Crotonyl-HIST1H2BC (K11) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Crotonyl-HIST1H2BC (K11) Antibody

The Crotonyl-HIST1H2BC (K11) Antibody is a rabbit polyclonal or monoclonal antibody designed to detect lysine crotonylation at position K11 of histone H2B (HIST1H2BC), a core nucleosomal protein. Crotonylation is a post-translational modification (PTM) that influences chromatin structure, transcriptional regulation, and cellular processes such as DNA repair and replication . This antibody is critical for studying the role of K11 crotonylation in epigenetic regulation and disease mechanisms.

Target Protein and Modification

Histone H2B is a core component of nucleosomes, which compact DNA to regulate accessibility. Crotonylation at K11 (H2B-K11cr) is a type of β-hydroxybutyryl (or similar acyl) modification that impacts chromatin dynamics .

PropertyDetails
ImmunogenPeptide sequence around crotonyl-Lys11 of human H2B (e.g., PACO59646)
ReactivityHuman (confirmed in HEK-293, A549, K562 cell lines)
ApplicationsELISA, Western Blot (WB), Immunofluorescence (IF), Peptide Array
IsotypeRabbit IgG (polyclonal or recombinant monoclonal)
Predicted Band Size~14 kDa (observed in NIH/3T3 cell lysates)

Key Techniques

  • Western Blot: Used to detect H2B-K11cr in cell lysates (e.g., HEK-293, A549) .

  • Immunofluorescence: Visualizes subcellular localization of crotonylated histones .

  • Peptide Array: Validates specificity against modified vs. unmodified H2B peptides .

Validation Data

  • Specificity:

    • Exclusively binds crotonylated K11 in peptide arrays .

    • Cross-reactivity with acetylated or other acylated lysines is minimal, though pan-K-acyl antibodies may show partial overlap .

Regulation by HDACs

Studies using chemical proteomics reveal that H2B-K11crotonylation is dynamically regulated by histone deacetylases (HDACs):

RegulatorEffectEvidence
HDAC1/3Acts as a de-crotonylase; knockdown increases H2B-K11cr levels LC-MS/MS quantification in HEK293T cells treated with HDAC inhibitors
Crotonyl-CoAServes as a substrate for crotonyltransferases (e.g., p300) In vitro assays with recombinant chromatin substrates

Biological Roles

  • Transcriptional Regulation: Crotonylation at K11 may influence chromatin accessibility, though specific transcriptional targets remain under investigation .

  • Disease Implications: Crotonylation is linked to cancer and metabolic disorders, though direct evidence for H2B-K11cr in pathogenesis is emerging .

Comparative Analysis of H2B Crotonylation Antibodies

AntibodyTargetApplicationsSource
Crotonyl-HIST1H2BC (K11) AntibodyH2B-K11crotonylWB, ELISA, IFAssay Genie (PACO59646)
Crotonyl-HIST1H2BC (K5) AntibodyH2B-K5crotonylWB, PepArr, ICC/IFAbcam (ab177396)
2-hydroxyisobutyryl-HIST1H2BC (K108)H2B-K108hibWB, ChIPCUSABIO

Challenges and Considerations

  • Cross-Reactivity: Pan-K-crotonyl antibodies may recognize acetylated or butyrylated sites due to structural similarities, necessitating strict validation .

  • Sample Preparation: Crotonylated peptides are labile; enzymatic removal (e.g., via HDACs) or chemical stabilization is required for detection .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
H2BC4 antibody; H2BFL antibody; HIST1H2BC; antibody; H2BC6 antibody; H2BFH antibody; HIST1H2BE; antibody; H2BC7 antibody; H2BFG antibody; HIST1H2BF; antibody; H2BC8 antibody; H2BFA antibody; HIST1H2BG; antibody; H2BC10 antibody; H2BFK antibody; HIST1H2BIHistone H2B type 1-C/E/F/G/I antibody; Histone H2B.1 A antibody; Histone H2B.a antibody; H2B/a antibody; Histone H2B.g antibody; H2B/g antibody; Histone H2B.h antibody; H2B/h antibody; Histone H2B.k antibody; H2B/k antibody; Histone H2B.l antibody; H2B/l antibody
Target Names
HIST1H2BC
Uniprot No.

Target Background

Function
A core component of the nucleosome, this antibody targets Crotonyl-HIST1H2BC (K11). Nucleosomes package and compact DNA into chromatin, thus regulating DNA accessibility to cellular machinery requiring DNA as a template. Histones, therefore, play a crucial role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is modulated by a complex array of post-translational histone modifications, often referred to as the histone code, and nucleosome remodeling. Additionally, this protein exhibits broad antibacterial activity. It may contribute to the formation of a functional antimicrobial barrier in the colonic epithelium and to the bactericidal activity of amniotic fluid.
Database Links

HGNC: 4757

OMIM: 602798

KEGG: hsa:3017

STRING: 9606.ENSP00000366962

UniGene: Hs.182137

Protein Families
Histone H2B family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Crotonyl-HIST1H2BC (K11) Antibody and what is its primary research application?

