Acetyl-HIST1H3A (K79) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Overview

Target: Acetylated lysine 79 on histone H3.1 (encoded by the HIST1H3A gene) .
Synonyms: H3/A, H3F3, H3FA, Histone H3.1 .
Species Reactivity: Primarily validated in Homo sapiens (human), with potential cross-reactivity in Canis familiaris (canine) based on immunogen design .

Key Applications

ApplicationValidation SourceDetails
Western BlotHeLa cell extracts Detects ~15 kDa band; enhanced signal in sodium butyrate-treated cells .
ChIPHeLa, U2OS cells Validated in chromatin immunoprecipitation assays for promoter analysis .
ImmunofluorescenceHeLa/U2OS Nuclear staining; specificity confirmed via peptide competition assays .
ELISAQuantitative assays Linear detection range: 0.2–100 pmol acetylated peptide .

Specificity

  • No cross-reactivity with acetylated H3K4, H3K9, H3K14, H3K18, H3K23, H3K27, H3K36, H3K56, or H3K122 .

  • Epitope recognition confirmed via peptide array and blocking experiments .

Functional Implications of H3K79 Acetylation

  • Transcriptional Activation: K79ac correlates with active chromatin regions, contrasting with K79 methylation (e.g., H3K79me3), which marks silent loci .

  • Cellular Response: Sodium butyrate treatment increases K79ac levels, suggesting a role in stress-induced chromatin remodeling .

Technical Considerations

  • Storage: Stable in 30% glycerol at -20°C; avoid freeze-thaw cycles .

  • Dilution Range:

    • WB: 1:500–1:2000

    • IF: 1:300–1:1000

    • ChIP: 2–5 µg per reaction .

Limitations

  • Species Restrictions: Limited reactivity outside mammals .

  • Exclusivity: Does not detect other H3 isoforms (e.g., H3.3) .

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 products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
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 the nucleosome, a fundamental unit of chromatin. Nucleosomes are responsible for packaging and compacting DNA, limiting its accessibility to cellular machinery that requires DNA as a template. Histones, therefore, play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex interplay of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer may be influenced by the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Studies have shown that increased expression of H3K27me3 during a patient's clinical course can be a helpful indicator for determining whether tumors are heterochronous. PMID: 29482987
  3. Recent findings indicate 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. Evidence suggests that the Ki-67 antigen proliferative index has limitations, and phosphohistone H3 (PHH3) is a potential alternative proliferative marker. PMID: 29040195
  5. These results identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms in the early developing human brain. 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 frequently associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Research demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, and deposits histone H3.3 onto them. PMID: 28981850
  9. Experiments have shown 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 study 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. 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. Research suggests 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 the 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) decrease 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) plays a crucial 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. 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 are 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 (K79) and its biological significance?

Acetyl-HIST1H3A (K79) refers to the acetylation of lysine 79 on histone H3.1, a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries that require DNA as a template. Histones play a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability through their post-translational modifications . Acetylation at K79, similar to other histone acetylation marks, is believed to contribute to transcriptional activation by loosening chromatin structure, though its specific functions may differ from better-characterized sites like K14, which functions in transcriptional activation, chromatin accessibility, cellular identity, and epigenetic memory .

What applications are suitable for Acetyl-HIST1H3A (K79) antibodies?

Acetyl-HIST1H3A (K79) antibodies are suitable for multiple experimental applications including:

  • Chromatin Immunoprecipitation (ChIP): For mapping genomic locations enriched with K79 acetylation

  • Western Blotting (WB): For detecting and quantifying total levels of K79 acetylation in protein extracts

  • Immunocytochemistry/Immunofluorescence (ICC/IF): For visualizing the nuclear distribution pattern of K79 acetylation

  • ELISA: For quantitative measurement of K79 acetylation levels

  • Peptide Array (PepArr): For testing antibody specificity against modified peptides

The working dilutions should be optimized for each application, starting with manufacturer recommendations such as 1:500-1:2000 for WB and 1:50-1:500 for ICC as reference points based on similar histone modification antibodies .

How do I validate the specificity of an Acetyl-HIST1H3A (K79) antibody?

Validating antibody specificity is crucial for reliable experimental results. A systematic approach includes:

  • Peptide competition assays: Pre-incubating the antibody with acetylated K79 peptides should abolish signal, while incubation with unmodified or differently modified peptides should not affect signal .

