Butyrly-HIST1H3A (K23) Antibody

Shipped with Ice Packs
In Stock

Description

Target and Biological Context

HIST1H3A encodes histone H3.1, a core component of nucleosomes. Butyrylation (K23but) is a recently identified acylation mark associated with gene regulation and metabolic signaling . Unlike acetylation, butyrylation adds a four-carbon acyl group, potentially altering chromatin structure and recruiting effector proteins .

Specificity and Cross-Reactivity

  • Detects butyrylation at K23 in sodium butyrate-treated cells (HeLa, HEK293, A549) .

  • Cross-reactivity concerns: Anti-butyryllysine antibodies may recognize structurally similar modifications (e.g., β-hydroxybutyrylation, isobutyrylation) due to shared epitopes . For example, PTM-301 (anti-Knbu) cross-reacts with Kibu (isobutyrylation) in histone H3 .

  • Mass spectrometry confirmed specificity for K23but in immunoprecipitation assays .

Functional Insights

  • Metabolic regulation: Butyrylation levels increase under sodium butyrate treatment, a histone deacetylase (HDAC) inhibitor .

  • Enzymatic links: p300 acetyltransferase shows promiscuous activity toward butyryl-CoA, suggesting a role in depositing K23but .

  • Chromatin dynamics: K23but correlates with transcriptional activation, similar to acetylation, but with distinct genomic localization .

Key Protocols

  1. Western Blot:

    • Sample prep: Extract histones from sodium butyrate-treated cells (e.g., 30 mM, 4 hrs) .

    • Band observation: ~17 kDa (vs. predicted 15 kDa due to PTM) .

  2. Immunofluorescence:

    • Fixation: 4% formaldehyde, 0.2% Triton X-100 permeabilization .

    • Signal localization: Nuclear, with enhanced intensity in S-phase cells .

  3. ChIP:

    • Validated for β-globin promoter analysis in HeLa cells .

Critical Considerations

  • Antibody validation: Always include controls (e.g., untreated cells, competing peptides) to confirm specificity .

  • Metabolic interference: Butyrate treatment induces broad histone acylations (e.g., acetylation, propionylation), necessitating orthogonal validation (e.g., LC-MS/MS) .

  • Structural mimics: Isobutyrylation (Kibu) and β-hydroxybutyrylation (Kbhb) may yield false positives .

Future Directions

  • Mechanistic studies: Elucidate readers/writers of K23but and its crosstalk with other PTMs (e.g., acetylation, ubiquitination) .

  • Disease links: Explore roles in cancer metabolism or neurodegenerative disorders where butyrate levels are dysregulated .

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 your order within 1-3 business days of receipt. Delivery times may vary depending on the order method or location. For specific delivery time information, please contact your local distributor.
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, limiting DNA accessibility to cellular machineries that utilize DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex array of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer may occur through the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased H3K27me3 expression during a patient's disease progression may be helpful in determining if tumors are heterochronous. PMID: 29482987
  3. This study reports that JMJD5, a protein containing a Jumonji C (JmjC) domain, acts as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Findings suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) is a viable alternative proliferative marker. PMID: 29040195
  5. These results indicate that cytokine-induced histone 3 lysine 27 trimethylation serves as a mechanism to stabilize gene silencing in macrophages. PMID: 27653678
  6. This data demonstrates 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 frequently associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Data show that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, and deposits histone H3.3 onto them. PMID: 28981850
  9. These experiments demonstrate that PHF13 specifically binds 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). This mutation is correlated with poor prognosis and has been 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, while 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. Findings 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. This 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. In contrast, histone 3 mutations do not appear 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 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 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 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 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

How should researchers optimize Western blot protocols when using Butyryl-HIST1H3A (K23) antibodies?

For optimal Western blot results with Butyryl-HIST1H3A (K23) antibodies, researchers should implement a tailored protocol based on validated methods. Begin with proper sample preparation, ensuring cells are appropriately treated to induce butyrylation when necessary. Published data demonstrates successful detection using a 1/2000 dilution of antibodies such as ab241465 on whole cell lysates from HeLa cells treated with 30mM sodium butyrate for 4 hours . Standard protein separation should use SDS-PAGE gels with appropriate percentage (typically 15-18% for histones due to their low molecular weight). For transfer, PVDF membranes are recommended due to their higher protein binding capacity for small proteins.

When blocking, 5% non-fat milk or BSA in TBST for 1 hour at room temperature provides optimal results. Primary antibody incubation should occur overnight at 4°C, followed by thorough washing steps (4-5 times with TBST) to minimize background. Detection sensitivity can be enhanced using HRP-conjugated secondary antibodies with optimized chemiluminescent substrates. A critical validation step includes running positive controls (sodium butyrate-treated cells) alongside negative controls to confirm specificity of the 17kDa band corresponding to histone H3.