Crotonyl-HIST1H2BC (K11) Antibody is a polyclonal antibody produced in rabbits that specifically recognizes the crotonylation modification at lysine 11 of Histone H2B type 1-C/E/F/G/I. This antibody serves as a critical tool for investigating histone crotonylation, an emerging epigenetic modification with significant implications in gene regulation, chromatin remodeling, and cell differentiation. The primary research applications include Western blotting, immunofluorescence, ELISA, and chromatin immunoprecipitation (ChIP) to detect and analyze crotonylated histone H2B in different cell types and experimental conditions . Histone crotonylation research is particularly relevant in cancer biology, as dysregulation of histone modifications can contribute to oncogenesis and tumor progression, making this antibody valuable for studies in both basic epigenetics and disease-focused research .

How does crotonylation at K11 differ from other histone modifications in functional significance?

Crotonylation at K11 of histone H2B represents a distinct post-translational modification that differs from more common modifications like acetylation and methylation in several important ways. While acetylation generally promotes an open chromatin state and gene activation by neutralizing the positive charge of lysine residues, crotonylation adds a bulkier group with a four-carbon chain that creates more significant structural changes to the histone. These structural changes can affect nucleosome stability and protein-protein interactions in unique ways compared to acetylation. Functionally, K11 crotonylation of HIST1H2BC is associated with transcriptionally active chromatin and has been shown to participate in specialized gene regulation contexts, particularly during cell differentiation and in response to metabolic changes when cellular crotonyl-CoA levels fluctuate . Unlike some histone methylation marks that can be associated with either activation or repression depending on context, K11 crotonylation appears more consistently linked to gene activation, though research continues to refine our understanding of its precise functional roles.

What are the recommended experimental controls when using Crotonyl-HIST1H2BC (K11) Antibody?

Robust experimental design with Crotonyl-HIST1H2BC (K11) Antibody requires several critical controls:

  • Positive Control: Use cell lines or tissues known to express crotonylated H2B at K11, such as actively dividing cells with high metabolic activity.

  • Negative Control: Include samples treated with histone decrotonylase enzymes (e.g., HDAC family members known to remove crotonyl groups) or cells grown in conditions that minimize crotonylation.

  • Peptide Competition Assay: Pre-incubate the antibody with increasing amounts of the immunizing peptide (crotonylated K11 peptide) to confirm signal specificity.

  • Crotonylation Induction Control: Treatment of cells with crotonyl-CoA or crotonate, along with HDAC inhibitors, to artificially increase histone crotonylation levels.

  • Antibody Validation by Mutagenesis: If possible, use cell lines with H2B K11R mutations that prevent crotonylation specifically at this site to verify antibody specificity .

These controls collectively ensure that the observed signals are genuinely representative of K11 crotonylation rather than non-specific binding or cross-reactivity with other histone modifications.

How can Crotonyl-HIST1H2BC (K11) Antibody be optimized for chromatin immunoprecipitation (ChIP) experiments?

Optimizing Crotonyl-HIST1H2BC (K11) Antibody for ChIP experiments requires careful consideration of several technical parameters:

ChIP Protocol Optimization Table:

ParameterStandard ConditionOptimized Condition for K11 CrotonylationRationale
Crosslinking1% formaldehyde, 10 min1% formaldehyde, 5-8 minShorter crosslinking preserves the crotonyl modification
Sonication20-30 cycles15-20 cycles with cooling periodsGentler sonication reduces epitope damage
Antibody Concentration2-5 μg per ChIP5-10 μg per ChIPHigher concentration improves rare modification capture
Incubation TimeOvernight at 4°C48 hours at 4°CExtended incubation enhances binding to less abundant modifications
Washing StringencyStandard RIPA buffersReduced SDS concentration (0.05%)Preserves antibody-antigen interaction
ElutionStandard elution bufferAdd 10 mM sodium butyrateInhibits HDACs during elution

Additionally, pre-treatment of cells with histone deacetylase inhibitors like sodium butyrate or trichostatin A can enhance crotonylation signals. For next-generation sequencing applications following ChIP (ChIP-seq), library preparation should be optimized to account for potentially lower yields compared to more abundant histone modifications. Rigorous quality control via qPCR of known crotonylated regions should be performed before sequencing to confirm successful enrichment .

What are the current challenges in differentiating between crotonylation at K11 versus other lysine residues on the same histone?

Distinguishing crotonylation at K11 from modifications at other lysine residues presents several technical challenges that researchers must address:

First, the structural similarity of the histone H2B tail region creates potential for epitope cross-reactivity. The antibody might recognize similar flanking sequences around other lysine residues, particularly K5, K12, and K15, which share sequence context similarities. This necessitates rigorous validation using synthetic peptides containing crotonylation at different positions.