  • Cross-reactivity testing: Using peptide arrays to confirm that the antibody does not cross-react with similar modifications (e.g., K79 methylation or acetylation at other lysine residues) .

  • Genetic validation: Using cells with mutations at K79 that prevent acetylation or with knockdown/knockout of relevant acetyltransferases.

  • Western blot analysis: Confirming a single band of appropriate molecular weight (~15-17 kDa for histone H3).

  • ChIP-seq with spike-in controls: Including exogenous chromatin with known K79 acetylation status as internal controls.

What are the recommended storage and handling conditions for these antibodies?

Based on standard practices for similar recombinant and polyclonal antibodies:

  • Storage temperature: Most histone modification antibodies should be stored at -20°C for long-term storage and at 4°C for short-term use .

  • Avoid freeze-thaw cycles: Aliquot antibodies upon receipt to minimize freeze-thaw cycles, which can degrade antibody quality.

  • Working dilution preparation: Dilute only the amount needed for immediate experiments in appropriate buffer containing a carrier protein (often BSA) to prevent adsorption to tubes.

  • Handling precautions: Follow non-reducing conditions for Western blot applications unless specifically recommended by the manufacturer.

How does K79 acetylation interact with other histone modifications in the histone code context?

The histone code hypothesis proposes that combinations of modifications create specific binding platforms for chromatin-associated proteins. For K79 acetylation:

  • Modification crosstalk: Similar to other histone acetylation marks, K79ac likely functions in concert with other modifications. The presence of K79 acetylation may influence the deposition or removal of nearby modifications, creating combinatorial patterns that dictate specific transcriptional outcomes.

  • Reader protein specificity: Different bromodomain-containing proteins may recognize K79ac in combination with other acetylation marks, leading to recruitment of specific transcriptional machinery.

  • Integration with methylation: K79 can also be methylated (mono-, di-, or tri-methylated) , suggesting a potential regulatory switch between acetylation and methylation at this residue similar to what occurs at other lysine residues like K4, K9, and K27.

  • Genome-wide distribution patterns: ChIP-seq analyses using K79ac antibodies can reveal how this modification co-localizes with other histone marks across the genome, providing insight into its functional roles in different chromatin contexts.

What are the experimental challenges in detecting K79 acetylation compared to other histone modifications?

Several technical challenges are specific to studying K79 acetylation:

  • Antibody specificity: Due to the similar chemical nature of acetylation at different lysine residues, ensuring antibody specificity is crucial. Rigorous validation using peptide arrays and competition assays is necessary .

  • Modification abundance: If K79 acetylation is less abundant than other well-studied acetylation sites (like K9, K14, K27), more sensitive detection methods may be required.

  • Sample preparation: Optimal fixation and extraction protocols need to be established to preserve K79 acetylation while efficiently extracting chromatin-bound histones.

  • Epitope masking: Neighboring modifications or protein interactions might mask the K79ac epitope, leading to false-negative results in some contexts.

  • ChIP efficiency: Optimization of sonication conditions, antibody concentrations, and immunoprecipitation protocols specifically for K79ac ChIP experiments is necessary for high signal-to-noise ratios.

How can I design effective ChIP-seq experiments using Acetyl-HIST1H3A (K79) antibodies?

Effective ChIP-seq experimental design for K79ac includes:

  • Antibody selection: Choose antibodies validated specifically for ChIP applications , preferably with published ChIP-seq datasets demonstrating their performance.

  • Controls:

    • Input controls: Sequencing chromatin before immunoprecipitation

    • IgG controls: Using matched isotype IgG for non-specific binding assessment

    • Spike-in controls: Using exogenous chromatin from different species for normalization

    • Positive/negative genomic regions: Including primers for regions known to be enriched/depleted for K79ac

  • Chromatin preparation:

    • Optimal crosslinking time (typically 10-15 minutes with 1% formaldehyde)

    • Appropriate sonication to generate 200-500 bp fragments

    • Quality assessment of sheared chromatin

  • Sequencing depth: Aim for 20-40 million uniquely mapped reads per sample for histone modification ChIP-seq.

  • Bioinformatic analysis pipeline:

    • Quality control metrics specific for histone acetylation marks

    • Appropriate peak calling algorithms (broad vs. narrow peaks)

    • Integration with other genomic datasets

How should I approach quantitative comparison of K79 acetylation levels across different experimental conditions?

For rigorous quantitative comparisons:

  • Standardized sample processing: Process all samples simultaneously using identical protocols for histone extraction, antibody incubation, and detection methods.

