HIST1H3A (Ab-11) Antibody

Shipped with Ice Packs
In Stock

Description

Chromatin Dynamics

  • Detects Histone H3.1 in nucleosome assembly during DNA replication .

  • Differentiates H3.1 (replication-dependent) from H3.3 (replication-independent) .

Epigenetic Regulation

  • Identifies post-translational modifications (PTMs) like acetylation (K56) and methylation (K9, K27) .

    • Example: Acetylation at K56 (H3K56ac) correlates with transcriptional activation .

    • Cross-Talk: Phosphorylation at Ser-11 (H3S10ph) inhibits K9 methylation, enabling acetylation .

Disease Research

  • Aberrant HIST1H3A expression is linked to pediatric acute myeloid leukemia (t(7;12) translocation) .

  • Overexpression in cancers disrupts chromatin stability, promoting oncogenesis .

Western Blot

  • Lane Specificity: Detects a single band at ~17 kDa in human, mouse, and rat lysates .

  • Blocking Controls: Specificity confirmed via peptide blocking assays .

Immunoprecipitation

  • Efficiently pulls down Histone H3 from HeLa cell extracts, validated by SDS-PAGE .

Immunofluorescence

  • Localizes to nuclei in fixed cells, with signal intensity correlating with cell cycle phase .

Key Research Findings

  1. Cell Cycle Dependency: HIST1H3A incorporation into chromatin peaks during S-phase .

  2. Modification Crosstalk:

    • Phosphorylation at Thr-4 (H3T3ph) by HASPIN regulates mitotic progression .

    • Acetylation at K56 enhances DNA repair efficiency .

  3. Disease Link: HIST1H3A mutations disrupt histone methylation patterns, implicated in gliomas .

Technical Considerations

  • Fixation: Compatible with methanol-fixed cells but requires antigen retrieval for formalin-fixed samples .

  • Buffer Optimization: High-salt nuclear extraction protocols recommended for WB .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
Histone H3 is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, restricting DNA accessibility to cellular machinery that utilizes DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex system of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer is mediated by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course may be helpful in determining whether the tumors are heterochronous. PMID: 29482987
  3. Studies have revealed that JMJD5, a Jumonji C (JmjC) domain-containing protein, functions as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Data suggests that the Ki-67 antigen proliferative index has notable limitations, and phosphohistone H3 (PHH3) may serve as an alternative proliferative marker. PMID: 29040195
  5. These findings indicate that cytokine-induced histone 3 lysine 27 trimethylation is a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data suggests that in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. Among a series of 47 diffuse midline gliomas, histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with BRAF-V600E mutation and was frequently associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research has shown that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral DNA, and deposits histone H3.3 onto it. PMID: 28981850
  9. Experiments have demonstrated that PHF13 specifically binds to DNA and two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl), acting as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This study provides the first description of the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3). This mutation is correlated with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Research indicates that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest, while the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggests that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail. This could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. These results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate the formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications have been observed in leukocytes exposed to traffic-derived airborne particulate matter. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. Histone 3 mutations do not appear to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggests that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. The functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. The authors have verified that histone H3 is a real substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. Circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Double mutations on the residues in the interface (L325A/D328A) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Minichromosome maintenance protein 2 (MCM2) binding is not required for the incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a crucial role in the maintenance of leukemia stem cells (LSCs). PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Lower-resolution mass spectrometry instruments can be utilized for the analysis of histone post-translational modifications (PTMs). PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. De novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What are the validated applications for HIST1H3A (Ab-11) antibody?

HIST1H3A antibodies have been validated for multiple experimental applications with specific optimal dilution ranges. These include Western Blot (WB) at 1:5000-1:50000 dilution, Immunoprecipitation (IP) using 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate, Immunohistochemistry (IHC) at 1:500-1:2000 dilution, Immunofluorescence (IF/ICC) at 1:500-1:2000 dilution, and Flow Cytometry (FC) using 0.40 μg per 10^6 cells in a 100 μl suspension . The antibody has been extensively validated in published literature, with at least 42 publications confirming its efficacy in Western blot applications and 2 publications for immunofluorescence applications .

Which cell lines and tissue samples have been validated for HIST1H3A antibody reactivity?

The HIST1H3A antibody has demonstrated positive Western blot detection in numerous cell lines including LNCaP, HEK-293, Jurkat, K-562, HSC-T6, NIH/3T3, MDCK, CHO, and HeLa cells . It has also shown reactivity with various tissue samples including chicken brain tissue, zebrafish, and wheat whole plant . The antibody has been confirmed to work in immunoprecipitation specifically with HeLa cells, and in immunohistochemistry with mouse testis tissue (with recommended antigen retrieval using TE buffer pH 9.0 or citrate buffer pH 6.0) . For immunofluorescence applications, it has been validated in MCF-7, A431, and HeLa cells .

What is the molecular weight of HIST1H3A and how does this impact Western blot interpretation?