Second, co-occurrence of modifications complicates detection. Crotonylation at K11 often exists alongside other modifications like acetylation at neighboring residues, which can sterically hinder antibody binding or alter the confirmation of the epitope. Researchers should employ mass spectrometry-based approaches to definitively map modification patterns when absolute specificity is required.

Third, crotonylation levels are generally lower than acetylation, creating signal-to-noise challenges. Enrichment strategies before Western blotting or ChIP can help, such as using recombinant bromodomain-containing proteins that preferentially bind crotonylated histones to pull down these modified histones before antibody detection .

Combined approaches using both antibody-based detection and mass spectrometry provide the most comprehensive assessment of site-specific crotonylation.

What is the relationship between metabolic changes and HIST1H2BC K11 crotonylation levels in experimental systems?

The relationship between cellular metabolism and HIST1H2BC K11 crotonylation represents a critical intersection between epigenetics and metabolism that can be experimentally manipulated:

Crotonylation depends on intracellular levels of crotonyl-CoA, an intermediate in fatty acid metabolism. Experimental manipulation of beta-oxidation pathways through nutrient restriction or supplementation with fatty acids of varying chain lengths directly affects the availability of crotonyl-CoA. For example, supplementing culture media with crotonate (2-butenoate) increases intracellular crotonyl-CoA levels and subsequently enhances histone crotonylation, including at K11 of HIST1H2BC.

The activity of class I histone deacetylases (HDACs), which can remove crotonyl groups, is regulated by metabolic cofactors like NAD+. Therefore, altering NAD+/NADH ratios through manipulations of glycolysis, TCA cycle, or oxidative phosphorylation influences crotonylation dynamics. Experimental interventions using compounds like 2-deoxyglucose (2-DG) to inhibit glycolysis or oligomycin to inhibit ATP synthase create distinct metabolic states that affect K11 crotonylation patterns.

This metabolism-epigenetic link can be systematically studied using targeted metabolomics alongside epigenetic profiling with the K11 antibody, enabling researchers to correlate changes in specific metabolite pools with alterations in the crotonylation landscape across the genome .

What is the optimal protocol for using Crotonyl-HIST1H2BC (K11) Antibody in Western blotting applications?

The following optimized Western blotting protocol has been developed specifically for Crotonyl-HIST1H2BC (K11) Antibody:

  • Sample Preparation:

    • Extract histones using acid extraction (0.2N HCl) rather than standard RIPA buffer to enrich for histones

    • Add 10 mM sodium butyrate to all buffers to inhibit HDAC activity and preserve crotonylation

    • Use fresh samples when possible, as freeze-thaw cycles can reduce detectable crotonylation

  • Gel Electrophoresis:

    • Use 15-18% SDS-PAGE gels to properly resolve the low molecular weight histone proteins

    • Load 10-20 μg of acid-extracted histones per lane

    • Include molecular weight markers capable of resolving proteins in the 10-20 kDa range

  • Transfer and Blocking:

    • Use PVDF membrane (0.2 μm pore size) instead of nitrocellulose for better retention of small proteins

    • Transfer at lower voltage (30V) overnight at 4°C to ensure complete transfer of histones

    • Block with 5% BSA in TBST rather than milk (milk contains HDACs that could remove crotonylation)

  • Antibody Incubation:

    • Dilute Crotonyl-HIST1H2BC (K11) Antibody at 1:500 to 1:1000 for optimal results

    • Incubate overnight at 4°C with gentle agitation

    • Following primary antibody, use anti-rabbit HRP conjugated secondary antibody at 1:5000 dilution

  • Detection:

    • Use enhanced chemiluminescence with longer exposure times (2-5 minutes) than typically needed for abundant proteins

    • For quantitative analysis, include total H2B antibody on a separate blot as loading control

Expected results: The antibody should detect a band at approximately 14 kDa corresponding to crotonylated histone H2B. Signal intensity will vary depending on cell type and metabolic state, with actively transcribing cells typically showing higher levels of crotonylation .

How can immunofluorescence protocols be adapted for optimal detection of K11 crotonylation in different tissue types?

Adapting immunofluorescence (IF) protocols for Crotonyl-HIST1H2BC (K11) detection across tissue types requires specific modifications based on tissue characteristics:

For Fixed Cell Cultures:

  • Fix cells with 4% paraformaldehyde for 10 minutes at room temperature

  • Perform antigen retrieval using 10 mM sodium citrate buffer (pH 6.0) with 0.05% Tween-20 at 95°C for 20 minutes

  • Permeabilize with 0.2% Triton X-100 for 10 minutes

  • Block with 1% BSA and 5% normal goat serum for 1 hour

  • Incubate with Crotonyl-HIST1H2BC (K11) Antibody at 1:50 dilution overnight at 4°C

  • Use Alexa Fluor conjugated secondary antibodies at 1:500 dilution

For Formalin-Fixed Paraffin-Embedded (FFPE) Tissues:

  • Deparaffinize and rehydrate sections following standard protocols

  • Critical modification: Extend antigen retrieval to 30 minutes in citrate buffer

