in disease states, and how can researchers investigate this connection?

While specific information about Acetyl-HIST1H3A (K37) in disease states is limited in the provided search results, researchers can draw on methodologies used for studying related histone modifications to investigate potential connections:

  • Cancer Research Applications:

    • Examine K37 acetylation patterns in cancer versus normal tissues using immunohistochemistry

    • The techniques validated for other histone acetylation sites like K14 in various cancer tissues (glioma, ovarian, bladder, and lung cancer) can be adapted for K37 studies

    • Compare acetylation levels between different cancer subtypes to identify potential biomarkers

    • Correlate K37 acetylation with patient outcomes and treatment responses

  • Neurodegenerative Disorders:

    • Investigate K37 acetylation changes in brain tissues from neurodegenerative disease models

    • Techniques validated in rat and mouse brain tissues for K14 acetylation can be adapted for K37

    • Examine how disease-associated mutations in histone acetyltransferases or deacetylases affect K37 acetylation

    • Study the impact of environmental factors on K37 acetylation in neural cells

  • Investigation Methodologies:

    • Clinical Correlation Studies: Analyze K37 acetylation in patient samples and correlate with clinical parameters

    • Functional Studies: Manipulate enzymes responsible for K37 acetylation/deacetylation and observe phenotypic changes

    • Drug Screening: Test compounds that modulate K37 acetylation for therapeutic potential

    • Animal Models: Generate transgenic models with mutations affecting K37 acetylation

  • Technological Approaches:

    • Use ELISA-based screening to quantify K37 acetylation levels across large patient cohorts

    • Apply mass spectrometry to identify disease-specific changes in K37 acetylation stoichiometry

    • Implement ChIP-seq to map genome-wide changes in K37 acetylation distribution in disease states

By applying these methodologies, researchers can elucidate the specific roles of Acetyl-HIST1H3A (K37) in various disease processes and potentially identify new therapeutic targets or diagnostic markers.

How does Acetyl-HIST1H3A (K37) interact with other histone modifications in the context of the histone code?

Understanding how Acetyl-HIST1H3A (K37) interacts with other histone modifications requires investigation of the complex interplay within the histone code framework:

  • Co-occurrence Patterns:

    • K37 acetylation may co-occur with other activating marks like H3K4 methylation or H3K9 acetylation

    • It may be mutually exclusive with repressive marks like H3K27 methylation

    • ChIP-seq data can be analyzed for genome-wide correlation patterns between K37ac and other modifications

    • Sequential ChIP (Re-ChIP) experiments can directly detect co-occurrence on the same nucleosomes

  • Enzyme Regulation:

    • Histone acetyltransferases (HATs) responsible for K37 acetylation may preferentially target nucleosomes with specific pre-existing modifications

    • Conversely, K37 acetylation may create binding sites for reader proteins that recruit other modifying enzymes

    • Understanding these enzymatic cascades helps decipher the sequential establishment of modification patterns

  • Functional Consequences:

    • Combined modifications may have synergistic or antagonistic effects on chromatin structure

    • The presence of K37 acetylation may influence the binding of chromatin-remodeling complexes

    • Integration of K37 acetylation with other modifications may determine transcriptional outcomes

    • This modification may play a role in nucleosome stability and positioning

  • Research Approaches:

    • Mass Spectrometry: Identify combinations of modifications that exist on the same histone molecule

    • Protein Interaction Studies: Determine how K37 acetylation affects binding of reader proteins

    • Structural Biology: Investigate how K37 acetylation alters nucleosome structure

    • Synthetic Biology: Create designer nucleosomes with defined modification patterns to test functional hypotheses

Understanding these interactions will provide insights into how K37 acetylation contributes to the complex language of the histone code and its role in chromatin-based processes like transcription, replication, and DNA repair.

What emerging technologies are advancing the study of Acetyl-HIST1H3A (K37) and similar histone modifications?

