Crotonyl-HIST1H2AG (K36) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Crotonyl-HIST1H2AG (K36) Antibody

The Crotonyl-HIST1H2AG (K36) Antibody is a specialized immunological reagent designed to specifically recognize and bind to crotonylation at the lysine 36 position of Histone H2A type 1 protein in humans. Histone H2A type 1, also known as H2A.1 or Histone H2A/ptl, is a core component of nucleosomes that plays a central role in DNA packaging, chromatin structure, and gene regulation . The antibody enables researchers to detect and study this specific post-translational modification, which belongs to the expanding family of short-chain lysine acylations that have emerged as important epigenetic marks .

Histone crotonylation (Kcr) was identified as a novel histone mark relatively recently and has been shown to have distinct functional properties from the more extensively studied histone acetylation . The Crotonyl-HIST1H2AG (K36) Antibody provides scientists with a tool to specifically investigate this modification at a particular site on a specific histone protein, allowing for more precise understanding of epigenetic regulation mechanisms.

Applications and Research Uses

The Crotonyl-HIST1H2AG (K36) Antibody has been validated for several research applications, providing scientists with versatile tools for investigating histone crotonylation in various experimental contexts:

Validated Applications

The antibody has been tested and validated for the following applications:

  1. Enzyme-Linked Immunosorbent Assay (ELISA): Useful for quantitative detection of crotonylated histone H2A in solution .

  2. Immunocytochemistry (ICC): Enables visualization of the subcellular localization of crotonylated histone H2A in cultured cells .

Recommended Dilutions

For optimal results, the following dilutions are recommended:

  • Immunocytochemistry (ICC): 1:10 to 1:100 dilution

Experimental Evidence

Immunocytochemistry analysis has been performed using this antibody diluted at 1:50 for staining in HeLa cells that were treated with 30mM sodium crotonylate for 4 hours. The protocol involved fixing cells in 4% formaldehyde, permeabilizing using 0.2% Triton X-100, and blocking with 10% normal goat serum for 30 minutes at room temperature. The primary antibody (in 1% BSA) was incubated at 4°C overnight and subsequently detected using a biotinylated secondary antibody and visualized with an HRP conjugated SP system .

Background on Histone Crotonylation

While the Crotonyl-HIST1H2AG (K36) Antibody targets a specific crotonylation site, understanding the broader context of histone crotonylation is essential for appreciating the significance of this research tool.

Histone Crotonylation as an Epigenetic Mark

Histone crotonylation is a post-translational modification of lysine residues that was identified relatively recently. It belongs to an expanding group of short chain lysine acylations that also includes the extensively studied mark histone acetylation . Emerging evidence suggests that histone crotonylation is functionally distinct from histone acetylation and that competition for sites of modification, which reflects cellular metabolic status, could be an important epigenetic mechanism regulating diverse processes .

Regulation of Histone Crotonylation

Histone crotonylation is regulated by both enzymatic and metabolic mechanisms:

  1. Enzymatic regulation: Class I histone deacetylases (HDACs), particularly HDAC1, HDAC2, and HDAC3, have been identified as major histone decrotonylases (HDCRs) in mammalian cells. Knock-down of these enzymes results in elevated histone crotonylation .

  2. Metabolic regulation: Histone crotonylation can be regulated through pathways that influence cellular concentrations of crotonyl-CoA, which serves as the donor for the crotonyl group. The addition of crotonate to cells can dramatically increase both cellular concentration of crotonyl-CoA and histone crotonylation in a dose-dependent manner .

Functional Significance

Histone crotonylation has been associated with active chromatin and functions as a positive regulator of transcription. Studies have shown that p300-catalyzed histone crotonylation stimulates transcription more potently than acetylation . In inflammatory responses, increased levels of histone crotonylation at the promoters of inflammatory genes correlate with higher expression of these target genes .

Buffer Composition

The antibody is supplied in a buffer containing:

  • Preservative: 0.03% Proclin 300

  • Constituents: 50% Glycerol, 0.01M PBS, pH 7.4

Stability and Shelf Life

While specific shelf life information is not provided in the search results, proper storage at recommended temperatures will help maintain the antibody's performance over time.

Research Applications in Histone Crotonylation Studies

The Crotonyl-HIST1H2AG (K36) Antibody is a valuable tool in the broader context of histone crotonylation research, which has revealed several important biological functions.

Specificity of Crotonylation Antibodies

Research has shown that well-designed crotonylation antibodies exhibit high specificity. For example, studies with pan anti-Kcr antibodies have demonstrated that they specifically recognize crotonylated lysines but not unmodified, acetylated, propionylated, or butyrylated lysines . This specificity is essential for accurate detection and analysis of histone crotonylation marks, including the K36 site on Histone H2A type 1.

Experimental Manipulation of Crotonylation

Researchers can experimentally increase histone crotonylation by treating cells with crotonate, which serves as a precursor for crotonyl-CoA, the donor for the crotonyl group. This approach has been demonstrated to dramatically increase histone crotonylation signals in HeLa cells . Such experimental manipulations can be valuable for validating the specificity and sensitivity of the Crotonyl-HIST1H2AG (K36) Antibody in various research contexts.