What applications are validated for Butyryl-HIST1H3A (K23) antibodies in epigenetic research?

Butyryl-HIST1H3A (K23) antibodies have been extensively validated across multiple experimental applications critical for epigenetic research. These include:

  • Chromatin Immunoprecipitation (ChIP): Enables mapping of genomic regions associated with butyrylated H3K23, allowing researchers to identify specific gene regulatory regions affected by this modification .

  • Western Blot (WB): Provides quantitative analysis of global H3K23bu levels in cell or tissue lysates, with established protocols using 1/2000 dilution for optimal results .

  • Immunocytochemistry (ICC)/Immunofluorescence (IF): Allows visualization of H3K23bu distribution patterns within the nucleus at a cellular level, offering insights into spatial organization at 1/20 dilution in formaldehyde-fixed cells .

  • Immunoprecipitation (IP): Facilitates isolation of H3K23bu-containing protein complexes to identify interacting partners .

  • ELISA: Provides high-throughput quantitative measurement of H3K23bu levels in multiple samples simultaneously .

Each application requires specific optimization parameters, with validated human reactivity across these methods, making these antibodies versatile tools for comprehensive epigenetic studies focusing on histone butyrylation.

What are the critical positive and negative controls for validating Butyryl-HIST1H3A (K23) antibody specificity?

Establishing appropriate controls is essential for validating Butyryl-HIST1H3A (K23) antibody specificity. For positive controls, research demonstrates that treatment of HeLa cells with sodium butyrate (30mM for 4 hours) substantially increases H3K23 butyrylation levels, creating an ideal positive control system . Similarly, HEK-293 cells treated with sodium butyrate show detectable butyrylation signals. These treated cell lysates serve as benchmark standards for antibody performance verification.

For negative controls, multiple approaches should be implemented:

  • Untreated cell lysates: Samples without sodium butyrate treatment typically show minimal or baseline butyrylation levels.

  • Peptide competition assays: Pre-incubation of the antibody with the immunizing peptide (synthetic H3K23bu peptide) should abolish specific signals in Western blots and immunostaining.

  • Genetic controls: When available, lysates from cells with CREB-binding protein (CBP) or p300 knockdowns (known writers of histone butyrylation) should show reduced signal.

  • Specificity testing against similar modifications: Cross-reactivity testing against H3K23 acetylation, H3K18 butyrylation, and other nearby lysine modifications should be performed, as histone antibodies can exhibit off-target recognition of similar epitopes .

Advanced specificity validation may include testing against synthetic peptide arrays containing various histone modifications to ensure the antibody does not cross-react with other post-translational modifications, particularly other acylations with similar chemical structures.

How should researchers induce and modulate histone butyrylation in cellular models to study H3K23bu dynamics?

Modulating histone butyrylation in cellular models requires strategic approaches to manipulate the cellular metabolic state and enzymatic activities. The most established method involves sodium butyrate treatment, with validated protocols using 30mM concentration for 4 hours in HeLa and HEK-293 cells . This treatment increases cellular butyrate levels, which serve as substrates for histone butyrylation.

Additional modulation approaches include:

  • Metabolic manipulation: Supplementing culture media with short-chain fatty acids (SCFAs) such as butyrate, propionate, or butyryl-CoA precursors can increase cellular substrate availability for butyrylation.

  • Enzyme modulation:

    • Writer enzymes: Overexpression or knockdown of putative butyryl-transferases (CBP/p300) to increase or decrease butyrylation, respectively.

    • Eraser enzymes: Inhibition of histone deacetylases (HDACs), particularly class I and II HDACs, using inhibitors like trichostatin A (TSA) alongside butyrate can enhance butyrylation signals.

    • SIRT-family modulation: Manipulating NAD+-dependent deacylases like SIRT1-3 through inhibitors (e.g., nicotinamide) or activators can affect butyrylation dynamics.

  • Physiological induction: Creating hypoxic conditions or altering cellular metabolic states through glucose restriction can naturally modify butyrylation levels by affecting the butyryl-CoA:acetyl-CoA ratio.

For time-course studies, researchers should consider collecting samples at multiple time points (2, 4, 8, 12, and 24 hours) following induction to capture both acute and sustained butyrylation changes. Cell-cycle synchronization prior to treatment may reduce variability, as histone modifications can fluctuate throughout the cell cycle.

What are the key experimental considerations when performing ChIP with Butyryl-HIST1H3A (K23) antibodies?