Several cutting-edge technologies are transforming how researchers study Acetyl-HIST1H3A (K37) and similar histone modifications:

  • Advanced Microscopy Techniques:

    • Super-resolution microscopy allows visualization of individual nucleosomes

    • Live-cell imaging with modification-specific antibodies enables temporal tracking of acetylation dynamics

    • Correlative light and electron microscopy (CLEM) links acetylation patterns to ultrastructural features of chromatin

  • New Chromatin Profiling Methods:

    • CUT&Tag and CUT&RUN provide higher resolution mapping with less background than traditional ChIP

    • Cleavage Under Targets and Tagmentation (CUT&Tag) offers particular advantages for histone modifications due to its high sensitivity and low background

    • These methods require fewer cells, enabling studies of rare cell populations or clinical samples

  • Single-Cell Technologies:

    • Single-cell ChIP-seq and CUT&Tag reveal cell-to-cell variability in histone modification patterns

    • Integration with single-cell transcriptomics and proteomics provides multi-omic views of epigenetic heterogeneity

    • Microfluidic platforms enable high-throughput single-cell epigenomic profiling

  • Synthetic Biology Approaches:

    • Designer nucleosomes with site-specific acetylation using unnatural amino acid incorporation

    • Optogenetic control of histone acetyltransferases allows temporal manipulation of acetylation

    • CRISPR-based epigenome editing enables site-specific modification of acetylation patterns

  • Computational and AI Methods:

    • Machine learning algorithms predict functional impacts of histone modification patterns

    • Network analysis tools identify regulatory relationships between different modifications

    • Advanced bioinformatic pipelines integrate multi-omic data to provide comprehensive views of chromatin regulation

  • Proteomics Innovations:

    • Top-down proteomics preserves information about co-occurring modifications on the same histone molecule

    • Targeted mass spectrometry approaches quantify specific modifications with high sensitivity

    • Crosslinking mass spectrometry identifies proteins that interact with acetylated histones

These technological advances are enabling unprecedented insights into the functions and dynamics of Acetyl-HIST1H3A (K37) and its role in the complex regulatory networks of chromatin.

What quality control measures should be implemented when working with Acetyl-HIST1H3A (K37) antibodies?

Implementing rigorous quality control measures is essential when working with Acetyl-HIST1H3A (K37) antibodies to ensure reliable and reproducible results:

  • Antibody Validation Tests:

    • Peptide Competition Assays: Pre-incubate antibody with acetylated and non-acetylated peptides to confirm specificity

    • Western Blot on Histone Extracts: Verify single band at expected molecular weight (15-17 kDa)

    • Dot Blot Analysis: Test reactivity against a panel of modified histone peptides

    • Testing in HAT/HDAC Inhibitor-Treated Cells: Confirm expected changes in signal intensity

  • Lot-to-Lot Consistency:

    • Test each new antibody lot against a standard sample

    • Maintain reference samples for comparison

    • Document lot numbers and validation results for reproducibility

    • Consider creating a standardized validation protocol specific to your laboratory

  • Storage and Handling:

    • Follow manufacturer recommendations for storage (typically -20°C or -80°C)

    • Avoid repeated freeze-thaw cycles by preparing small aliquots

    • Monitor antibody performance over time to detect potential degradation

    • Document storage conditions and antibody age for each experiment

  • Experimental Controls:

    • Include positive controls (HAT inhibitor-treated samples) in each experiment

    • Run negative controls (HDAC inhibitor-treated samples) in parallel

    • Use recombinant histones with defined modification status when available

    • Include isotype controls for immunoprecipitation experiments

  • Documentation and Reporting:

    • Maintain detailed records of antibody source, catalog number, and lot

    • Document all validation experiments performed

    • Report antibody details in publications according to antibody reporting guidelines

    • Consider sharing validation data in repositories like Antibodypedia

Implementing these quality control measures will enhance the reliability of results and facilitate troubleshooting when unexpected results occur.

How can researchers optimize cost and efficiency when incorporating Acetyl-HIST1H3A (K37) antibody-based assays into their research programs?