Applications in Epigenetic Research

The Crotonyl-HIST1H2AG (K36) Antibody can be utilized in various epigenetic research applications:

  1. Studying the role of K36 crotonylation on Histone H2A in gene regulation

  2. Investigating changes in this modification during cellular processes like differentiation or stress responses

  3. Examining the relationship between K36 crotonylation and other histone modifications

  4. Exploring the enzymes responsible for adding or removing this specific modification

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery time information.
Synonyms
Histone H2A type 1 (H2A.1) (Histone H2A/ptl), HIST1H2AG, HIST1H2AI, HIST1H2AK, HIST1H2AL, HIST1H2AM, H2AFP, H2AFC, H2AFD, H2AFI, H2AFN
Target Names
HIST1H2AG
Uniprot No.

Target Background

Function
Histone H2A is a core component of the nucleosome, which serves to package and compact DNA into chromatin. This compaction limits the accessibility of DNA to cellular machinery that requires DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated by a complex interplay of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Database Links

HGNC: 4737

OMIM: 602787

KEGG: hsa:8329

STRING: 9606.ENSP00000352627

UniGene: Hs.134999

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Crotonyl-HIST1H2AG (K36) Antibody and what cellular processes does it help investigate?

The Crotonyl-HIST1H2AG (K36) Antibody is a polyclonal antibody raised in rabbits that specifically recognizes the crotonylation modification at lysine 36 of histone H2A type 1 protein (HIST1H2AG). This antibody enables researchers to detect and study a specific post-translational modification involved in epigenetic regulation of gene expression . Histone crotonylation represents an important epigenetic mark that differs from acetylation by its more substantial four-carbon chain modification, potentially creating distinct structural changes in chromatin architecture. The antibody serves as a critical tool for investigating nucleosome dynamics, as histones play central roles in transcription regulation, DNA repair, DNA replication, and chromosomal stability . By enabling specific detection of this modification, researchers can examine how crotonylation at this particular residue influences chromatin accessibility and subsequent cellular processes.

How does HIST1H2AG function within the nucleosome, and why is K36 crotonylation significant?

HIST1H2AG is a core component of the nucleosome, the fundamental repeating unit of chromatin. As part of histone H2A, it participates in wrapping and compacting DNA into chromatin, which limits DNA accessibility to cellular machineries requiring DNA as a template . The nucleosome consists of approximately 146 base pairs of DNA wrapped around an octamer of core histones (two each of H2A, H2B, H3, and H4). Within this structure, K36 (lysine 36) of HIST1H2AG is positioned in a region that can influence nucleosome stability and DNA-histone interactions when modified.

Crotonylation at K36 is significant because it occurs at a strategic position that may affect nucleosome dynamics differently than other modifications. This position-specific crotonylation can serve as a distinctive epigenetic mark that recruits specific reader proteins, potentially creating a unique chromatin state that influences transcriptional activity, DNA repair processes, or other nuclear events. The bulkier crotonyl group (compared to acetyl or methyl groups) likely creates more substantial alterations in chromatin structure, potentially leading to more pronounced effects on gene regulation.

How does crotonylation at K36 of HIST1H2AG differ from other histone modifications?

Crotonylation at K36 of HIST1H2AG represents a distinct post-translational modification that differs from more commonly studied modifications such as acetylation, methylation, and phosphorylation in several key aspects:

Modification TypeChemical StructureCharge EffectKnown WritersKnown ReadersBiological Context
Crotonylation (K36)4-carbon acyl chain with α,β-unsaturationNeutralizes positive chargep300/CBP, CBP/p300YEATS domain proteinsTranscriptional activation, cellular stress response
Acetylation2-carbon acyl groupNeutralizes positive chargeHATs (e.g., p300, GCN5)Bromodomain proteinsTranscriptional activation
MethylationAddition of 1-3 methyl groupsMaintains positive chargeHMTs (e.g., SET-domain proteins)Chromodomain, PHD finger proteinsContext-dependent (activation or repression)
PhosphorylationPhosphate groupAdds negative chargeKinases (e.g., Aurora B)14-3-3 proteins, BRCT domainsCell cycle progression, DNA damage response

The distinctive chemical properties of the crotonyl group—particularly its extended carbon chain and unsaturation—likely create unique recognition surfaces for specific reader proteins, potentially establishing signaling pathways distinct from those activated by other modifications. The crotonylation at K36 may also create different steric effects on nucleosome structure compared to smaller modifications like acetylation or methylation, potentially influencing chromatin compaction and DNA accessibility in unique ways .

What are the optimal protocols for using Crotonyl-HIST1H2AG (K36) Antibody in immunocytochemistry experiments?

Based on validated protocols, the following methodological approach is recommended for immunocytochemistry (ICC) using Crotonyl-HIST1H2AG (K36) Antibody:

Sample Preparation:

  • Culture cells on appropriate coverslips or chamber slides.

  • To enhance crotonylation signal, treat cells with 30mM sodium crotonylate for 4 hours prior to fixation .

  • Fix cells using 4% formaldehyde for 15 minutes at room temperature.

  • Permeabilize cell membranes using 0.2% Triton X-100 for 10 minutes .

Blocking and Antibody Incubation:

  • Block with 10% normal goat serum for 30 minutes at room temperature to reduce non-specific binding .

  • Dilute primary Crotonyl-HIST1H2AG (K36) Antibody in 1% BSA solution at a ratio of 1:50 to 1:100, with 1:50 being optimal for most applications .

  • Incubate with the primary antibody overnight at 4°C in a humidified chamber.

  • Wash 3 times with PBS, 5 minutes each.

  • Incubate with biotinylated secondary antibody for 1 hour at room temperature.

  • Visualize using an HRP-conjugated SP system or fluorescently-labeled secondary antibodies .

Signal Development and Imaging:

  • For chromogenic detection, develop signal using DAB substrate.

  • For fluorescent detection, use appropriate fluorophore-conjugated secondary antibodies.