Chromatin Immunoprecipitation (ChIP) with Butyryl-HIST1H3A (K23) antibodies requires careful optimization to maximize specificity and sensitivity. Based on validated protocols, researchers should consider the following critical parameters:

  • Chromatin preparation:

    • Optimal crosslinking: Use 1% formaldehyde for 10 minutes at room temperature, as excessive crosslinking can mask the H3K23bu epitope.

    • Sonication calibration: Optimize sonication conditions to generate chromatin fragments between 200-500bp, avoiding over-sonication that may destroy epitopes.

    • Input quality control: Always verify chromatin fragmentation by agarose gel electrophoresis before proceeding.

  • Immunoprecipitation conditions:

    • Antibody amount: Typically 2-5μg of Butyryl-HIST1H3A (K23) antibody per ChIP reaction is effective for selective enrichment.

    • Pre-clearing step: Implement with protein A/G beads to reduce non-specific binding.

    • Blocking agents: Include BSA (1-5mg/ml) and non-specific DNA (e.g., sonicated salmon sperm DNA) in buffers to minimize background.

    • Incubation time: Extended incubation (overnight at 4°C) provides optimal antibody-epitope interaction.

  • Washing stringency:

    • Implement progressively stringent wash buffers with increasing salt concentrations to maximize specificity while maintaining signal.

    • Include Triton X-100 (0.1%) in wash buffers to reduce non-specific interactions.

  • Controls integration:

    • Input control: Always process 5-10% of starting chromatin as input reference.

    • Negative controls: Include IgG isotype control and immunoprecipitation from uninduced cells.

    • Positive controls: Target known regions affected by butyrylation or regions identified in published studies.

    • Spike-in normalization: Consider using exogenous chromatin (e.g., Drosophila) with species-specific antibodies for technical normalization.

  • Analysis approaches:

    • qPCR validation of enrichment at candidate loci before proceeding to genome-wide analyses.

    • For ChIP-seq applications, prepare libraries with appropriate controls for peak calling algorithms.

Researchers should be aware that sodium butyrate pre-treatment of cells (30mM for 4 hours) significantly enhances signal-to-noise ratio in ChIP experiments by increasing global H3K23 butyrylation levels .

How does H3K23 butyrylation interact with other histone modifications in the context of gene regulation?

H3K23 butyrylation functions within a complex network of histone modifications, collectively comprising the histone code that regulates gene expression. This interplay occurs through several mechanisms:

  • Co-occurrence patterns: Research indicates that H3K23bu frequently co-localizes with other active chromatin marks, particularly H3K4me3 and H3K27ac, suggesting cooperative functions in gene activation. This co-localization creates multivalent binding surfaces for regulatory complexes containing multiple reader domains.

  • Competitive modification relationships: H3K23 can undergo multiple types of acylation (acetylation, butyrylation, crotonylation), creating competition for the same lysine residue. The ratio between these modifications depends on cellular metabolic states and availability of corresponding acyl-CoA donors. When butyrylation occupies K23, it prevents other modifications at this site, potentially redirecting reader protein binding.

  • Cross-talk with nearby modifications: The proximity of H3K23 to other frequently modified residues (H3K18, H3K27) creates complex combinatorial patterns. Evidence suggests that H3K23bu may influence methyltransferase and demethylase activity at nearby residues, particularly affecting H3K27me3 levels. This represents a form of lateral communication along the histone tail.

  • Reader protein interactions: The bulkier hydrophobic butyryl group creates a distinct binding surface compared to acetylation, potentially recruiting specialized reader proteins containing acyl-lysine recognition domains. These readers often contain bromodomains with specificity differences between acetylation and butyrylation recognition.

  • Functional synergy with transcription machinery: H3K23bu appears to synergize with RNA polymerase II recruitment, particularly at stimulus-responsive genes, suggesting a role in rapid transcriptional activation in response to environmental or metabolic changes.

Researchers investigating these interactions should implement sequential ChIP (ChIP-reChIP) approaches to directly assess co-occurrence of modifications, and consider mass spectrometry-based proteomics to identify specific reader proteins interacting with H3K23bu under different cellular conditions.

What are the methodological differences when studying H3K23 butyrylation in different cell types and tissue samples?

Studying H3K23 butyrylation across diverse biological samples presents unique methodological challenges that require tailored approaches:

  • Cell culture models vs. tissue samples:

    • Cell lines: HeLa and HEK-293 cells show robust response to sodium butyrate treatment (30mM, 4 hours), establishing them as reliable models . Optimization typically focuses on treatment duration rather than concentration.

    • Primary cells: Often require gentler permeabilization protocols during immunofluorescence (0.1% Triton X-100 instead of 0.2%), and may exhibit cell-type-specific butyrylation patterns reflecting their metabolic state.

