Histone H3 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Core Characteristics

Histone H3 monoclonal antibodies are immunoglobulin molecules produced by identical immune cells cloned from a single parent cell. They exhibit high specificity for defined epitopes on histone H3, including unmodified, phosphorylated, methylated, or cleaved forms. Key features include:

  • Target Specificity: Recognition of specific residues (e.g., phosphorylated Ser28, unmodified Lys4) or cleavage sites (e.g., after Arg49 in NETosis) .

  • Species Reactivity: Broad cross-reactivity across humans, mice, rats, and other vertebrates, depending on the clone .

  • Applications: Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), flow cytometry (FC), and chromatin immunoprecipitation (ChIP) .

Cell Cycle and Mitosis

  • Phospho-Ser28 Detection (HTA28 Clone): Identifies M-phase cells by targeting histone H3 phosphorylated at serine 28, aiding in the study of mitotic activity in tissues like regenerating rat liver .

  • CENP-A Variant Recognition: Certain clones (e.g., D1H2) detect the centromere-specific histone H3 variant CENP-A, useful in chromosome segregation studies .

Epigenetic Modifications

  • Lysine Methylation: Antibodies like RM140 (monomethyl-Lys4) and RM340 (trimethyl-Lys4) enable mapping of epigenetic marks associated with transcriptional activation or repression .

  • Unmodified Lys4 Detection (RM186 Clone): Distinguishes unmodified histone H3 from acetylated or methylated forms, critical for studying gene silencing .

Disease Mechanisms

  • NETosis Detection: The 3D9 clone targets histone H3 cleaved at Arg49 during neutrophil extracellular trap (NET) formation, serving as a biomarker for NET-related pathologies like thrombosis or autoimmune diseases .

Antibody Clones and Their Specificities

The table below compares prominent histone H3 monoclonal antibody clones:

CloneSpecificity/ModificationApplicationsHost/IsotypeCross-ReactivitySource
HTA28Phospho-Ser28IHC, IFMouse/IgGRat, human[PMC]
RM186Unmodified Lys4WB, ICC, ChIPRabbit/IgGHuman, vertebratesRevMab
D1H2Total H3, CENP-AWB, IHC, IF, FCRabbit/IgGHuman, mouse, rat, monkeyCell Signaling
3D9Cleaved H3 (Arg49)WB, IFMouse/IgGHuman[PMC]
RM140Monomethyl-Lys4WB, ChIP, ICCRabbit/IgGHuman, vertebratesRevMab

Validation and Optimization

  • Housekeeping Protein Validation: Histone H3 antibodies used for normalization (e.g., loading controls) require validation to confirm stable expression under experimental conditions .

  • Titration: Optimal dilution varies by application; for example, RM140 requires 0.5–2 µg/mL for ChIP vs. 1:50–1:500 for IF .

Research Findings and Innovations

  • Mitotic Index Quantification: HTA28 immunostaining doubled the detection sensitivity of M-phase cells compared to traditional histology in rat liver studies, reducing false positives from pyknotic nuclei .

  • NET-Specific Cleavage: The 3D9 antibody revealed histone H3 cleavage at Arg49 as a conserved NETosis marker, detectable in human tissues and murine models .

  • Epigenetic Crosstalk: RM186 demonstrated that unmodified Lys4 coexists with Thr3 phosphorylation, suggesting interplay between PTMs in transcriptional regulation .

Limitations and Challenges

  • Cross-Reactivity Risks: Some clones (e.g., D1H2) may detect homologous epitopes in non-target species without validation .

  • Fixation Sensitivity: Delayed tissue fixation abolishes phospho-Ser28 signal, necessitating rapid processing for mitosis studies .

Product Specs

Buffer
Phosphate Buffered Saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Our standard lead time for dispatching orders is 1-3 business days upon receipt. Delivery times may vary depending on the order fulfillment method and destination. For specific delivery timelines, please consult your local distributor.
Uniprot No.

Q&A

What is Histone H3 and why is it significant in epigenetic research?