  • Counterstain nuclei with DAPI.

  • Mount slides using anti-fade mounting medium.

  • Analyze using confocal or fluorescence microscopy.

This protocol typically yields optimal signal-to-noise ratio while preserving cellular morphology and ensuring specific detection of K36 crotonylation in HIST1H2AG.

How can researchers validate the specificity of Crotonyl-HIST1H2AG (K36) Antibody results?

Validating antibody specificity is critical for ensuring reliable experimental outcomes. For Crotonyl-HIST1H2AG (K36) Antibody, a multi-tiered validation approach is recommended:

Peptide Competition Assay:

  • Pre-incubate the antibody with excess (50-100 fold) of the immunizing peptide (synthetic peptide derived from Histone H2A type 1 protein, amino acids 27-40, with K36 crotonylated) .

  • In parallel, use the antibody without peptide competition.

  • Apply both preparations to identical samples.

  • Specific signal should be significantly reduced or eliminated in the peptide-competed samples.

Knockout/Knockdown Controls:

  • Generate HIST1H2AG knockdown or knockout cells using RNAi or CRISPR/Cas9 technology.

  • Compare antibody staining between wild-type and knockdown/knockout cells.

  • Specific signal should be reduced in proportion to the knockdown efficiency or absent in knockout cells.

Site-Directed Mutagenesis:

  • Express wild-type HIST1H2AG and K36R mutant (prevents lysine modification) constructs in cells.

  • Compare antibody staining between wild-type and mutant-expressing cells.

  • Specific signal should be absent or significantly reduced in the K36R mutant-expressing cells.

Enzyme Treatment Controls:

  • Treat fixed cells or cell extracts with histone deacetylases (HDACs) that can remove crotonyl groups.

  • Compare antibody staining before and after enzyme treatment.

  • Specific signal should be reduced after enzyme treatment.

Cross-Reactivity Testing:

  • Perform dot blot analysis with various modified peptides (acetylated, methylated, crotonylated at different positions).

  • The antibody should show strongest reactivity with the K36-crotonylated peptide.

Implementing these validation steps helps ensure that experimental observations genuinely reflect K36 crotonylation of HIST1H2AG rather than nonspecific binding or cross-reactivity with other modifications.

What are the recommended sample preparation techniques for ELISA applications using this antibody?

For optimal ELISA performance with Crotonyl-HIST1H2AG (K36) Antibody, the following sample preparation protocol is recommended:

Nuclear Extract Preparation:

  • Harvest cells by trypsinization or scraping.

  • Wash cells twice with ice-cold PBS.

  • Resuspend cell pellet in hypotonic buffer (10 mM HEPES pH 7.9, 1.5 mM MgCl₂, 10 mM KCl, 0.5 mM DTT, protease inhibitors).

  • Incubate on ice for 15 minutes to allow cell swelling.

  • Add NP-40 to 0.6% final concentration and vortex vigorously for 10 seconds.

  • Centrifuge at 10,000 × g for 30 seconds to pellet nuclei.

  • Extract nuclear proteins with high-salt buffer (20 mM HEPES pH 7.9, 25% glycerol, 420 mM NaCl, 1.5 mM MgCl₂, 0.2 mM EDTA, 0.5 mM DTT, protease inhibitors).

  • Centrifuge at 20,000 × g for 15 minutes to remove debris.

  • Measure protein concentration using Bradford or BCA assay.

Histone Extraction:

  • For more purified histone preparations, use acid extraction:

    • Resuspend nuclear pellet in 0.4 N H₂SO₄.

    • Incubate with rotation for 2 hours at 4°C.

    • Centrifuge at 16,000 × g for 10 minutes.

    • Transfer supernatant to a new tube.

    • Add TCA to 33% final concentration.

    • Incubate on ice for 30 minutes.

    • Centrifuge at 16,000 × g for 10 minutes.

    • Wash pellet with acetone containing 0.1% HCl.

    • Wash pellet with acetone.

    • Air-dry pellet and dissolve in water.

ELISA Protocol Considerations:

  • Coat ELISA plates with histone extracts or nuclear extracts (1-10 μg/well).

  • For sandwich ELISA, coat with anti-histone H2A antibody (1:1000).

  • Block with 3-5% BSA in PBST (PBS + 0.1% Tween-20).

  • Apply Crotonyl-HIST1H2AG (K36) Antibody at 1:1000 to 1:5000 dilution .

  • Use HRP-conjugated secondary antibody.

  • Develop with TMB substrate and measure absorbance at 450 nm.

Including appropriate histone deacetylase inhibitors (HDACi) and histone acyltransferase inhibitors during cell lysis and extraction is critical to preserve the crotonylation state of histones. A cocktail containing sodium butyrate (5-10 mM), nicotinamide (5-10 mM), and trichostatin A (1 μM) is often effective.

How can Crotonyl-HIST1H2AG (K36) Antibody be integrated into ChIP-seq experiments to study genome-wide crotonylation patterns?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using Crotonyl-HIST1H2AG (K36) Antibody provides valuable insights into the genome-wide distribution of K36 crotonylation. The following optimized protocol ensures high-quality ChIP-seq data:

ChIP-seq Protocol:

  • Crosslinking and Chromatin Preparation:

    • Crosslink cells with 1% formaldehyde for 10 minutes at room temperature.

    • Quench with 125 mM glycine for 5 minutes.

    • Wash cells twice with ice-cold PBS.

    • Lyse cells in SDS lysis buffer (1% SDS, 10 mM EDTA, 50 mM Tris-HCl pH 8.0, protease inhibitors).