  • Add 10 mM sodium butyrate to all buffers to inhibit HDACs

  • Increase primary antibody concentration to 1:10-1:30 dilution

  • Extend primary antibody incubation to 48 hours at 4°C for deeper tissue penetration

  • Use tyramide signal amplification (TSA) systems for enhanced detection

For Fresh Frozen Tissues:

  • Use cold 4% paraformaldehyde fixation for 1 hour

  • Perform mild antigen retrieval (10 minutes) to preserve tissue architecture

  • Block endogenous peroxidases with 0.3% H₂O₂ before antibody incubation

  • Dilute antibody to 1:30-1:50 in buffer containing 0.1% Triton X-100

  • Include DAPI counterstain to visualize nuclear localization

For all tissue types, confirm specificity by pre-incubating the antibody with crotonylated peptides as a blocking control. Co-staining with markers of active transcription (e.g., H3K27ac or RNA Pol II) can provide functional context for the crotonylation patterns observed .

What are the most effective methods for quantifying changes in HIST1H2BC K11 crotonylation across experimental conditions?

Quantifying changes in HIST1H2BC K11 crotonylation requires selection of appropriate techniques based on experimental scale and resolution requirements:

1. Western Blot Densitometry:

  • Most accessible method for relative quantification

  • Normalize crotonyl-K11 signal to total H2B levels

  • Linear dynamic range is typically 5-10 fold change

  • Suitable for comparing treatments with substantial effects

2. Quantitative Mass Spectrometry:

  • Gold standard for absolute quantification

  • Sample preparation:

    • Digest histones with trypsin under propionylation conditions

    • Enrich for crotonylated peptides using antibody-based pulldown

  • Use targeted approaches like parallel reaction monitoring (PRM) or SWATH-MS

  • Create calibration curves with synthetic crotonylated peptides for absolute quantification

  • Can detect changes as small as 5-10% in modification levels

3. ChIP-qPCR and ChIP-seq:

  • For measuring genome-wide distribution changes

  • Normalize to input DNA and to appropriate controls (IgG, total H2B)

  • For ChIP-seq, use spike-in normalization with exogenous chromatin (e.g., Drosophila) to account for global changes

  • Bioinformatic analysis should include peak calling specifically optimized for histone modifications

4. Quantitative Immunofluorescence:

  • For spatial analysis within cellular compartments

  • Use automated image acquisition and analysis software

  • Normalize fluorescence intensity to DAPI or other nuclear markers

  • Z-stack imaging ensures complete nuclear signal capture

Comparative Table of Quantification Methods:

MethodSensitivityThroughputSpatial ResolutionRequired SampleTechnical Complexity
Western BlotMediumMediumNone10-20 μg histonesLow
MSVery HighLowNone50-100 μg histonesVery High
ChIP-qPCRHighLowGene-specific1-5×10⁶ cellsMedium
ChIP-seqHighHighGenome-wide5-10×10⁶ cellsHigh
IFMediumMediumSubcellularFixed cells/tissueMedium

For rigorous quantification, combining at least two complementary methods is recommended to validate observed changes in crotonylation levels .

How should researchers address unexpected cross-reactivity when using Crotonyl-HIST1H2BC (K11) Antibody?

When encountering unexpected cross-reactivity with Crotonyl-HIST1H2BC (K11) Antibody, implement this systematic troubleshooting workflow:

  • Verify the unexpected bands/signals:

    • Repeat the experiment with positive and negative controls

    • Compare molecular weights of unexpected bands to other known histone variants

    • Determine if cross-reactivity occurs consistently across different sample types

  • Conduct peptide competition assays:

    • Pre-incubate antibody with:

      • Target peptide (crotonylated K11 peptide)

      • Similar peptides with crotonylation at different lysine residues (K5, K12, K15)

      • Peptides with different modifications (acetylation, butyrylation) at K11

    • Decreasing signal with only the specific crotonylated K11 peptide confirms specificity

  • Modify antibody conditions:

    • Titrate antibody concentration (try 1:100, 1:500, 1:1000, 1:2000 dilutions)

    • Increase washing stringency with higher salt concentrations (150mM to 300mM NaCl)

    • Shorten incubation time to reduce non-specific binding

    • Test different blocking agents (switch between BSA, casein, or commercial blockers)

  • Enzymatic validation:

    • Treat samples with recombinant histone deacetylases/decrotonylases

    • If cross-reactive bands persist after treatment while the K11 signal disappears, this indicates non-specific binding

  • Confirm with orthogonal methods:

    • Use mass spectrometry to identify the actual proteins/modifications detected

    • Test a different antibody against the same epitope from another vendor

    • For ChIP applications, validate peaks by motif analysis to ensure enrichment at expected genomic features

Document all troubleshooting steps and findings to establish valid working parameters for the specific experimental system and application .

What factors can affect the stability and shelf-life of Crotonyl-HIST1H2BC (K11) Antibody, and how should it be stored for optimal performance?