Histone H3 is one of the four core components (along with H2A, H2B, and H4) that make up the nucleosome core particle, the fundamental unit of chromatin. Nucleosomes consist of 146 base pairs of DNA wrapped around an octamer of these core histone proteins . Histone H3 plays a central role in regulating DNA accessibility through its N-terminal tail, which undergoes various post-translational modifications that influence chromatin structure and gene expression . These modifications, including acetylation, methylation, and phosphorylation, constitute part of the "histone code" that regulates transcription, DNA repair, replication, and chromosomal stability . The ability to detect and quantify Histone H3 and its modifications is therefore critical for understanding epigenetic regulation mechanisms.

What distinguishes monoclonal antibodies from polyclonal antibodies when studying Histone H3?

Monoclonal antibodies for Histone H3 offer several advantages over polyclonal alternatives in research applications:

FeatureMonoclonal AntibodiesPolyclonal Antibodies
SourceSingle B-cell clone (e.g., mouse) Multiple B-cells (e.g., rabbit)
Epitope recognitionSingle epitope (highly specific)Multiple epitopes
Batch consistencyHigh lot-to-lot reproducibilityPotential variability between lots
Background signalTypically lowerCan be higher
ApplicationsExcellent for specific modificationsBetter for detecting total protein

What are the primary applications of Histone H3 monoclonal antibodies in epigenetic research?

Histone H3 monoclonal antibodies are versatile tools employed across multiple research techniques:

  • Chromatin Immunoprecipitation (ChIP): Used to identify genomic regions associated with specific histone modifications, revealing regulatory elements and transcription factor binding sites .

  • ChIP-Sequencing (ChIP-Seq): Combines ChIP with next-generation sequencing to map genome-wide distribution of histone modifications and assess global epigenetic landscapes .

  • Western Blotting (WB): Quantifies Histone H3 protein levels and specific modifications in different experimental conditions, typically using a 1:1000 dilution for optimal results .

  • Immunofluorescence (IF)/Immunocytochemistry (ICC): Visualizes the nuclear distribution of Histone H3 and its modifications within cells, providing spatial information about chromatin organization .

  • Immunohistochemistry (IHC): Examines Histone H3 distribution in tissue sections, often used in cancer research to correlate histone modifications with pathological conditions .

  • Immunoprecipitation (IP): Isolates Histone H3-containing complexes to study protein-protein interactions within chromatin-remodeling machinery .

  • ELISA: Provides quantitative analysis of specific histone modifications in purified samples .

  • CUT&RUN/CUT&Tag: Advanced techniques that offer higher resolution than traditional ChIP for mapping histone modifications .

How should I optimize ChIP-Seq experiments using Histone H3 monoclonal antibodies?

Optimizing ChIP-Seq with Histone H3 monoclonal antibodies requires careful consideration of several factors:

  • Antibody Selection: Choose antibodies validated specifically for ChIP-Seq applications. For instance, antibodies raised against the N-terminus of histone H3 often perform well in chromatin immunoprecipitation .

  • Crosslinking Conditions: Standard formaldehyde fixation (1% for 10 minutes) is generally appropriate for Histone H3 ChIP, but modifications at different residues may require optimization of crosslinking time.

  • Sonication Parameters: Aim for chromatin fragments of 200-500 bp for optimal resolution. Histone H3 ChIP typically requires less aggressive sonication than transcription factor ChIP due to the abundance of the target.

  • Antibody Concentration: Use the manufacturer's recommended amount; excess antibody can increase background. For monoclonal Histone H3 antibodies, 3-5 μg per ChIP reaction is often sufficient .

  • Controls: Always include:

    • Input DNA (non-immunoprecipitated chromatin)

    • Negative control (IgG from the same species as your primary antibody)

    • Positive control (a known binding region for your histone mark)

  • Washing Stringency: Balance between removing non-specific binding and maintaining specific interactions; generally, more stringent washes are possible with monoclonal antibodies due to their high specificity.

  • Library Preparation: Use library preparation methods that accommodate the typically smaller amounts of DNA obtained from histone ChIP compared to transcription factor ChIP.

  • Sequencing Depth: For broad histone marks, 20-30 million mapped reads per sample is often sufficient; for more focal marks, deeper sequencing may be required.