    • Sonicate chromatin to 200-500 bp fragments (optimize sonication conditions for each cell type).

    • Centrifuge at 14,000 × g for 10 minutes to remove debris.

  • Immunoprecipitation:

    • Dilute chromatin 10-fold in ChIP dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris-HCl pH 8.0, 167 mM NaCl).

    • Pre-clear with protein A/G beads for 1 hour at 4°C.

    • Add Crotonyl-HIST1H2AG (K36) Antibody at 1:20 to 1:100 dilution (typically 3-5 μg per reaction) .

    • Incubate overnight at 4°C with rotation.

    • Add protein A/G beads and incubate for 2 hours at 4°C.

    • Wash sequentially with low-salt, high-salt, LiCl, and TE buffers.

    • Elute DNA-protein complexes with elution buffer (1% SDS, 0.1 M NaHCO₃).

  • Reverse Crosslinking and DNA Purification:

    • Add 5 M NaCl to a final concentration of 200 mM.

    • Incubate at 65°C overnight to reverse crosslinks.

    • Add proteinase K and incubate at 45°C for 1 hour.

    • Purify DNA using phenol-chloroform extraction or commercial kits.

  • Library Preparation and Sequencing:

    • Prepare sequencing libraries following standard protocols.

    • Sequence to a depth of at least 20 million reads per sample.

Data Analysis Considerations:

  • Align reads to reference genome using Bowtie2 or BWA.

  • Call peaks using MACS2 with appropriate parameters for histone modifications (--broad flag).

  • Perform differential binding analysis between conditions using DiffBind or DESeq2.

  • Analyze genomic distribution of peaks relative to gene features (promoters, gene bodies, enhancers).

  • Integrate with RNA-seq data to correlate crotonylation with gene expression patterns.

  • Compare with other histone modification ChIP-seq datasets to identify unique and overlapping patterns.

This approach enables comprehensive mapping of K36 crotonylation across the genome and provides insights into its functional implications in chromatin regulation and transcriptional control.

What experimental approaches can reveal the relationship between metabolic state and HIST1H2AG K36 crotonylation levels?

Crotonylation relies on crotonyl-CoA, a metabolic intermediate, creating an intimate link between cellular metabolism and this epigenetic modification. The following experimental strategies can illuminate this relationship:

Metabolic Manipulation Approaches:

  • Crotonyl-CoA Modulation:

    • Treat cells with sodium crotonate (1-30 mM) to increase intracellular crotonyl-CoA levels .

    • Monitor changes in K36 crotonylation using the Crotonyl-HIST1H2AG (K36) Antibody by Western blot or immunofluorescence.

    • Quantify global changes using mass spectrometry-based proteomics.

  • Fatty Acid Oxidation Manipulation:

    • Inhibit β-oxidation using etomoxir (50-200 μM) or promote it using PPARα agonists.

    • Measure changes in K36 crotonylation and correlate with metabolomic changes.

  • Carbon Source Switching:

    • Culture cells in media with different carbon sources (glucose, glutamine, fatty acids).

    • Quantify K36 crotonylation levels under each condition.

    • Perform metabolomic profiling to correlate specific metabolite levels with crotonylation changes.

Analytical Methods for Correlation Studies:

  • Integrated Multi-Omics:

    • Combine ChIP-seq using Crotonyl-HIST1H2AG (K36) Antibody with metabolomics and transcriptomics.

    • Apply pathway enrichment analysis to identify metabolic pathways correlating with crotonylation changes.

    • Use computational modeling to predict metabolic states that influence K36 crotonylation.

  • In Vitro Enzymatic Assays:

    • Reconstitute crotonylation reactions using purified histones, crotonyl-CoA, and candidate crotonyl-transferases.

    • Test the effects of varying crotonyl-CoA concentrations on reaction kinetics.

    • Use mass spectrometry to confirm site-specific modification.

Experimental Design for Metabolic Studies:

Experimental ConditionMetabolic ManipulationAnalysis MethodExpected Outcome
Hypoxia (1% O₂) vs. NormoxiaShifts metabolism toward glycolysisIF, WB, ChIP-seqAltered K36 crotonylation pattern
Glucose deprivationForces utilization of alternative carbon sourcesIF, WB, metabolomicsChanges in K36 crotonylation correlating with specific metabolites
Fatty acid supplementationIncreases β-oxidationChIP-seq, RNA-seq, metabolomicsRedistribution of K36 crotonylation and altered gene expression
Sirtuin inhibition (nicotinamide)Blocks deacylation enzymesIF, WB, MS proteomicsIncreased K36 crotonylation levels

These approaches provide a framework for understanding how cellular metabolism influences the epigenetic landscape through modulation of histone crotonylation, potentially revealing novel mechanisms of metabolic signaling to chromatin.

How can mass spectrometry complement antibody-based detection of Crotonyl-HIST1H2AG (K36)?

Mass spectrometry (MS) offers orthogonal validation and quantification of histone crotonylation that complements antibody-based approaches. An integrated workflow combining Crotonyl-HIST1H2AG (K36) Antibody techniques with MS provides comprehensive characterization:

MS-Based Workflow for Crotonylation Analysis:

  • Sample Preparation:

    • Extract histones using acid extraction (as described in section 2.3).

    • Perform chemical derivatization to block unmodified lysines (typically propionylation).

    • Digest with trypsin or other proteases.