Multiple factors affect the stability and performance of Crotonyl-HIST1H2BC (K11) Antibody, requiring careful storage and handling:

Critical Storage Parameters:

  • Temperature Considerations:

    • Store unopened antibody at -20°C for long-term stability

    • Working aliquots can be stored at 4°C for up to 2 weeks

    • Avoid repeated freeze-thaw cycles; create single-use aliquots (10-20 μL)

    • Temperature fluctuations accelerate degradation of polyclonal antibodies

  • Buffer Composition Effects:

    • The standard storage buffer (50% glycerol, 0.01M PBS, pH 7.4, 0.03% Proclin 300) maintains stability

    • Do not add sodium azide as preservative as it can affect epitope recognition

    • Monitor for precipitation; if observed, centrifuge before use

    • For extended storage periods (>6 months), verify pH remains 7.2-7.6

  • Light Exposure:

    • Protect from direct light exposure, particularly if conjugated to fluorophores

    • Store in amber tubes or wrapped in aluminum foil

    • Excessive light exposure can cause oxidative damage to antibody proteins

  • Contamination Prevention:

    • Use sterile technique when handling

    • Filter buffers used for dilution through 0.22 μm filters

    • Add antimicrobial agents only if antibody will be stored at 4°C for extended periods

  • Stability Testing Protocol:

    • Periodically test antibody performance with consistent positive controls

    • Create a reference sample at first use and compare signal intensity over time

    • Document batch variations and sensitivity changes

Recommended Storage and Handling Table:

Storage DurationRecommended ConditionHandling Notes
<1 month4°CNo aliquoting needed if used frequently
1-6 months-20°C in aliquotsAllow to reach room temperature before opening
>6 months-80°C in aliquotsTest activity before critical experiments
Working dilutions4°C for <1 weekAdd 0.1% BSA to prevent adsorption to tubes

Under optimal storage conditions, the antibody maintains >90% activity for approximately 12 months, after which gradual decline in sensitivity may be observed .

How can researchers distinguish between true crotonylation signals and artifacts when using Crotonyl-HIST1H2BC (K11) Antibody?

Distinguishing genuine crotonylation signals from artifacts requires a comprehensive validation approach:

  • Technical Validation Controls:

    • Secondary antibody-only control to identify non-specific binding

    • IgG isotype control to establish background signal levels

    • Pre-immune serum control (if available from antibody manufacturer)

    • Signal persistence after general histone deacetylase inhibition but reduction after specific decrotonylase treatment

  • Biological Validation Approaches:

    • Genetic manipulation of crotonylation machinery:

      • Knockdown/knockout of known regulators (p300/CBP with crotonylation activity)

      • Overexpression of verified decrotonylases

    • Metabolic manipulation:

      • Increase crotonylation by supplementing media with crotonate or crotonyl-CoA

      • Decrease crotonylation by restricting carbon sources that feed into crotonyl-CoA production

  • Signal Pattern Analysis:

    • True K11 crotonylation typically shows:

      • Nuclear localization in IF experiments

      • Enrichment at transcriptionally active regions in ChIP experiments

      • Correlation with other active histone marks (H3K27ac, H3K4me3)

      • Molecular weight consistent with histone H2B (approximately 14 kDa)

    • Common artifacts include:

      • Cytoplasmic staining

      • Multiple bands on Western blots distant from expected molecular weight

      • Signals that don't respond to known crotonylation modulators

      • Enrichment at heterochromatic regions (unusual for crotonylation)

  • Orthogonal Confirmation:

    • Mass spectrometry validation of crotonylation at K11

    • Comparison with results from a different crotonyl-K11 antibody

    • Multiple application testing (if signal appears in WB but not in ChIP or IF, question specificity)

  • Quantitative Assessment:

    • Signal-to-noise ratio should be >3:1 for reliable detection

    • Compare signal dynamic range across experimental conditions to expected biological variance

How can Crotonyl-HIST1H2BC (K11) Antibody be integrated into multi-omics approaches to study epigenetic regulation?

Integrating Crotonyl-HIST1H2BC (K11) Antibody into multi-omics frameworks requires thoughtful experimental design to establish meaningful correlations between crotonylation and other molecular features:

Integrated Experimental Design Approach:

  • ChIP-seq + RNA-seq Integration:

    • Perform ChIP-seq with Crotonyl-HIST1H2BC (K11) Antibody and RNA-seq on the same biological samples

    • Computational analysis workflow:

      • Map K11 crotonylation peaks relative to transcription start sites

      • Calculate correlation between peak intensity and transcript abundance

      • Identify gene sets where K11 crotonylation shows strongest correlation with expression

    • Expected outcome: Identification of gene networks specifically regulated by K11 crotonylation

  • Metabolomics + Crotonylation Profiling:

    • Conduct targeted metabolomics focusing on acyl-CoA intermediates alongside histone crotonylation analysis

    • Extract cells using dual-purpose protocols that preserve both metabolites and histones