  • Normalization strategies:

    • For Western blots: Normalize K79ac signal to total H3 levels

    • For ChIP-qPCR: Use percentage of input or normalization to invariant regions

    • For ChIP-seq: Implement spike-in normalization using exogenous chromatin

  • Technical replicates: Include multiple technical replicates to assess method variability.

  • Biological replicates: Analyze at least three independent biological replicates to account for biological variation.

  • Statistical analysis: Apply appropriate statistical tests based on experimental design and data distribution.

  • Dynamic range considerations: Ensure detection methods remain within linear dynamic range using standard curves if applicable.

What are common troubleshooting strategies for weak or inconsistent signals in K79ac detection?

When encountering weak or inconsistent signals:

  • Antibody concentration optimization: Titrate antibody concentrations to determine optimal working dilutions for each application .

  • Epitope retrieval methods: For fixed samples, test different antigen retrieval methods (heat-induced vs. enzymatic) to expose the K79ac epitope.

  • Blocking conditions: Optimize blocking reagents (BSA, non-fat milk, serum) and durations to reduce background while preserving specific signal.

  • Detection system sensitivity: Consider using more sensitive detection methods such as enhanced chemiluminescence for Western blots or amplification systems for immunofluorescence.

  • Sample preparation: Ensure histones are properly extracted and denatured; consider using histone extraction kits specifically designed to preserve acetylation marks.

  • Fresh antibody aliquots: Antibody activity may decrease with repeated freeze-thaw cycles; use fresh aliquots when possible.

How can I resolve contradictory results when mapping K79 acetylation using different antibody clones?

When different antibody clones yield contradictory results:

  • Epitope differences: Different antibodies may recognize slightly different epitopes surrounding K79ac, leading to context-dependent detection differences. Map the exact epitope recognized by each antibody.

  • Clone comparison experiments: Perform side-by-side experiments using multiple antibody clones (monoclonal and polyclonal) on identical samples .

  • Orthogonal validation: Confirm results using non-antibody methods like mass spectrometry to directly detect K79ac.

  • Sequential ChIP: Perform sequential ChIP (re-ChIP) using different antibody clones to identify regions recognized by both antibodies.

  • Genetic validation: Use systems where K79 is mutated or where relevant acetyltransferases are depleted to confirm specificity.

  • Context specificity: Determine if contradictions are context-specific (e.g., cell type-dependent, locus-specific) by systematically varying experimental conditions.

What controls should I include when studying the dynamics of K79 acetylation during cellular processes?

For studying K79 acetylation dynamics, include:

  • Time-course controls: Sample collection at consistent time points across biological replicates.

  • Treatment controls:

    • Positive controls: HDAC inhibitors (like TSA or sodium butyrate) to increase global acetylation levels

    • Negative controls: Acetyltransferase inhibitors to reduce acetylation

    • Vehicle controls: For any treatments involving solvents

  • Cellular state markers: Monitor markers of cell cycle, differentiation status, or stress response relevant to your experimental context.

  • Other histone modifications: Track well-characterized histone modifications (H3K27ac, H3K9ac) as comparative controls.

  • Total histone levels: Monitor total H3 levels to account for potential changes in histone abundance.

  • Pathway validation: Include readouts of pathways known to influence histone acetylation (e.g., metabolic state indicators like Acetyl-CoA levels).

How can I integrate K79 acetylation data with other omics approaches for comprehensive epigenetic profiling?

Multi-omics integration strategies include:

  • ChIP-seq integration with transcriptomic data: Correlate K79ac enrichment patterns with RNA-seq data to establish functional relationships with gene expression.

  • ATAC-seq or DNase-seq correlation: Determine relationship between K79ac and chromatin accessibility.

  • HiC or chromosome conformation capture techniques: Investigate potential roles of K79ac in three-dimensional chromatin organization.

  • Single-cell approaches: Apply single-cell versions of ChIP-seq, ATAC-seq, and RNA-seq to understand heterogeneity in K79ac distribution and its functional consequences.

  • Computational integration: Use machine learning approaches to integrate K79ac ChIP-seq with other histone modifications to predict functional genomic elements or expression patterns.

  • Mass spectrometry integration: Combine antibody-based detection with proteomic approaches to identify proteins associated with K79-acetylated histones.

What emerging technologies are enhancing the study of K79 acetylation and its functional consequences?

Cutting-edge technologies for K79ac research include:

  • CUT&RUN and CUT&Tag: These techniques offer higher signal-to-noise ratios than traditional ChIP-seq and require fewer cells, enabling more sensitive detection of K79ac.