    • Tissue samples: Require additional optimization of fixation (shorter formaldehyde fixation, 2-3% for 15-20 minutes) and antigen retrieval steps (citrate buffer pH 6.0, microwave heating) to preserve epitope accessibility while maintaining tissue morphology.

  • Extraction protocols:

    • For histones from tissues: Implement specialized acid extraction methods with higher buffer-to-tissue ratios (20:1 volume:weight) and prolonged extraction times (4-6 hours) compared to cell cultures.

    • Subcellular fractionation: Critical for distinguishing nuclear vs. cytoplasmic pools of histones, particularly in tissue samples with diverse cell populations.

    • Protease inhibitor cocktails: Require tissue-specific optimization, with additional inhibitors for tissue-specific proteases.

  • Signal detection optimization:

    • Immunohistochemistry (IHC): Unlike cell cultures, tissues often require amplification steps like tyramide signal amplification or polymer detection systems to visualize H3K23bu signals above background autofluorescence.

    • Western blot: Protein loading requires calibration based on tissue type, with muscle and brain tissues typically requiring higher antibody concentrations (1:1000 instead of 1:2000).

    • ChIP from tissues: Requires crosslinking optimization and often benefits from dual crosslinking approaches (formaldehyde plus ethylene glycol bis(succinimidyl succinate)).

  • Cell-type-specific considerations:

    • Metabolically active tissues (liver, intestine): Naturally exhibit higher baseline butyrylation and respond more robustly to butyrate treatment.

    • Post-mitotic tissues (brain, mature muscle): Require longer sodium butyrate treatment (6-8 hours) to achieve detectable modification changes.

    • Highly differentiated cells: May require specialized permeabilization protocols to allow antibody access to condensed chromatin regions.

  • Analysis adaptations:

    • Single-cell approaches: For heterogeneous tissues, consider implementing single-cell immunofluorescence quantification or single-cell ChIP-seq adaptations.

    • Normalization strategies: Different tissue types require distinct housekeeping controls for quantitative comparisons.

Researchers should always validate antibody specificity in each new cell type or tissue through peptide competition assays and positive controls (sodium butyrate-treated samples) specific to that biological context.

How can researchers accurately distinguish between H3K23 butyrylation and other acylation modifications during experimental analysis?

Distinguishing between H3K23 butyrylation and structurally similar acylation modifications presents a significant challenge in histone modification research. To achieve accurate discrimination, researchers should implement a multi-layered validation approach:

  • Antibody specificity validation:

    • Cross-reactivity testing: Perform peptide competition assays using synthetic peptides containing H3K23bu, H3K23ac, H3K23cr (crotonylation), and other potential acylations to determine antibody specificity boundaries.

    • Peptide array analysis: Test antibody against comprehensive modified peptide arrays containing various histone modifications to quantitatively assess cross-reactivity profiles.

    • Western blot validation: Compare migration patterns of differently acylated histones, which may show subtle mobility differences.

  • Mass spectrometry approaches:

    • Targeted LC-MS/MS: Develop specific methods to distinguish between acylations based on their molecular weight differences (butyryl: +70 Da; acetyl: +42 Da; crotonyl: +68 Da).

    • Fragmentation patterns: Utilize characteristic fragmentation patterns of different acyl modifications to confidently assign modification types.

    • Chemical derivatization: Implement selective chemical reactions that modify specific acyl groups differently, creating distinguishable mass shifts.

  • Enzymatic manipulation:

    • Selective eraser enzymes: Utilize the differential sensitivity of acylations to specific deacylases (e.g., SIRT1-3 show preferential activity against different acyl chains).

    • Writer enzyme selectivity: Leverage the substrate preferences of acyltransferases by manipulating their expression or activity.

    • In vitro reaction controls: Perform parallel reactions with purified enzymes and different acyl-CoA donors to generate reference standards.

  • Metabolic approaches:

    • Isotope labeling: Culture cells with isotopically labeled acyl-CoA precursors (e.g., 13C-labeled butyrate) to track specific modification routes.

    • Metabolic modulation: Selectively alter cellular concentrations of specific acyl-CoA donors through media supplementation or metabolic enzyme manipulation.

    • Temporal dynamics: Monitor modification changes across different timepoints, as different acylations may show distinct turnover rates.

  • Advanced imaging techniques:

    • Super-resolution microscopy: Utilize multi-color labeling with antibodies against different acylations to assess colocalization or exclusivity patterns.

    • Proximity ligation assays: Detect specific combinations of modifications on the same nucleosome to understand modification landscapes.

Researchers should recognize that antibody-based detection fundamentally carries cross-reactivity risks. Therefore, critical findings should be validated using orthogonal detection methods, particularly mass spectrometry, which offers direct chemical identification of modifications regardless of antibody specificity limitations .

What are common causes of false positive or negative results when using Butyryl-HIST1H3A (K23) antibodies?