Using specialized ChIP kits designed for histone modifications, such as high-sensitivity kits, can improve results when working with limited samples or challenging modifications .

What are the key considerations for Western blot analysis of Histone H3?

Western blotting for Histone H3 requires specific adaptations to standard protocols:

  • Sample Preparation:

    • Acid extraction methods are preferred for enriching histones

    • Use HeLa acid extract as a positive control

    • Include protease and phosphatase inhibitors to preserve modifications

  • Gel Selection:

    • Use high percentage (15-18%) SDS-PAGE gels to resolve the 15-17 kDa Histone H3 protein

    • Consider specialized Triton-Acid-Urea (TAU) gels for separating differently modified histone variants

  • Transfer Conditions:

    • PVDF membranes typically work better than nitrocellulose for histone proteins

    • Use shorter transfer times and lower methanol concentration to improve small protein transfer

  • Antibody Dilution:

    • For most Histone H3 monoclonal antibodies, a 1:1000 dilution is recommended for Western blotting

    • Always prepare antibodies in fresh blocking buffer

  • Blocking Conditions:

    • 5% non-fat dry milk in TBST is generally effective

    • For phospho-specific antibodies, BSA is preferred over milk

  • Detection Method:

    • Enhanced chemiluminescence provides sufficient sensitivity for most applications

    • Fluorescent detection allows for multiplex analysis of different modifications

  • Stripping and Reprobing:

    • Mild stripping conditions are recommended if the membrane will be reprobed

    • Consider running multiple gels instead of stripping when studying multiple modifications

When analyzing post-translational modifications, loading controls are essential - typically total Histone H3 serves as an appropriate normalization control for modification-specific antibodies.

How can I validate the specificity of a Histone H3 monoclonal antibody?

Rigorous validation is critical when working with Histone H3 antibodies to ensure experimental reliability:

  • Peptide Competition Assay: Pre-incubate the antibody with increasing concentrations of the immunizing peptide before use in your application. Specific signal should decrease proportionally with increasing peptide concentration.

  • Knockout/Knockdown Controls:

    • Use genetic knockouts of the target histone variant when possible

    • For modifications, employ cells treated with inhibitors of the relevant modifying enzymes

    • CRISPR-edited cells with specific mutations at modification sites provide excellent controls

  • Cross-Reactivity Testing:

    • Test against related histone variants (H3.1, H3.2, H3.3)

    • Check reactivity with the same modification at different residues (e.g., ensure H3K9ac antibody doesn't detect H3K14ac)

  • Multiple Detection Methods: Confirm findings using orthogonal techniques (e.g., if using ChIP-Seq, validate key regions by ChIP-qPCR).

  • Dot Blot Analysis: Test antibody specificity against a panel of modified and unmodified histone peptides.

  • Mass Spectrometry Correlation: Compare antibody-based detection with mass spectrometry results when feasible.

  • Species Cross-Reactivity Assessment: If working with non-human samples, verify the antibody works in your species of interest. While many Histone H3 antibodies show broad species reactivity due to sequence conservation , experimental validation is still necessary.

  • Batch Testing: When receiving a new lot of antibody, compare it to previous lots using the same experimental conditions to ensure consistent performance.

Why might I observe unexpected molecular weight bands when detecting Histone H3?

Unexpected bands in Histone H3 Western blots can result from several biological and technical factors:

  • Histone Modifications: Extensive post-translational modifications can alter the apparent molecular weight of Histone H3. While the calculated molecular weight is approximately 15 kDa , modifications like poly-ubiquitination can significantly increase the observed size.

  • Histone Variants: The human genome contains multiple Histone H3 genes encoding variants (H3.1, H3.2, H3.3, CENP-A, etc.) that may appear as distinct bands despite similar sizes.

  • Histone Dimers/Oligomers: Incomplete denaturation can result in histone complexes appearing at higher molecular weights, typically as multiples of the monomer size.

  • Proteolytic Degradation: Improper sample handling can lead to degradation products appearing as lower molecular weight bands. Always use fresh protease inhibitors and maintain samples at appropriate temperatures.