  • CRISPR-based epigenome editing: Using dCas9 fused to acetyltransferases to specifically introduce K79ac at defined genomic loci to test functional consequences.

  • Live-cell imaging of acetylation dynamics: Developing specific sensors for real-time visualization of K79ac fluctuations in living cells.

  • Single-molecule approaches: Applying techniques like single-molecule FRET to study how K79ac affects nucleosome stability and dynamics at the molecular level.

  • Long-read sequencing applications: Using long-read technologies to map K79ac across repetitive regions or to link distant modifications within the same chromatin fragment.

  • Spatial omics approaches: Developing methods to map K79ac distribution in a spatially resolved manner within tissue contexts.

How can I design experiments to elucidate the specific cellular functions of K79 acetylation?

To determine K79ac functions:

  • Site-specific mutagenesis: Generate K79R (non-acetylatable) or K79Q (acetylation-mimicking) mutations and assess phenotypic consequences.

  • Acetyltransferase/deacetylase identification: Perform enzymatic activity assays or protein interaction studies to identify enzymes that regulate K79 acetylation.

  • Reader protein identification: Use techniques like SILAC-MS with acetylated vs. non-acetylated K79 peptide pulldowns to identify proteins that specifically recognize K79ac.

  • Temporal dynamics studies: Track K79ac levels during processes like cell cycle progression, differentiation, or response to stimuli to identify potential regulatory roles.

  • Locus-specific manipulation: Use CRISPR-based approaches to target acetyltransferases or deacetylases to specific loci to determine local effects of K79ac modulation.

  • Disease model studies: Examine K79ac alterations in disease states to uncover potential pathological roles.

What are the optimal sample preparation protocols for maximizing K79 acetylation detection?

For optimal K79ac detection:

  • Cell/tissue fixation: Use freshly prepared 1% formaldehyde for 10-15 minutes at room temperature, followed by quenching with glycine.

  • Histone extraction protocol selection:

    • Acid extraction: Particularly effective for preserving histone modifications

    • Triton extraction: Useful for nuclear fraction enrichment

    • Commercial histone extraction kits optimized for preserving acetylation marks

  • Protease and HDAC inhibitors: Include comprehensive protease inhibitor cocktails and HDAC inhibitors (sodium butyrate, TSA) during extraction to prevent degradation and deacetylation.

  • Gentle handling: Minimize mechanical stress and processing time to preserve labile modifications.

  • Storage considerations: Store extracted histones at -80°C in small aliquots to avoid repeated freeze-thaw cycles.

How should I optimize ChIP-qPCR experiments specifically for K79 acetylation studies?

For K79ac ChIP-qPCR optimization:

  • Antibody amount titration: Test multiple antibody concentrations (2-10 μg per ChIP reaction) to determine optimal signal-to-noise ratio.

  • Chromatin amount optimization: Standardize input chromatin quantity (typically 20-50 μg per reaction).

  • Primer design considerations:

    • Design primers for regions expected to be enriched for K79ac (based on literature or hypothesis)

    • Include primers for negative regions (heterochromatic regions)

    • Include primers for positive control regions (known acetylation-rich promoters)

  • Sonication optimization: Aim for chromatin fragments of 200-500 bp, verified by gel electrophoresis.

  • Washing stringency: Optimize salt concentration in wash buffers to reduce background while maintaining specific signal.

  • Data normalization: Calculate percent input or fold enrichment over IgG control for accurate quantification.

What quantitative approaches provide the most accurate measurement of K79 acetylation levels?

For accurate K79ac quantification:

  • ELISA-based methods: Commercial ELISA kits can provide quantitative measurement with detection ranges as low as 23.5 pg/mL and sensitivity of 5.8 pg/mL .

  • Western blotting with standard curves: Include a dilution series of recombinant acetylated histones to create standard curves for quantification.

  • Mass spectrometry approaches:

    • Targeted MS using isotope-labeled internal standards

    • Multiple reaction monitoring (MRM) for sensitive and specific quantification

    • SILAC labeling for relative quantification between conditions

  • ChIP-qPCR absolute quantification: Use spike-in controls of known concentration for absolute quantification of enrichment.

  • Flow cytometry: For single-cell quantification of global K79ac levels using permeabilized cells and fluorescently-labeled antibodies.

  • Image analysis quantification: For immunofluorescence data, use appropriate image analysis software with background subtraction and internal calibration standards.

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.