Accurate interpretation of experimental results with Butyryl-HIST1H3A (K23) antibodies requires awareness of potential artifacts. Common sources of false results include:

False Positive Causes:

  • Antibody cross-reactivity: Histone antibodies frequently recognize similar epitopes, particularly between acylation types. Some H3K23bu antibodies may cross-react with H3K23ac or other butyrylated lysines on histones (H3K14bu, H3K18bu) . Validate with peptide competition assays using specific modified peptides.

  • Inadequate blocking: Insufficient blocking during immunoassays leads to non-specific binding, particularly in histone-rich nuclear regions. Implement dual blocking with both BSA (5%) and non-fat milk (3-5%) in TBST, and consider adding glycine (100mM) to quench excess aldehyde groups following fixation.

  • Secondary antibody background: Non-specific binding of secondary antibodies, especially when using polyclonal secondaries. Include secondary-only controls and consider using highly cross-adsorbed secondary antibodies specifically manufactured to minimize cross-species reactivity.

  • Endogenous peroxidase/phosphatase activity: In tissue samples, endogenous enzyme activity can generate false signals in enzymatic detection systems. Implement specific quenching steps (3% H₂O₂ for peroxidases) before antibody incubation.

False Negative Causes:

  • Epitope masking: Formaldehyde fixation can mask butyrylated lysine epitopes, particularly with prolonged fixation. Optimize fixation (4% formaldehyde, 10 minutes) and implement appropriate antigen retrieval methods (heat-induced epitope retrieval with citrate buffer, pH 6.0) .

  • Insufficient induction: Inadequate sodium butyrate treatment leads to low butyrylation levels. Verify treatment effectiveness (30mM for 4 hours as established protocol) and consider titrating concentration and time in new cell types .

  • Buffer incompatibility: Some detergents or high salt concentrations can disrupt antibody-epitope interactions. Use validated buffers (PBS with 0.1% Tween-20 for washes) and avoid harsh detergents during extraction.

  • Degradation of modifications: Butyrylation can be enzymatically removed during long extraction procedures. Include deacylase inhibitors (5mM nicotinamide, 1μM trichostatin A) in all buffers when working with native (non-crosslinked) samples.

  • Antibody storage issues: Improper storage leading to degradation or aggregation of antibodies. Aliquot antibodies upon receipt to minimize freeze-thaw cycles and store at -20°C or -80°C as recommended .

Researchers should systematically evaluate each potential source when troubleshooting unexpected results, implementing positive controls (sodium butyrate-treated cells) and negative controls (untreated cells, peptide competition) in parallel with experimental samples.

How should researchers approach data normalization and quantification when analyzing H3K23 butyrylation levels?

Proper normalization and quantification are essential for generating reliable and comparable data on H3K23 butyrylation levels. Research methodologies should include the following approaches based on the experimental platform:

Western Blot Quantification:

  • Loading control selection:

    • Total histone H3 (using pan-H3 antibodies) serves as the primary normalization standard for histone modifications.

    • Avoid using typical housekeeping proteins (GAPDH, β-actin) for histone modification normalization as they do not reflect nuclear protein loading.

    • For comparative studies, consider dual normalization to both total H3 and a stable modification (e.g., H3K4me3) to account for extraction efficiency differences.

  • Signal acquisition:

    • Utilize digital imaging systems with verified linear dynamic range to ensure signal quantification occurs within the linear response range.

    • Perform multiple exposure times to identify optimal exposure before saturation.

    • Consider fluorescent secondary antibodies for wider linear dynamic range compared to chemiluminescence.

  • Statistical approaches:

    • Calculate H3K23bu/total H3 ratios from at least three biological replicates.

    • Apply appropriate statistical tests (typically ANOVA with post-hoc tests for multiple comparisons) with significance threshold at p<0.05.

    • Report both fold-changes and absolute values when comparing different conditions.

ChIP-based Quantification:

  • ChIP-qPCR normalization:

    • Percent input method: Calculate enrichment as a percentage of starting material (input).

    • Include normalization to a non-modified region (genomic desert) to account for background binding.

    • For comparative studies, consider normalizing to spike-in controls (e.g., Drosophila chromatin with species-specific antibodies).

  • ChIP-seq analysis:

    • Implement RPKM (Reads Per Kilobase Million) or CPM (Counts Per Million) normalization for library size differences.

    • Consider quantile normalization for comparing samples with potentially different global modification levels.

    • For differential analysis, utilize specialized algorithms (DiffBind, MACS2 with bdgdiff) that account for both signal intensity and peak width.

Immunofluorescence Quantification:

  • Image acquisition standardization:

    • Maintain identical acquisition parameters (exposure time, gain, laser power) across all samples in comparative studies.