    • Correlate intracellular crotonyl-CoA levels with K11 crotonylation intensity

    • This approach reveals how metabolic flux affects the epigenetic landscape

  • Proteomics + Crotonylation:

    • Employ proteomics to identify "readers" of K11 crotonylation using protein pull-down assays

    • Use synthetic peptides with K11 crotonylation as bait

    • Identify proteins that differentially bind to crotonylated versus non-crotonylated peptides

    • Map the protein interaction network surrounding K11-crotonylated histones

  • Single-Cell Multi-omics:

    • Adapt CUT&Tag protocols for Crotonyl-HIST1H2BC (K11) Antibody for single-cell applications

    • Combine with single-cell RNA-seq in platforms like 10X Genomics

    • This reveals cell-to-cell heterogeneity in crotonylation patterns and their relationship to transcriptional states

  • Spatial Omics Integration:

    • Utilize Crotonyl-HIST1H2BC (K11) Antibody in spatial profiling techniques

    • Combine immunofluorescence detection of K11 crotonylation with spatial transcriptomics

    • Map the spatial distribution of crotonylation in relation to gene expression zones within tissues

These multi-omics approaches collectively provide a systems-level understanding of how K11 crotonylation functions within the broader context of cellular regulation, revealing both regulatory mechanisms and functional consequences of this epigenetic modification .

What role does HIST1H2BC K11 crotonylation play in cancer epigenetics, and how can this antibody advance cancer research?

HIST1H2BC K11 crotonylation has emerging significance in cancer epigenetics, with the antibody enabling several critical research directions:

  • Altered Crotonylation Patterns in Cancer:
    Research utilizing the Crotonyl-HIST1H2BC (K11) Antibody has revealed distinct crotonylation patterns in various cancer types compared to normal tissues. In particular, studies have shown that K11 crotonylation is frequently dysregulated in colorectal and prostate cancers, where it associates with altered expression of genes involved in cell proliferation and metabolism. The antibody enables precise mapping of these alterations through ChIP-seq and immunohistochemistry applications, revealing cancer-specific epigenetic signatures that may serve as biomarkers.

  • Metabolic Reprogramming and Crotonylation:
    Cancer cells undergo metabolic reprogramming that affects acyl-CoA pools, including crotonyl-CoA. The antibody allows researchers to directly link these metabolic changes to epigenetic alterations by monitoring K11 crotonylation under different metabolic conditions. This connection is particularly relevant in cancers with mitochondrial dysfunction or altered fatty acid metabolism, where crotonylation may serve as an epigenetic readout of the cancer metabolic state.

  • Therapeutic Targeting Opportunities:
    Enzymes regulating crotonylation (writers, erasers, readers) represent potential therapeutic targets. The antibody enables:

    • Screening compounds that modulate K11 crotonylation levels

    • Monitoring target engagement in drug development pipelines

    • Assessing combination therapies with existing epigenetic drugs (HDAC inhibitors)

    • Identifying synthetic lethal interactions between crotonylation machinery and cancer mutations

  • Translational Applications:
    In the clinical research setting, the antibody facilitates:

    • Development of prognostic indicators based on K11 crotonylation patterns

    • Patient stratification for precision medicine approaches

    • Monitoring epigenetic changes during treatment response and resistance development

  • Technical Applications Table for Cancer Research:

    ApplicationCancer Research UseTechnical Considerations
    ChIP-seqGenome-wide mapping of crotonylation changesUse patient-derived xenografts for human-specific antibody applications
    IHC/IFTissue-specific localization in tumor samplesOptimize antigen retrieval for FFPE cancer tissue microarrays
    Western BlotQuantitative assessment across cancer cell linesCompare with other acylation marks to establish cancer-specific patterns
    CUT&TagSingle-cell analysis of tumor heterogeneityModify protocol for low-input samples from clinical specimens

By leveraging the Crotonyl-HIST1H2BC (K11) Antibody in these applications, cancer researchers can explore how this specific epigenetic modification contributes to oncogenesis and identify novel therapeutic strategies targeting the crotonylation pathway .

How can Crotonyl-HIST1H2BC (K11) Antibody be used to study the interplay between histone crotonylation and other epigenetic modifications?

The Crotonyl-HIST1H2BC (K11) Antibody enables sophisticated investigation of crotonylation's relationship with other epigenetic marks through several methodological approaches:

  • Sequential ChIP (Re-ChIP) Methodology:
    This technique allows detection of co-occurrence of K11 crotonylation with other modifications on the same nucleosomes:

    • First round: Immunoprecipitate with Crotonyl-HIST1H2BC (K11) Antibody

    • Elute under mild conditions to preserve protein-DNA complexes

    • Second round: Immunoprecipitate with antibodies against other modifications (H3K27ac, H3K4me3, etc.)

    • Analysis reveals genomic regions where both modifications co-exist

    Critical optimization: Adjust elution conditions between rounds to maintain epitope integrity while releasing immune complexes (use low pH glycine buffer rather than SDS-containing buffers).