  • Non-specific Binding: Some antibodies may cross-react with other proteins, particularly other histones. Check the antibody datasheet for known cross-reactivity .

  • Isoform Recognition: Some antibodies may preferentially recognize specific isoforms or modified forms of Histone H3, resulting in multiple bands.

To address these issues:

  • Use purified recombinant Histone H3 as a positive control

  • Include acid-extracted histones from HeLa cells as a reference

  • Consider using gradient gels for better resolution of closely spaced bands

  • If studying specific modifications, include appropriate controls for that modification

How do I resolve conflicting ChIP-Seq data from different Histone H3 monoclonal antibodies?

Discrepancies between ChIP-Seq datasets generated using different Histone H3 antibodies require systematic investigation:

  • Epitope Mapping: Determine the exact epitopes recognized by each antibody. Antibodies raised against different regions of Histone H3 (e.g., N-terminal tail vs. globular domain) may yield different results due to epitope accessibility in chromatin .

  • Modification Interference: Some antibodies against unmodified Histone H3 may have reduced binding when nearby residues are modified. For example, phosphorylation at Ser10 can affect the recognition of acetylation at Lys9.

  • Antibody Class Effects: Compare results from monoclonal versus polyclonal antibodies. Monoclonal antibodies provide high specificity but may be more sensitive to epitope masking, while polyclonal antibodies may offer broader recognition .

  • Technical Validation:

    • Perform sequential ChIP (re-ChIP) to determine if the targets overlap

    • Validate selected regions by ChIP-qPCR using both antibodies

    • Assess correlation between replicates for each antibody

  • Bioinformatic Analysis:

    • Compare peak distributions relative to genomic features

    • Examine peak shapes and signal-to-noise ratios

    • Analyze enrichment of transcription factor motifs within peaks

    • Use peak overlap statistics to quantify similarities and differences

  • Correlation with Functional Data: Integrate RNA-Seq or other functional genomic data to determine which antibody's binding profile better correlates with expected biological functions.

  • Literature Consistency: Compare your findings with published datasets using the same antibodies to identify potential technical artifacts versus biologically meaningful differences.

When reporting conflicting results, clearly document the exact antibody clone used, lot number, and experimental conditions to aid reproducibility and interpretation.

What approaches can resolve issues with high background in immunofluorescence studies using Histone H3 antibodies?

High background in Histone H3 immunofluorescence experiments can significantly impair data quality but can be addressed through multiple strategies:

  • Fixation Optimization:

    • Test different fixatives (4% paraformaldehyde vs. methanol)

    • Adjust fixation time (shorter times may preserve epitope accessibility)

    • For some modifications, dual fixation methods may be optimal

  • Permeabilization Adjustments:

    • Optimize detergent concentration (0.1-0.5% Triton X-100)

    • Consider shorter permeabilization times to prevent over-extraction

  • Blocking Improvements:

    • Increase blocking time (1-2 hours at room temperature or overnight at 4°C)

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Include 0.1-0.3% Triton X-100 in blocking buffer to reduce non-specific binding

  • Antibody Dilution Optimization:

    • Titrate primary antibody; for Histone H3 monoclonal antibodies, start with 1:200-1:1000

    • Include 0.05% Tween-20 in antibody dilution buffer

    • Extend primary antibody incubation (overnight at 4°C often yields better signal-to-noise)

  • Washing Protocol Enhancement:

    • Increase number of washes (5-6 washes of 5-10 minutes each)

    • Use PBS with 0.05-0.1% Tween-20 for more stringent washing

  • Secondary Antibody Considerations:

    • Further dilute secondary antibody (1:500-1:2000)

    • Pre-adsorb secondary antibody against fixed cells

    • Select highly cross-adsorbed secondary antibodies

  • Autofluorescence Reduction:

    • Include an autofluorescence quenching step (0.1% sodium borohydride or commercial quenchers)

    • Adjust imaging parameters to minimize autofluorescence detection

  • Controls:

    • Include no-primary-antibody controls

    • Use isotype controls at the same concentration as the primary antibody

    • When possible, include genetic controls (knockdown/knockout)

  • Mounting Media Selection:

    • Use anti-fade mounting media with DAPI for nuclear counterstaining

    • Some mounting media formulations can reduce background fluorescence

These optimizations should be performed systematically, changing one parameter at a time while documenting the effects on signal-to-noise ratio.