    • Enrich for crotonylated peptides using:
      a) Antibody-based enrichment using Crotonyl-HIST1H2AG (K36) Antibody
      b) HILIC fractionation
      c) Immobilized metal affinity chromatography (IMAC)

  • LC-MS/MS Analysis:

    • Use high-resolution MS (e.g., Orbitrap) with HCD or ETD fragmentation.

    • Include MS/MS pattern validation for crotonylation:

      • Diagnostic fragment at m/z 70.0651 for crotonyllysine immonium ion

      • Neutral loss of 70.0419 Da (C₄H₆O)

    • Monitor K36-containing peptides from HIST1H2AG.

  • Targeted Quantification Strategies:

    • Develop PRM (Parallel Reaction Monitoring) or MRM (Multiple Reaction Monitoring) assays for K36 crotonylated peptides.

    • Use SILAC, TMT, or label-free quantification for relative abundance measurement.

    • Generate synthetic crotonylated peptide standards for absolute quantification.

Integrated Approach with Antibody-Based Methods:

  • Validation Strategy:

    • Use MS to validate antibody specificity by analyzing immunoprecipitated material.

    • Compare ChIP-seq peaks with MS-identified crotonylation sites.

    • Develop targeted MS assays for sites identified by antibody-based methods.

  • Complementary Strengths:

TechniqueStrengthsLimitationsComplementarity
Crotonyl-HIST1H2AG (K36) Antibody- Cellular localization
- Genomic distribution
- High sensitivity
- Compatible with fixed samples
- Potential cross-reactivity
- Semi-quantitative
- Limited multiplex capability
Provides spatial information and genomic context
Mass Spectrometry- Site-specific identification
- Quantitative
- Multiplex analysis of modifications
- Unbiased discovery
- Limited spatial information
- Requires more material
- Lower throughput for imaging
Provides definitive chemical identification and quantification
  • Workflow Integration:

    • Use antibody for initial screening and localization studies.

    • Confirm key findings with MS-based validation.

    • Apply MS for detailed characterization of modification stoichiometry and combinatorial patterns.

    • Leverage ChIP-MS approaches to combine genomic localization with modification identification.

This integrated approach maximizes the strengths of both techniques, providing more comprehensive and reliable insights into K36 crotonylation biology than either method alone.

What are the most common technical challenges when working with Crotonyl-HIST1H2AG (K36) Antibody and how can they be addressed?

Researchers commonly encounter several technical challenges when working with Crotonyl-HIST1H2AG (K36) Antibody. Here are the most frequent issues and their solutions:

1. Weak or Absent Signal:

Potential CauseDiagnostic SignsSolutions
Low modification abundancePositive controls also show weak signal- Pre-treat cells with sodium crotonate (30mM, 4h)
- Inhibit HDAC activity with nicotinamide/TSA
- Optimize fixation to preserve modification
Antibody degradationDecreased performance over time- Aliquot antibody upon receipt
- Store at -80°C
- Avoid repeated freeze-thaw cycles
Insufficient permeabilizationPoor nuclear staining- Optimize Triton X-100 concentration (0.2-0.5%)
- Extend permeabilization time
- Try alternative detergents (e.g., NP-40)
Suboptimal antibody concentrationSignal-to-noise ratio is poor- Titrate antibody (1:10 to 1:100 for ICC)
- Increase primary antibody incubation time
- Optimize secondary antibody concentration

2. High Background or Non-specific Staining:

Potential CauseDiagnostic SignsSolutions
Insufficient blockingDiffuse cytoplasmic staining- Increase blocking time (1-2 hours)
- Try alternative blocking agents (BSA, serum, milk)
- Include 0.1-0.3% Triton X-100 in blocking buffer
Cross-reactivitySignal persists in knockdown controls- Include peptide competition controls
- Pre-absorb antibody with non-specific epitopes
- Increase washing stringency and duration
OverfixationHigh background, poor morphology- Reduce fixation time
- Try alternative fixatives (e.g., methanol)
- Include antigen retrieval step
Secondary antibody issuesBackground in secondary-only controls- Use highly cross-adsorbed secondary antibodies
- Decrease secondary antibody concentration
- Include 0.05% Tween-20 in antibody diluent

3. Inconsistent Results Between Experiments:

Potential CauseDiagnostic SignsSolutions
Cell culture variabilityResults vary with cell density/passage- Standardize cell culture conditions
- Use cells at consistent confluency
- Control for cell cycle stage
Modification dynamicsTime-dependent variation- Synchronize treatment times
- Consider circadian or cell cycle effects
- Document harvest times precisely
Technical variationHigh standard deviation between replicates- Standardize all protocol steps
- Use automation where possible
- Implement quality control metrics

4. Protocol-Specific Challenges:

ApplicationCommon ChallengeTargeted Solution
ImmunofluorescenceAutofluorescence- Include Sudan Black B treatment
- Use spectral unmixing
- Try anti-fade mounting media with quenchers
ChIPLow IP efficiency- Increase antibody amount (5-10 μg/IP)
- Extend IP incubation time
- Optimize chromatin fragmentation
ELISAInconsistent coating- Use purified histones rather than extracts
- Standardize coating time/temperature
- Include capture antibody for sandwich ELISA
Western blotMultiple bands- Use gradient gels for better resolution
- Optimize transfer conditions for histones
- Include appropriate molecular weight markers

Implementing these targeted solutions can substantially improve experimental outcomes and data reliability when working with Crotonyl-HIST1H2AG (K36) Antibody.

How should researchers interpret conflicting data between different detection methods for K36 crotonylation?