  • Mass Spectrometry-Based Combinatorial Analysis:

    • Enrich histones containing K11 crotonylation using the antibody

    • Subject enriched fraction to bottom-up proteomics with multiple enzyme digestion

    • Analyze resulting peptides to identify modifications co-occurring on the same histone molecule

    • This approach can reveal previously unknown modification patterns and their relative abundances

  • Proximity Ligation Assay (PLA) Applications:

    • Use Crotonyl-HIST1H2BC (K11) Antibody alongside antibodies against other modifications

    • PLA signal indicates co-occurrence within 40nm (approximately nucleosomal distance)

    • Quantify signal frequency in different nuclear regions or cell types

    • This technique provides spatial information about modification co-occurrence that ChIP methods cannot

  • Targeted Epigenetic Editing:

    • Use CRISPR-dCas9 fusion systems with crotonylating enzymes to increase K11 crotonylation at specific loci

    • Monitor changes in other modifications using ChIP-qPCR

    • This approach reveals causal relationships between crotonylation and other modifications

  • Inhibitor Studies with Modification-Specific Quantification:

    • Treat cells with inhibitors of specific epigenetic writers/erasers

    • Measure changes in K11 crotonylation using the antibody

    • Create modification networks based on inhibitor effects

    • Example: HDAC inhibitors may increase both acetylation and crotonylation, while p300 inhibitors might reduce both

This multi-method approach reveals whether K11 crotonylation operates independently or synergistically with other modifications, providing insights into the combinatorial epigenetic code. Current research suggests K11 crotonylation works cooperatively with acetylation at nearby residues to maintain open chromatin states while potentially competing with methylation marks at the same residue .

What emerging technologies might enhance the research applications of Crotonyl-HIST1H2BC (K11) Antibody?

Several cutting-edge technologies are positioned to dramatically expand the research applications of Crotonyl-HIST1H2BC (K11) Antibody:

  • Next-Generation CUT&Tag and CUT&RUN Applications:
    The adaptation of these techniques for Crotonyl-HIST1H2BC (K11) Antibody offers significant advantages over traditional ChIP:

    • Reduced input material (as few as 1,000 cells)

    • Improved signal-to-noise ratio for detecting low-abundance modifications

    • Compatibility with single-cell applications to reveal cell-to-cell variation in crotonylation patterns

    • Enhanced spatial resolution to precisely map K11 crotonylation relative to transcription factor binding sites

    Implementation requires optimization of tagmentation conditions specifically for the K11 antibody, with particular attention to salt concentrations that maintain epitope recognition while allowing efficient tagmentation.

  • Live-Cell Imaging of Crotonylation Dynamics:

    • Development of recombinant nanobodies derived from Crotonyl-HIST1H2BC (K11) Antibody

    • Fusion of nanobodies with fluorescent proteins for live-cell applications

    • This approach would enable real-time monitoring of K11 crotonylation changes during cell cycle, differentiation, or in response to metabolic perturbations

    • Current limitation: development of modification-specific nanobodies requires extensive screening and validation

  • Spatial Multi-omics Integration:

    • Combining Crotonyl-HIST1H2BC (K11) Antibody immunofluorescence with spatial transcriptomics

    • Technologies like 10X Visium or Slide-seq can be paired with immunofluorescence

    • This correlation reveals spatial relationships between crotonylation patterns and gene expression domains

    • Particularly valuable for studying crotonylation in complex tissues like brain, where metabolic gradients may create epigenetic zonation

  • Cryo-Electron Microscopy Applications:

    • Using Crotonyl-HIST1H2BC (K11) Antibody fragments for cryo-EM studies

    • This approach can reveal structural consequences of K11 crotonylation on nucleosome organization

    • Potential to visualize how reader proteins interact with crotonylated nucleosomes

    • Technical challenge: adapting the antibody for cryo-EM compatibility while maintaining specificity

  • AI-Enhanced Analysis Pipelines:

    • Machine learning algorithms trained on ChIP-seq data from Crotonyl-HIST1H2BC (K11) Antibody

    • Prediction of crotonylation patterns based on DNA sequence and other epigenetic markers

    • Integration with multi-omics data to build comprehensive regulatory networks

    • These computational approaches extend the value of experimental data by enabling in silico predictions of crotonylation patterns in unstudied conditions

These emerging technologies collectively enhance the spatial, temporal, and contextual understanding of K11 crotonylation, moving beyond simple detection toward functional integration within broader cellular systems .

What are the current limitations of the Crotonyl-HIST1H2BC (K11) Antibody, and how might future antibody development address these challenges?

Current Crotonyl-HIST1H2BC (K11) Antibody limitations and potential future improvements include:

  • Cross-Reactivity Challenges:
    Current limitation: Potential cross-reactivity with similar histone modifications (especially acetylation) or crotonylation at other lysine residues due to the polyclonal nature of the antibody.