How can Histone H3 monoclonal antibodies be used to study chromatin dynamics during cell differentiation?

Investigating chromatin remodeling during differentiation using Histone H3 antibodies requires multifaceted approaches:

  • Temporal ChIP-Seq Analysis:

    • Perform ChIP-Seq for multiple Histone H3 modifications (H3K4me3, H3K27me3, H3K27ac, H3K9me3) at defined timepoints during differentiation

    • Track the dynamics of bivalent domains (H3K4me3/H3K27me3) that often mark developmental genes

    • Integrate with transcription factor binding data to identify pioneering factors

  • Single-Cell Applications:

    • Apply CUT&Tag or CUT&RUN technologies for single-cell epigenomic profiling

    • Correlate histone modification patterns with differentiation trajectories

    • Identify heterogeneity in chromatin states within seemingly uniform populations

  • Live-Cell Imaging:

    • Use fluorescently-tagged nanobodies derived from Histone H3 monoclonal antibodies

    • Track real-time changes in histone modification distribution during differentiation

    • Combine with fluorescent reporters for lineage-specific genes

  • Nucleosome Positioning Analysis:

    • Integrate Histone H3 ChIP-Seq with nucleosome positioning assays (MNase-Seq)

    • Analyze changes in nucleosome occupancy at regulatory elements

    • Identify pioneer factor binding sites by nucleosome displacement patterns

  • Histone Turnover Studies:

    • Use SNAP-tagged histone variants combined with immunoprecipitation

    • Correlate modification patterns with histone turnover rates

    • Identify regions of high chromatin dynamics during cell fate transitions

  • Mass Spectrometry Integration:

    • Immunoprecipitate modified histones for quantitative mass spectrometry

    • Identify combinatorial modification patterns (modifications occurring on the same histone tail)

    • Quantify changes in modification stoichiometry during differentiation

  • Functional Validation:

    • Use CRISPR-mediated targeting of histone modifying enzymes to specific loci

    • Validate the functional importance of specific modifications in differentiation

    • Correlate changes in chromatin accessibility (ATAC-Seq) with histone modifications

This integrated approach provides comprehensive insights into the epigenetic regulation of cell differentiation, revealing both global trends and locus-specific mechanisms.

What strategies enable the study of Histone H3 modifications in limited clinical samples or rare cell populations?

Investigating histone modifications in scarce samples requires specialized approaches:

  • Low-Input ChIP-Seq Protocols:

    • Utilize carrier ChIP methods (adding exogenous chromatin from another species)

    • Employ tagmentation-based library preparation to reduce material loss

    • Consider spike-in normalization for quantitative comparisons between samples

  • CUT&RUN and CUT&Tag Adaptations:

    • These techniques require substantially fewer cells than conventional ChIP (1,000-50,000 cells)

    • Use automated CUT&RUN/CUT&Tag protocols to minimize handling losses

    • Optimize wash conditions for maximum specificity with minimal cell loss

  • Microfluidic Approaches:

    • Implement microfluidic devices for ChIP with as few as 100 cells

    • Combine with on-chip library preparation to minimize transfer losses

    • Consider parallel processing of multiple modifications from the same sample

  • Cell Isolation Strategies:

    • Use gentle FACS sorting with histone modification-preserving buffers

    • Consider nuclei isolation instead of whole cells for better purity

    • Implement laser capture microdissection for tissue-specific analysis

  • Signal Amplification Methods:

    • Employ antibody-DNA conjugates with rolling circle amplification

    • Use proximity ligation assays to detect specific modification patterns

    • Consider tyramide signal amplification for immunofluorescence detection

  • Single-Cell Multi-Omics Integration:

    • Combine scRNA-Seq with scCUT&Tag to correlate transcription with histone modifications

    • Implement computational methods to infer modification patterns from accessibility data

    • Use multi-modal assays that simultaneously profile chromatin and transcription

  • Preservation Protocols:

    • Develop optimized fixation protocols for biobanked samples

    • Validate histone modification stability in differently preserved specimens

    • Implement calibrated spike-in controls to account for preservation artifacts

These advanced techniques have revolutionized epigenetic analysis of rare clinical samples, enabling new insights into disease-specific chromatin states that were previously inaccessible due to material limitations.