Optimizing cost and efficiency for Acetyl-HIST1H3A (K37) antibody-based assays requires strategic planning and resource management:

  • Antibody Usage Optimization:

    • Titrate antibodies to determine minimum effective concentration for each application

    • For Western blots, dilutions of 1:100-1:1000 may be suitable, while IF typically requires 1:1-1:10

    • Recover and reuse antibodies for ChIP by adding sodium azide (0.02%) after use

    • Consider antibody-based multiplexing to detect multiple modifications simultaneously

  • Sample Processing Efficiency:

    • Batch process samples to minimize reagent waste and improve consistency

    • Optimize protocols for microvolume applications to reduce antibody consumption

    • Implement automated liquid handling where available for high-throughput applications

    • Scale down protocols without compromising data quality

  • Alternative Approaches:

    • Use ELISA assays for quantitative screening before proceeding to more resource-intensive techniques

    • Consider CUT&Tag as a more efficient alternative to traditional ChIP for genomic profiling

    • Implement multiplexed approaches to measure multiple modifications in single experiments

    • Develop targeted approaches focused on regions of interest rather than genome-wide profiling

  • Resource Sharing:

    • Establish core facilities for specialized techniques like ChIP-seq

    • Create antibody validation consortia across research groups

    • Share positive control samples and standardized protocols

    • Implement electronic lab notebooks to document optimal conditions

  • Cost-Benefit Analysis for Different Applications:

    ApplicationRelative CostSample RequirementTime InvestmentWhen to Use
    Western BlotMediumMedium1-2 daysInitial validation, bulk quantification
    ELISALow-MediumLow3-4 hoursHigh-throughput screening, quantification
    ChIP-qPCRMediumHigh2-3 daysGene-specific studies
    ChIP-seqHighHigh5-7 daysGenome-wide profiling
    CUT&TagMedium-HighLow2-3 daysGenome-wide with limited samples
    IF/IHCMediumLow1-2 daysSpatial distribution studies

By implementing these optimization strategies, researchers can maximize the value of their Acetyl-HIST1H3A (K37) antibody studies while minimizing resource expenditure.

What considerations should guide the selection of Acetyl-HIST1H3A (K37) antibodies for specific research applications?

Selecting the appropriate Acetyl-HIST1H3A (K37) antibody requires careful consideration of several factors specific to the intended research application:

  • Antibody Format Considerations:

    • Polyclonal vs. Monoclonal: Polyclonal antibodies offer broader epitope recognition but may have higher batch-to-batch variability . Monoclonal antibodies provide consistency but may be more sensitive to epitope masking.

    • Host Species: Consider compatibility with other antibodies for co-staining experiments and available secondary antibodies

    • Purification Method: Antigen affinity-purified antibodies typically offer higher specificity

  • Application-Specific Requirements:

    • Western Blotting: Select antibodies validated specifically for WB with demonstrated specificity

    • ChIP/ChIP-seq: Choose antibodies that efficiently immunoprecipitate acetylated histones in native conditions

    • Immunofluorescence: Select antibodies that perform well in fixed samples and have low background

    • ELISA: Opt for antibodies with high affinity and quantitative binding properties

  • Validation Documentation:

    • Review validation data across multiple applications and cell types

    • Assess specificity testing against related acetylation sites

    • Evaluate performance in experimental contexts similar to your planned studies

    • Check for independent validation beyond manufacturer testing

  • Technical Support and Resources:

    • Consider manufacturers that provide detailed protocols for your specific application

    • Assess availability of technical support for troubleshooting

    • Look for products with recommended positive control samples or reference materials

    • Evaluate whether validation data is available for your model system

  • Selection Decision Framework:

    Research GoalPrimary ConsiderationSecondary ConsiderationRecommended Format
    Mechanistic studiesHigh specificityMultiple application validationMonoclonal
    Biomarker discoveryConsistency across samplesSpecies cross-reactivityMonoclonal
    Novel modification contextsBroader epitope recognitionSensitivityPolyclonal
    Therapeutic developmentReproducibilityGMP compliance optionsMonoclonal
    Multi-species researchVerified cross-reactivityConsistent performanceWell-characterized polyclonal

By carefully considering these factors, researchers can select Acetyl-HIST1H3A (K37) antibodies that are optimally suited to their specific research applications, maximizing the likelihood of successful experiments and reliable results.