    Future developments:

    • Generation of monoclonal antibodies with enhanced specificity through advanced selection techniques

    • Development of recombinant antibodies through phage display specifically targeting the unique structural features of the crotonyl group at K11

    • Use of synthetic biology approaches to create high-specificity binding proteins based on modified reader domains that naturally recognize crotonylation

  • Sensitivity Limitations:
    Current limitation: Detection of low-abundance crotonylation can be challenging, especially in samples with limited material or when crotonylation occurs at low stoichiometry.

    Future developments:

    • Integration with signal amplification technologies like tyramide signal amplification or rolling circle amplification

    • Development of proximity-based detection methods with improved sensitivity

    • Creation of branched DNA technologies adapted for antibody-based crotonylation detection

    • Engineering antibodies with higher affinity constants through directed evolution

  • Application-Specific Optimization Requirements:
    Current limitation: Extensive optimization needed for each experimental application (ChIP, IF, WB) and for different sample types.

    Future developments:

    • Development of application-specific antibody formulations

    • Creation of validation kits with positive and negative controls for each application

    • Standardized protocols optimized for specific tissue types

    • Machine learning-based prediction of optimal conditions for new sample types based on previous data

  • Technological Advances on the Horizon:

    Current LimitationEmerging SolutionTimelineTechnical Hurdles
    Batch-to-batch variabilityRecombinant antibody technology1-3 yearsExpression system optimization
    Limited quantificationMass cytometry-compatible antibodies2-4 yearsMetal conjugation efficiency
    Low multiplexing capacityDNA-barcoded antibodies for CITE-seq1-2 yearsMaintaining epitope recognition after barcoding
    Fixed timepoint analysisEngineered biosensors based on antibody binding domains3-5 yearsProtein engineering challenges
  • Integration with Non-Antibody Technologies:
    Future developments will likely include hybrid approaches that combine the specificity of antibody recognition with other technologies:

    • CRISPR-based genomic screening to correlate genetic factors with K11 crotonylation patterns

    • Nanopore sequencing adaptations to directly detect modified histones during DNA sequencing

    • Integration with emerging epigenetic editing tools to manipulate crotonylation at specific genomic loci

These advancements will collectively address current limitations and expand the utility of Crotonyl-HIST1H2BC (K11) detection across a broader range of experimental systems and biological questions .

How might computational methods enhance the analysis of data generated using Crotonyl-HIST1H2BC (K11) Antibody?

Advanced computational approaches can significantly enhance the value of data generated with Crotonyl-HIST1H2BC (K11) Antibody through several methodological innovations:

  • Machine Learning for Signal Enhancement:

    • Convolutional neural networks (CNNs) can improve signal-to-noise ratio in ChIP-seq data

    • Supervised learning algorithms can be trained on high-confidence crotonylation datasets to identify patterns

    • Example implementation: DeepSignal or similar deep learning frameworks adapted for K11 crotonylation peak calling

    • This approach reduces false positives and recovers weak signals that might be missed by conventional analysis

  • Integrative Multi-Omics Analysis Frameworks:

    • Bayesian network models to integrate K11 crotonylation data with:

      • Transcriptomics (RNA-seq)

      • Other histone modifications (ChIP-seq)

      • Chromatin accessibility (ATAC-seq)

      • Metabolomics data

    • Implementation using tools like MOFA+ (Multi-Omics Factor Analysis) or similar frameworks

    • This creates comprehensive regulatory networks linking crotonylation to functional outcomes

  • Motif Analysis and Sequence Determinants:

    • Identification of DNA sequence motifs associated with K11 crotonylation enrichment

    • Analysis workflow:

      • Extract sequences under crotonylation peaks

      • Use tools like MEME Suite to identify enriched motifs

      • Compare to known transcription factor binding sites

      • Build predictive models of where crotonylation will occur based on sequence

    • This approach reveals potential regulatory factors recruiting crotonylation machinery

  • Comparative Epigenomics:

    • Systematic comparison of K11 crotonylation patterns across:

      • Cell types and tissues

      • Disease states versus healthy controls

      • Developmental timepoints

      • Species (for evolutionarily conserved patterns)

    • Implementation through analysis pipelines like Cistrome or custom R/Python workflows

    • This reveals context-specific functions and conserved roles of K11 crotonylation

  • Spatial Analysis for Immunofluorescence Data:

    • Advanced image analysis for K11 crotonylation IF data:

      • Nuclear segmentation algorithms to identify individual nuclei

      • Quantification of signal intensity, distribution patterns, and co-localization

      • Cell-type identification in heterogeneous tissues

    • Implementation through platforms like CellProfiler or custom ImageJ/Python scripts

    • This approach quantifies subtle changes in crotonylation patterns that may be missed by visual inspection

These computational approaches transform descriptive antibody-generated data into predictive models of K11 crotonylation function. As datasets accumulate, these methods will enable meta-analyses across studies, creating increasingly robust and generalizable insights into the biological roles of this epigenetic modification .

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.