How can Histone H3 monoclonal antibodies be applied in multiplexed epigenomic profiling?

Multiplexed approaches enable simultaneous analysis of multiple histone modifications, providing comprehensive epigenomic insights:

  • Sequential ChIP (Re-ChIP):

    • Perform initial ChIP with one Histone H3 antibody, then re-immunoprecipitate with a second antibody

    • Identifies genomic regions with co-occurring modifications (e.g., H3K4me3 and H3K27ac)

    • Requires careful optimization of elution conditions between immunoprecipitations

  • Co-ChIP Methods:

    • Use antibodies with different host species for simultaneous immunoprecipitation

    • Separate the bound complexes using species-specific secondary antibodies

    • Enables analysis of mutually exclusive marks from the same sample

  • Barcoded Antibody Approaches:

    • Conjugate different DNA barcodes to Histone H3 monoclonal antibodies

    • Perform parallel ChIP reactions followed by pooled sequencing

    • Demultiplex based on barcode sequences during data analysis

  • Mass Cytometry (CyTOF) Applications:

    • Label Histone H3 antibodies with different metal isotopes

    • Analyze dozens of histone modifications simultaneously at single-cell resolution

    • Correlate with cellular phenotypic markers for comprehensive profiling

  • Imaging-Based Multiplexing:

    • Use iterative antibody staining and stripping for sequential imaging

    • Apply spectral unmixing for simultaneous detection of multiple modifications

    • Implement DNA-Exchange imaging for highly multiplexed detection

  • Combinatorial Indexing Strategies:

    • Combine cell/nucleus barcoding with modification-specific antibodies

    • Enable thousands of single-cell epigenomic profiles in parallel

    • Integrate with transcriptomic profiling for multi-modal analysis

  • Computational Integration:

    • Develop algorithms to infer combinatorial modification patterns from individual ChIP-Seq datasets

    • Use machine learning approaches to predict modification co-occurrence

    • Integrate multiple epigenomic datasets to construct comprehensive regulatory networks

For optimal results, monoclonal antibodies should be carefully validated for specificity and performance in multiplexed settings, as some antibodies may show altered binding characteristics when used in combination .

What metrics should be used to evaluate batch-to-batch consistency of Histone H3 monoclonal antibodies?

Ensuring consistency between antibody batches is critical for reproducible epigenetic research:

  • Western Blot Quantification:

    • Compare signal intensity at the target molecular weight (15-17 kDa)

    • Assess background binding patterns across multiple dilutions

    • Calculate signal-to-noise ratios using densitometry

  • Immunofluorescence Pattern Analysis:

    • Evaluate nuclear localization pattern and intensity

    • Quantify coefficient of variation in nuclear staining across cells

    • Compare distribution patterns relative to DAPI or other nuclear markers

  • ChIP-qPCR Benchmarking:

    • Test enrichment at 5-10 well-characterized genomic regions

    • Compare percent input values between batches

    • Calculate enrichment relative to negative control regions

  • Epitope Binding Affinity:

    • Measure antibody affinity using surface plasmon resonance

    • Determine EC50 values in peptide ELISA assays

    • Compare titration curves between antibody lots

  • Cross-Reactivity Assessment:

    • Test against a panel of related histone peptides

    • Evaluate species cross-reactivity consistency

    • Confirm specificity for particular modifications when applicable

  • Reproducibility Metrics:

    • Calculate intra-lot and inter-lot coefficients of variation

    • Perform correlation analysis between ChIP-Seq datasets from different lots

    • Assess peak overlap statistics and peak intensity correlations

  • Standard Reference Materials:

    • Use recombinant histone standards and defined cell lines as references

    • Include archived positive control samples from previous successful experiments

    • Consider creating internal reference standards for long-term studies

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 2024 Thebiotek. All Rights Reserved.