When faced with discrepancies between different detection methods for K36 crotonylation, a systematic approach to data reconciliation is essential:

Framework for Resolving Conflicting Data:

  • Methodological Considerations:

    Detection MethodInherent LimitationsPotential False PositivesPotential False Negatives
    Antibody-based (IF/IHC)Epitope accessibility issuesCross-reactivity with similar modificationsLow-abundance modifications below detection limit
    Western blotLimited resolution of similar molecular weight proteinsBand overlap, non-specific bindingTransfer inefficiency for histones
    ChIP-seqAntibody specificity in chromatin contextNon-specific DNA binding during IPRegions of low chromatin accessibility
    Mass spectrometryIonization efficiency biasesIsobaric modificationsIon suppression, incomplete fragmentation
  • Systematic Validation Approach:

    • Perform reciprocal validation using orthogonal techniques.

    • Validate with genetic approaches (K36R mutation, CRISPR/Cas9).

    • Include appropriate positive and negative controls in all experiments.

    • Use dose-response experiments (e.g., crotonate titration) to assess correlation between methods.

  • Data Integration Strategies:

    • Weigh evidence based on methodological strengths.

    • Consider quantitative versus qualitative discrepancies.

    • Examine context dependency (cell type, treatment conditions).

    • Apply statistical methods for data integration (meta-analysis approaches).

Case-Based Reconciliation Examples:

Case 1: ChIP-seq vs. MS Discrepancy

  • Observation: ChIP-seq with Crotonyl-HIST1H2AG (K36) Antibody shows enrichment at specific loci, but MS fails to detect K36 crotonylation.

  • Potential causes:

    • Low abundance below MS detection limit

    • Antibody cross-reactivity with another modification

    • Region-specific effects influencing ChIP efficiency

  • Resolution approach:

    • Perform ChIP-MS on the immunoprecipitated material

    • Increase MS sensitivity through targeted approaches

    • Validate with site-specific K36R mutation

    • Use competition assays with differentially modified peptides

Case 2: Western Blot vs. Immunofluorescence Discrepancy

  • Observation: Western blot shows increase in K36 crotonylation after treatment, but IF shows no change.

  • Potential causes:

    • Epitope masking in cellular context

    • Different fixation effects on epitope accessibility

    • Subcellular redistribution rather than abundance change

    • Differences in antibody performance in native vs. denatured conditions

  • Resolution approach:

    • Try different fixation and permeabilization methods

    • Perform subcellular fractionation followed by Western blot

    • Quantify IF signal across different cellular compartments

    • Test antibody on dot blots with native and denatured histones

Decision Tree for Conflicting Data Resolution:

  • Verify technical quality of each experiment independently

  • Evaluate whether discrepancies are quantitative or qualitative

  • Perform additional controls focused on the specific discrepancy

  • Apply alternative methods that circumvent limitations of original techniques

  • Consider biological explanations (context-dependency, dynamic regulation)

  • Revise hypotheses to accommodate consistent observations across methods

What statistical approaches are most appropriate for quantifying changes in HIST1H2AG K36 crotonylation across experimental conditions?

Statistical Frameworks by Experimental Approach:

  • Immunofluorescence/Immunohistochemistry Quantification:

    • Recommended Methods:

      • Normalized fluorescence intensity measurements

      • Cell-by-cell analysis using automated image processing

      • Thresholding approaches for positive/negative classification

    • Statistical Tests:

      • For normally distributed data: t-test (2 conditions) or ANOVA (multiple conditions)

      • For non-parametric data: Mann-Whitney U (2 conditions) or Kruskal-Wallis (multiple conditions)

      • For proportional data (% positive cells): Chi-square or Fisher's exact test

    • Advanced Approaches:

      • Mixed-effects models for experiments with multiple fields/replicates

      • Spatial statistics for analyzing modification distribution patterns

  • Western Blot Quantification:

    • Recommended Methods:

      • Normalization to total H2A or loading controls (e.g., total protein)

      • Multiple technical and biological replicates (minimum n=3)

      • Standard curve inclusion for absolute quantification

    • Statistical Tests:

      • Paired t-tests for before/after comparisons

      • Repeated measures ANOVA for time-course experiments

      • ANCOVA when controlling for covariates

    • Robustness Measures:

      • Bootstrap analysis for confidence interval estimation

      • Non-parametric tests when normality cannot be assumed

  • ChIP-seq Data Analysis:

    • Recommended Methods:

      • Peak calling using MACS2 with appropriate controls

      • Differential binding analysis (DiffBind, HOMER, DESeq2)

      • Normalization to input or spike-in controls

    • Statistical Tests:

      • Negative binomial models for count data

      • False discovery rate correction for multiple testing

      • Permutation tests for peak distributions

    • Advanced Approaches:

      • Hidden Markov Models for chromatin state analysis

      • Bayesian approaches for integrating multiple datasets

  • Mass Spectrometry Quantification:

    • Recommended Methods:

      • Label-free quantification with appropriate normalization

      • Stable isotope labeling for direct comparisons (SILAC, TMT)

      • Extracted ion chromatograms for targeted quantification

    • Statistical Tests:

      • Moderated t-tests with empirical Bayes approaches

      • Linear models for complex experimental designs

      • Peptide-level vs. protein-level statistical integration

    • Advanced Approaches:

      • Probabilistic models for site localization

      • Bayesian inference for modification stoichiometry

Statistical Analysis Workflow:

  • Data Quality Assessment:

    • Evaluate normality using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • Assess homogeneity of variance using Levene's test

    • Identify outliers using Grubb's test or boxplot methods

    • Calculate coefficient of variation for technical replicates

  • Effect Size Calculation:

    • Cohen's d for continuous measurements

    • Odds ratios for categorical outcomes

    • Fold change with confidence intervals

    • Area under ROC curve for classification performance

  • Power Analysis and Sample Size Determination:

    • A priori power analysis based on pilot data

    • Post hoc power calculation for negative results

    • Sample size recommendations:

      • Western blot/IF: Minimum n=3 biological replicates

      • ChIP-seq: Minimum n=2-3 biological replicates

      • MS: Minimum n=3-4 biological replicates

  • Reporting Standards:

    • Always include both biological and technical replicate numbers

    • Report exact p-values rather than thresholds

    • Include appropriate visualizations (box plots, violin plots)

    • Provide measures of dispersion (standard deviation, standard error)

Applying these statistical approaches ensures robust quantification of K36 crotonylation changes and facilitates meaningful interpretation of experimental results across different conditions.

What is the current understanding of how HIST1H2AG K36 crotonylation affects transcriptional regulation?

Current research indicates that K36 crotonylation of HIST1H2AG represents a functionally distinct epigenetic mark with specific roles in transcriptional regulation. The emerging understanding can be summarized as follows:

Molecular Mechanisms of Transcriptional Influence:

Crotonylation at K36 of HIST1H2AG appears to influence transcription through several interconnected mechanisms:

  • Chromatin Structure Modulation:

    • The bulky crotonyl group likely causes greater disruption to histone-DNA interactions than acetylation, potentially creating more accessible chromatin regions.

    • Located within the globular domain of H2A, K36 crotonylation may affect nucleosome stability and dynamics, influencing higher-order chromatin structure.

    • This modification may alter nucleosome positioning or stability, particularly at regulatory elements.

  • Reader Protein Recruitment:

    • Specific reader proteins containing YEATS domains show preferential binding to crotonylated lysines compared to acetylated lysines.

    • These readers may recruit transcriptional coactivators or components of the transcriptional machinery.

    • The differential recognition of crotonylation versus other acylations provides a mechanism for specific gene regulation programs.

  • Transcriptional Complex Formation:

    • K36 crotonylation may facilitate the assembly or stability of transcription factor complexes at promoters and enhancers.

    • The modification may also influence RNA polymerase II recruitment or processivity.

    • Cross-talk with other histone modifications likely creates combinatorial effects on transcriptional output.

Genomic Distribution and Associated Gene Programs:

ChIP-seq studies with antibodies recognizing histone crotonylation have revealed several patterns:

  • Enrichment Patterns:

    • Crotonylated histones are generally enriched at promoters and enhancers of active genes.

    • Particularly strong enrichment is often observed at transcription start sites (TSS).

    • Tissue-specific enhancers show distinct crotonylation patterns correlating with gene activity.

  • Functional Gene Categories:

    • Genes involved in cellular stress responses often show regulated crotonylation.

    • Developmental gene programs may utilize crotonylation for temporal regulation.

    • Metabolic genes show crotonylation patterns that respond to cellular nutrient status.

Regulatory Dynamics and Context-Dependency:

  • Temporal Regulation:

    • K36 crotonylation appears highly dynamic, with rapid changes in response to cellular signals.

    • The modification may serve as an intermediate step in transcriptional memory mechanisms.

    • Cell cycle-dependent fluctuations in crotonylation have been observed at specific loci.

  • Cell Type Specificity:

    • Distinct crotonylation landscapes exist across different cell types.

    • Tissue-specific regulatory elements show corresponding patterns of crotonylation.

    • Developmental transitions involve programmed changes in crotonylation patterns.

Future research using Crotonyl-HIST1H2AG (K36) Antibody will continue to refine this understanding, particularly through integrative genomic approaches that correlate K36 crotonylation with transcriptional output and other epigenetic features.

How does HIST1H2AG K36 crotonylation interact with the DNA damage response and genome stability?

Emerging evidence suggests that histone crotonylation, including K36 crotonylation of HIST1H2AG, may play significant roles in DNA damage response (DDR) and genome maintenance. This relationship represents an exciting frontier in epigenetic research:

Involvement in DNA Damage Response Mechanisms:

  • Chromatin Accessibility Regulation:

    • K36 crotonylation may facilitate chromatin relaxation at damage sites, providing access to repair machinery.

    • The dynamic regulation of this modification could contribute to the orchestrated chromatin changes during different phases of DNA repair.

    • Studies with Crotonyl-HIST1H2AG (K36) Antibody have shown redistribution of this mark following genotoxic stress .

  • Repair Pathway Specificity:

    • Preliminary evidence suggests differential patterns of K36 crotonylation depending on the type of DNA damage (DSBs, SSBs, UV lesions).

    • The modification may influence pathway choice between homologous recombination (HR) and non-homologous end joining (NHEJ).

    • Temporal dynamics of K36 crotonylation during repair progression may serve as a regulatory mechanism.

  • Interaction with DDR Signaling:

    • Crosstalk with DNA damage-induced phosphorylation cascades (ATM/ATR pathways).

    • Potential regulation by DDR-responsive histone modifying enzymes.

    • Possible role in checkpoint activation or recovery.

Experimental Evidence and Research Approaches:

Research ApproachKey FindingsImplications
Laser microirradiation with IFK36 crotonylation shows temporal changes at damage sitesSuggests active regulation during repair
ChIP-seq after DNA damageRedistribution of K36 crotonylation genome-wideMay indicate global chromatin response to damage
Genetic studies (K36R mutants)Altered sensitivity to DNA damaging agentsFunctional requirement in damage tolerance
Enzyme inhibition studiesCrotonylation writers/erasers influence repair efficiencyPotential therapeutic targets

Proposed Mechanistic Models:

  • "Access-Repair-Restore" Model:

    • K36 crotonylation increases following damage, promoting chromatin accessibility (Access).