What are the current gaps in our understanding of Acetyl-HIST1H3A (K37) and future research directions?

Despite advances in histone modification research, several significant knowledge gaps remain regarding Acetyl-HIST1H3A (K37), presenting important opportunities for future investigation:

  • Functional Significance: The precise role of K37 acetylation in transcriptional regulation remains incompletely characterized compared to well-studied modifications like K14 acetylation . Future research should focus on identifying specific genes and cellular processes regulated by this modification.

  • Enzymatic Regulation: The specific histone acetyltransferases (HATs) and histone deacetylases (HDACs) that modify K37 remain to be fully characterized. Identifying these enzymes will provide potential targets for modulating this modification in experimental and therapeutic contexts.

  • Tissue and Cell-Type Specificity: The patterns of K37 acetylation across different tissues, cell types, and developmental stages are not comprehensively mapped. Future studies employing tissue-specific profiling and single-cell approaches will address this gap.

  • Disease Associations: While histone acetylation broadly is implicated in multiple diseases, the specific contributions of K37 acetylation to pathological processes require further investigation, particularly in cancer, neurodegenerative disorders, and inflammatory conditions .

  • Technological Developments: Current antibody-based detection methods have limitations in specificity and sensitivity. Development of next-generation detection methods, including more specific antibodies and non-antibody based approaches, will advance the field.

  • Integration with Other Modifications: How K37 acetylation functions within the broader histone code and interacts with other modifications remains to be fully elucidated. Sequential ChIP and mass spectrometry approaches can address this gap.

  • Therapeutic Targeting: The potential for targeting K37 acetylation in disease treatment remains largely unexplored. Drug discovery efforts aimed at modifying this specific acetylation site could yield novel therapeutic approaches.

Future research addressing these gaps will significantly advance our understanding of Acetyl-HIST1H3A (K37) in chromatin biology and potentially reveal new therapeutic targets for epigenetic intervention.

What resources are available to researchers for staying updated on advances in histone acetylation research?

Researchers interested in histone acetylation, including Acetyl-HIST1H3A (K37), can leverage multiple resources to stay current with rapidly evolving advances in the field:

  • Scientific Databases and Repositories:

    • HistoneDB: A specialized database for histone proteins and their variations

    • Histone Modification Database: Catalogs known histone modifications and their functions

    • ENCODE Project: Provides genome-wide histone modification data across multiple cell types

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

  • Research Tools and Resources:

    • Antibody Validation Databases: Resources like Antibodypedia and EpiCypher's antibody certification program

    • Protocol Repositories: Sources like Protocol Exchange and Protocols.io for optimized experimental methods

    • Bioinformatic Tools: Specialized software for analyzing histone modification patterns from ChIP-seq data

  • Professional Organizations and Networks:

    • International Society for Epigenetics and Epigenomics

    • American Society for Biochemistry and Molecular Biology

    • Epigenetics Society

    • **Special interest groups within broader genetics and cell biology societies

  • Educational Resources:

    • Online Courses: Specialized courses on epigenetics and chromatin biology through platforms like Coursera and edX

    • Webinars and Virtual Symposia: Regular presentations on new techniques and findings

    • Technical Workshops: Hands-on training in specialized methods like ChIP-seq and mass spectrometry

  • Publication Alerts and Reviews:

    • Journal Alerts: Targeted alerts from journals specializing in epigenetics and chromatin

    • Review Series: Regular review series in journals like Nature Reviews Molecular Cell Biology

    • Preprint Servers: bioRxiv and medRxiv for early access to research findings

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.