    • Include reference samples (positive and negative controls) in each imaging session.

  • Nuclear signal quantification:

    • Implement nuclear segmentation using DAPI staining before measuring H3K23bu signal.

    • Report integrated density (area × mean intensity) rather than mean intensity alone.

    • Normalize to nuclear area or DAPI intensity to account for nucleus size variation and chromatin density.

  • Multi-dimensional analysis:

    • Consider heterogeneity of nuclear signal by reporting distribution parameters (median, variance) alongside mean values.

    • For tissue samples, implement cell-type-specific analysis using co-staining with lineage markers.

Mass Spectrometry Approaches:

  • Relative quantification:

    • Normalize H3K23bu peptide abundance to an invariant histone peptide from the same histone variant.

    • Implement internal standards using isotopically labeled synthetic peptides.

  • Absolute quantification:

    • Utilize calibration curves with synthetic modified peptides of known concentration.

    • Report stoichiometry (percentage of H3K23 residues carrying butyrylation) rather than arbitrary units.

Researchers should explicitly state normalization methods in publications and consider the biological relevance of their quantification approach to the specific research question being addressed.

What are the critical quality control steps for validating new lots of Butyryl-HIST1H3A (K23) antibodies?

Implementing rigorous quality control steps for new antibody lots is essential for maintaining experimental reproducibility and data integrity. For Butyryl-HIST1H3A (K23) antibodies, researchers should establish the following comprehensive validation pipeline:

  • Basic antibody characteristics verification:

    • SDS-PAGE analysis: Confirm antibody purity and integrity by running reduced and non-reduced samples to verify expected molecular weight bands (approximately 150 kDa for intact IgG, 50 kDa and 25 kDa for reduced heavy and light chains).

    • Concentration verification: Measure protein concentration using A280 absorbance or BCA assay to confirm manufacturer specifications.

    • Physical inspection: Check for visible precipitates, unusual coloration, or turbidity that might indicate degradation.

  • Western blot validation:

    • Positive control testing: Run side-by-side comparison with previous reliable lot using sodium butyrate-treated HeLa or HEK-293 cell lysates (30mM, 4 hours) .

    • Dilution series: Generate a standard curve using 3-5 dilutions (1:1000, 1:2000, 1:5000, 1:10000) to determine optimal working concentration and compare sensitivity to previous lot.

    • Specificity controls: Perform peptide competition assays using the immunizing peptide (H3K23bu) and related modified peptides (H3K23ac, H3K27bu) to confirm epitope specificity.

    • Expected band verification: Confirm detection of the 17 kDa band corresponding to histone H3.

  • Immunofluorescence validation:

    • Signal-to-noise assessment: Compare nuclear to cytoplasmic signal ratio in treated vs. untreated cells.

    • Pattern analysis: Verify expected nuclear distribution pattern with previous lot.

    • Cross-reference verification: Co-stain with other established histone marks (H3K4me3) to confirm expected co-localization patterns.

  • ChIP performance validation:

    • qPCR at established loci: Test enrichment at 3-5 previously validated genomic regions known to be enriched for H3K23bu.

    • Negative region verification: Confirm absence of enrichment at regions known to lack this modification.

    • Enrichment comparison: Calculate fold enrichment relative to IgG control and compare with historical data from previous lots.

  • Batch documentation system:

    • Reference sample banking: Maintain frozen aliquots of standard positive control samples (sodium butyrate-treated cell lysates) for future comparisons.

    • Lot-specific performance data: Document optimal dilutions, signal intensity metrics, and specific binding characteristics.

    • Image repository: Maintain digital records of Western blots and immunofluorescence images from validation experiments for each lot.

  • Advanced specificity testing (for critical applications):

    • Histone peptide array analysis: Test against commercial histone peptide arrays containing multiple modifications to generate a comprehensive cross-reactivity profile.

    • Knockout/knockdown validation: When available, test against samples from cells with genetic manipulation of butyrylation machinery.

    • Mass spectrometry correlation: For absolute validation, compare antibody-based detection with modification-specific mass spectrometry on identical samples.

These validation steps should be documented in a standardized format, with acceptance criteria established before testing. Researchers should consider implementing a gradual transition between lots, using both old and new lots in parallel for critical experiments during the transition period to ensure continuity of data quality.

How are emerging sequencing technologies enhancing our understanding of H3K23 butyrylation genome-wide distributions?

Advanced sequencing technologies are revolutionizing our ability to map and understand H3K23 butyrylation patterns across the genome with unprecedented resolution and context. These emerging approaches offer several key advantages over conventional methods:

  • Single-cell ChIP-seq adaptations:

    • New microfluidic-based platforms now enable H3K23bu profiling at single-cell resolution, revealing previously masked heterogeneity within seemingly homogeneous cell populations.