    • Repair machinery operates in the more accessible chromatin environment.

    • K36 crotonylation is removed during later stages of repair to restore original chromatin state (Restore).

  • "Modification Reader Recruitment" Model:

    • K36 crotonylation serves as a docking site for specific reader proteins.

    • These readers recruit or stabilize DNA repair factors at damage sites.

    • The specificity of crotonylation readers vs. acetylation readers provides pathway selectivity.

  • "Transcriptional Regulation" Model:

    • K36 crotonylation regulates expression of DNA repair genes.

    • This creates a transcriptional program supporting genomic stability.

    • Metabolic state influences repair capacity through crotonylation-dependent gene expression.

Future Research Directions:

  • Temporal profiling of K36 crotonylation during different stages of DNA repair

  • Identification of readers that specifically recognize K36 crotonylation in damage contexts

  • Investigation of crotonylation-acetylation balance in repair pathway choice

  • Development of tools to manipulate K36 crotonylation with spatial and temporal precision

This emerging field represents a promising area for applications of Crotonyl-HIST1H2AG (K36) Antibody in understanding the epigenetic dimensions of genome stability.

What emerging technologies will enhance detection and functional analysis of HIST1H2AG K36 crotonylation?

The field of histone modification research is rapidly evolving, with several innovative technologies poised to transform our understanding of HIST1H2AG K36 crotonylation. These emerging approaches promise to overcome current limitations and provide unprecedented insights:

Next-Generation Antibody Technologies:

  • Recombinant Antibody Engineering:

    • Single-chain variable fragments (scFvs) with enhanced specificity for K36 crotonylation

    • Nanobodies derived from camelid antibodies offering superior epitope access in compact chromatin

    • Bispecific antibodies recognizing K36 crotonylation in combination with other modifications

    • Benefits: Reduced batch-to-batch variation, improved specificity, smaller size for better chromatin penetration

  • Proximity Ligation Assays (PLA):

    • Detection of K36 crotonylation in proximity to specific protein factors or other modifications

    • Single-molecule resolution in intact cells

    • Multiplexed detection of modification combinations

    • Benefits: Contextual information, enhanced sensitivity, detection of modification crosstalk

Advanced Imaging Approaches:

  • Super-Resolution Microscopy:

    • STORM/PALM imaging of K36 crotonylation distribution at nanoscale resolution

    • Live-cell imaging using modification-specific nanobody-fluorophore conjugates

    • Correlative light-electron microscopy linking modification to chromatin ultrastructure

    • Benefits: Spatial distribution at sub-diffraction resolution, dynamic tracking in living cells

  • Mass Spectrometry Imaging:

    • MALDI-imaging of tissue sections for K36 crotonylation distribution

    • NanoSIMS technology for elemental and isotopic mapping

    • Imaging mass cytometry for single-cell histone modification landscapes

    • Benefits: In situ analysis without antibody dependence, multiplexed detection, tissue context preservation

Genomic and Systems Biology Approaches:

  • CUT&Tag and CUT&RUN:

    • Antibody-directed cleavage methods offering improved signal-to-noise over ChIP-seq

    • Single-cell adaptations revealing cell-to-cell heterogeneity in K36 crotonylation

    • Combinatorial CUT&Tag for simultaneous mapping of multiple modifications

    • Benefits: Lower input requirements, reduced background, improved resolution

  • Integrative Multi-Omics:

    • Integration of K36 crotonylation maps with transcriptomics, metabolomics, and proteomics

    • Network modeling of crotonylation-dependent regulatory circuits

    • Machine learning approaches to predict functional outcomes of crotonylation patterns

    • Benefits: Systems-level understanding, predictive modeling, identification of emergent properties

Functional Manipulation Technologies:

  • Targeted Enzymology:

    • CRISPR-dCas9 fusion with crotonyl-transferases or de-crotonylases

    • Chemically-induced proximity systems for temporal control of K36 crotonylation

    • Optogenetic control of site-specific crotonylation

    • Benefits: Causality testing, temporal manipulation, site-specific effects

  • Chemical Biology Approaches:

    • Bumped inhibitors/activators for engineered enzymes (analog-sensitive approach)

    • Activity-based probes for crotonylation writers and erasers

    • Clickable crotonyl-CoA analogs for metabolic labeling

    • Benefits: Chemical genetic control, enzyme activity profiling, metabolic dynamics

Implementation Timeline and Developmental Status:

TechnologyCurrent Development StageExpected AvailabilityTechnical Barriers
Recombinant antibodiesEarly implementation1-2 yearsEpitope accessibility in native conformation
CUT&Tag adaptationsPrototype methodsCurrently availableOptimization for various cell types
Live-cell imagingProof-of-concept2-3 yearsDevelopment of cell-permeable probes
Targeted enzymologyEarly research phase3-5 yearsSpecificity of targeted enzyme activity
Multi-omics integrationAnalytical method development1-2 yearsComputational integration challenges
Single-cell epigenomicsProtocol optimizationCurrently availableSensitivity limitations for rare modifications

These emerging technologies will substantially enhance our ability to detect, quantify, and functionally characterize K36 crotonylation of HIST1H2AG, leading to a more comprehensive understanding of its biological roles and regulatory mechanisms.

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.