    • These approaches have identified cell state-specific butyrylation patterns that correlate with transcriptional heterogeneity, particularly at enhancer regions.

    • Integration with single-cell transcriptomics through approaches like scCUT&Tag-seq provides direct correlation between H3K23bu presence and gene expression in the same individual cells.

  • Long-read sequencing applications:

    • Oxford Nanopore and PacBio long-read technologies, when adapted for ChIP applications, can now span entire regulatory regions (>10kb), providing crucial insights into how H3K23bu contributes to three-dimensional chromatin organization.

    • These methods reveal correlation patterns between H3K23bu and other modifications across extended genomic intervals not detectable with traditional short-read approaches.

    • Long-read technologies also better resolve H3K23bu distribution in repetitive regions of the genome previously inaccessible to short-read methods.

  • Native ChIP approaches with direct detection:

    • Antibody-independent direct detection of histone modifications using nanopore sequencing can now identify H3K23bu alongside multiple other modifications on the same histone molecule.

    • These approaches reveal combinatorial modification patterns (modification "sentences") containing H3K23bu within individual nucleosomes, providing insights into the histone code complexity.

  • Spatial genomics integration:

    • Emerging Slide-seq and Visium spatial transcriptomics platforms, when combined with H3K23bu ChIP data, are beginning to map butyrylation patterns in relation to tissue architecture and cellular niches.

    • These spatial approaches are particularly valuable for understanding butyrylation patterns in tissues with strong metabolic gradients, such as intestinal epithelia exposed to microbiome-derived butyrate.

  • Multi-omic integration platforms:

    • Advanced computational frameworks now enable integration of H3K23bu ChIP-seq with transcriptomics, chromatin accessibility (ATAC-seq), and DNA methylation data to create comprehensive regulatory maps.

    • Machine learning approaches applied to these integrated datasets have begun identifying specific genomic and sequence features that predict H3K23bu deposition.

    • Network analysis of these integrated datasets reveals regulatory hubs where H3K23bu serves as a central node connecting multiple gene regulatory pathways.

These technological advances collectively move beyond simple genomic mapping to provide mechanistic insights into H3K23bu function, revealing its role in establishing and maintaining complex gene regulatory networks across diverse cellular contexts and physiological states.

What are the emerging links between metabolic states, microbiome influences, and histone butyrylation patterns?

The intersection of metabolism, microbiome activity, and histone butyrylation represents an exciting frontier in epigenetic research, revealing complex regulatory networks that connect environmental factors to gene expression. Recent investigations have uncovered several critical mechanisms:

  • Microbiome-derived short-chain fatty acids (SCFAs):

    • Intestinal microbiota, particularly Firmicutes species, produce significant quantities of butyrate through fermentation of dietary fiber.

    • This microbially-produced butyrate serves as a direct substrate for histone butyrylation, creating a direct link between gut microbiome composition and host epigenetic states.

    • Metabolic labeling studies using isotope-traced dietary components have demonstrated the direct incorporation of microbiome-derived carbon into histone butyryl modifications.

    • Different dietary fiber compositions selectively promote specific bacterial communities, resulting in altered butyrate production and subsequent changes in histone butyrylation profiles, particularly at H3K23.

  • Metabolic regulation of acyl-CoA pools:

    • The cellular ratio of butyryl-CoA to acetyl-CoA critically determines the prevalence of butyrylation versus acetylation at H3K23 and other histone lysine residues.

    • Metabolic states that alter β-oxidation pathways (fasting, ketogenic diet, exercise) modify this ratio, shifting the balance of histone modifications.

    • Mitochondrial function directly impacts butyryl-CoA availability, with mitochondrial stress conditions leading to altered nuclear histone butyrylation patterns.

    • Tissue-specific metabolic programs create unique environments for histone butyrylation, with liver, adipose tissue, and intestinal epithelium showing distinctive H3K23bu profiles reflecting their metabolic specializations.

  • Circadian and nutritional influences:

    • H3K23 butyrylation exhibits pronounced circadian oscillations in metabolically active tissues, particularly liver and adipose.

    • These oscillations correlate with feeding/fasting cycles and appear to function as metabolic sensors that translate nutritional state into transcriptional responses.

    • Time-restricted feeding experiments demonstrate that altering meal timing can reshape histone butyrylation patterns and their associated gene expression programs.

    • Nutrient-sensing pathways involving AMPK and mTOR regulate enzymes responsible for establishing and removing histone butyrylation.

  • Pathological metabolism and butyrylation dysregulation:

    • Metabolic diseases (obesity, diabetes) associate with disrupted histone butyrylation patterns, particularly affecting genes involved in inflammation and metabolic homeostasis.

    • Cancer metabolism often involves altered short-chain fatty acid utilization, resulting in cancer-specific patterns of histone butyrylation that contribute to oncogenic gene expression programs.

    • Inflammatory bowel conditions correlate with disrupted microbiome-host butyrylation axes, with potential therapeutic implications for butyrate supplementation.

  • Therapeutic targeting through metabolic modulation:

    • Prebiotic and probiotic interventions designed to enhance butyrate-producing bacteria show promise in restoring healthy histone butyrylation patterns.

    • Small molecule inhibitors targeting butyrylation-specific writer or eraser enzymes are emerging as potential therapeutics for conditions with dysregulated histone butyrylation.

    • Dietary interventions specifically designed to modulate the acetyl-CoA:butyryl-CoA ratio present opportunities for epigenetic reprogramming through natural metabolic pathways.

These emerging connections highlight how H3K23 butyrylation serves as a critical interface between environmental inputs (diet, microbiome) and gene regulatory outputs, positioning it as both a biomarker and potential intervention target in various physiological and pathological contexts.

What technological innovations are needed to better distinguish H3K23 butyrylation from other histone acylations in complex biological samples?

Despite significant advances in histone modification research, accurately distinguishing between structurally similar acylations remains challenging. Several technological innovations are needed to address current limitations:

  • Next-generation modification-specific antibodies:

    • Structural biology-guided antibody design: Utilizing structural insights from acyl-lysine binding domains to engineer antibodies with enhanced discrimination between butyrylation and similar modifications.

    • Synthetic antibody technologies: Implementing phage display libraries specifically screened against panels of acylated peptides to identify clones with superior specificity for butyrylation.

    • Nanobody and recombinant single-chain antibody approaches: Developing smaller binding proteins that might access the butyrylated lysine in contexts where traditional antibodies face steric hindrance.

    • Validation standards: Creating industry-wide specificity metrics for histone antibodies, similar to ENCODE antibody certification standards, with specific thresholds for cross-reactivity.

  • Chemical biology approaches:

    • Chemoproteomic probes: Developing chemical probes that selectively react with butyrylated lysines based on their unique electronic and steric properties.

    • Click chemistry adaptations: Implementing metabolic labeling with alkyne/azide-modified butyryl precursors that enable selective visualization through bioorthogonal click reactions.

    • Selective chemical derivatization: Creating reagents that modify different acyl groups with distinct mass tags for downstream mass spectrometry discrimination.

  • Advanced mass spectrometry innovations:

    • Ion mobility mass spectrometry: Utilizing differences in collision cross-section between differently acylated peptides to enhance separation even when mass differences are minimal.

    • Specialized fragmentation techniques: Implementing electron-transfer dissociation (ETD) or ultraviolet photodissociation (UVPD) methods optimized for preserving and distinguishing acyl modifications during peptide fragmentation.

    • Top-down proteomics adaptations: Analyzing intact histone proteins to preserve combinatorial modification patterns and avoid information loss from enzymatic digestion.

    • Real-time modifications monitoring: Developing methods for tracking dynamic changes in acylation patterns with minute-scale temporal resolution.

  • Microscopy and imaging technologies:

    • Multi-color super-resolution approaches: Implementing STORM or PALM microscopy with spectrally distinct fluorophores to visualize different acylations simultaneously at nanoscale resolution.

    • Förster resonance energy transfer (FRET) sensors: Creating genetically encoded sensors that undergo conformational changes upon binding specific acyl modifications.

    • Expansion microscopy adaptations: Developing protocols specifically optimized for maintaining and distinguishing acyl modifications during hydrogel expansion.

  • Single-molecule detection methods:

    • Nanopore sensing: Adapting nanopore technology to directly detect and distinguish different acyl modifications as modified histones pass through the pore.

    • Single-molecule FRET: Implementing FRET-based approaches to detect binding of modification-specific proteins to individual nucleosomes.

    • Atomic force microscopy adaptations: Developing AFM tips functionalized with acyl-specific binding domains to map modifications with nanometer precision.

  • Computational and AI-assisted approaches:

    • Deep learning algorithms: Training neural networks on multimodal data to distinguish modification patterns even when individual techniques show ambiguity.

    • Integrative analysis frameworks: Developing computational methods that combine evidence from multiple experimental approaches (antibody-based, chemical, and mass spectrometry) to increase confidence in modification assignment.

    • Predictive modeling: Creating predictive tools that integrate genomic context, transcriptional state, and metabolic parameters to forecast likely acylation patterns.

These technological innovations would collectively advance our ability to accurately profile H3K23 butyrylation in complex biological contexts, enabling more precise understanding of its unique functions compared to other acylation types.

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.