The observed molecular weight of HIST1H3A is approximately 15 kDa . This information is critical for accurate interpretation of Western blot results. When working with histones, it's important to note that their small size requires appropriate gel concentration selection (typically 15-18% acrylamide gels) for optimal resolution. Additionally, researchers should be aware that histone proteins may demonstrate slight variations in migration patterns due to post-translational modifications. Therefore, when analyzing Western blot results, bands slightly above or below the expected 15 kDa mark should be carefully evaluated in the context of potential modifications rather than immediately dismissed as non-specific binding.

How should sample preparation be optimized for histone research using HIST1H3A antibodies?

For optimal detection of HIST1H3A, researchers should implement specialized sample preparation protocols that preserve histone integrity and post-translational modifications. Acid extraction using 0.2N HCl or 0.4N H2SO4 is recommended for isolating histones from nuclei. This approach helps to separate histones from DNA and other nuclear proteins, resulting in enriched histone preparations.

For tissue samples, a recommended protocol includes:

  • Homogenizing tissue in nuclear isolation buffer (15mM Tris-HCl pH 7.5, 60mM KCl, 15mM NaCl, 5mM MgCl2, 1mM CaCl2, 250mM sucrose) with protease and phosphatase inhibitors

  • Filtering the homogenate through a 100μm cell strainer

  • Centrifuging at 1000g for 10 minutes to isolate nuclei

  • Resuspending the nuclear pellet in 0.2N HCl and incubating on ice for 30 minutes

  • Centrifuging at 16,000g for 10 minutes

  • Neutralizing the supernatant containing histones with 1/10 volume of 2M NaOH

This method ensures optimal preservation of histone modifications which is crucial when studying epigenetic markers. For antibody applications, buffers should be supplemented with deacetylase inhibitors (e.g., sodium butyrate) and phosphatase inhibitors to maintain modification states.

What controls are essential when using HIST1H3A antibodies in epigenetic research?

When designing experiments with HIST1H3A antibodies, several critical controls should be included:

  • Positive Controls: Include cell lines with known HIST1H3A expression patterns such as HeLa, HEK-293, or Jurkat cells .

  • Negative Controls:

    • Primary antibody omission controls

    • Isotype controls using mouse IgG2b (matching the HIST1H3A antibody isotype)

    • Where possible, HIST1H3A-depleted samples via siRNA knockdown

  • Peptide Competition Assays: Pre-incubating the antibody with the immunizing peptide before application to verify specificity.

  • Cross-reactivity Controls: Since HIST1H3A shares sequence homology with other histone H3 variants, researchers should include tests for cross-reactivity with H3.3 or other variants when specificity is crucial.

  • Modified Histone Controls: When studying specific histone modifications, include controls for different modification states (e.g., methylated vs. acetylated histones) to ensure the antibody detection is not affected by post-translational modifications at or near the epitope.

These controls help validate findings and ensure that observed signals are specific to HIST1H3A rather than artifacts or cross-reactive signals.

How do different fixation methods affect HIST1H3A epitope accessibility in immunostaining applications?

Fixation methods significantly impact HIST1H3A epitope accessibility, particularly for applications like immunohistochemistry and immunofluorescence. The choice of fixation protocol can affect both signal intensity and specificity:

  • Paraformaldehyde (PFA) Fixation: While commonly used (4% PFA for 10-15 minutes), this method can mask some histone epitopes due to extensive protein crosslinking. For HIST1H3A detection, shorter fixation times (5-10 minutes) are often preferable.

  • Methanol Fixation: Ice-cold methanol (100%) for 10 minutes provides good nuclear architecture preservation while maintaining accessibility to many histone epitopes. This method is often superior for detecting nuclear proteins like HIST1H3A.

  • Hybrid Protocols: A sequential approach using a brief PFA fixation (3-5 minutes) followed by methanol permeabilization often provides optimal results for maintaining both cellular morphology and epitope accessibility.

For antigen retrieval in tissue sections, HIST1H3A antibody performance is optimized using TE buffer at pH 9.0, although citrate buffer at pH 6.0 can also be effective . When working with formalin-fixed paraffin-embedded (FFPE) samples, heat-induced epitope retrieval is critical, with optimal conditions typically involving 20 minutes of heating in retrieval buffer.

How can HIST1H3A antibodies be utilized to study the functional impact of histone H3 mutations in cancer research?

Recent research has identified specific mutations in histone variants, particularly H3.3, in a high proportion of pediatric brain cancers . While these studies focused on H3.3 mutations (encoded by H3F3A), similar methodological approaches can be applied to investigate potential HIST1H3A mutations.

For studying histone H3 mutations in cancer research using HIST1H3A antibodies, researchers can employ these methodological approaches:

  • Mutation-Specific Antibodies: When available, use antibodies that specifically recognize mutated histones (such as K27M or G34R/V mutations). When using general HIST1H3A antibodies, complement with sequencing data to correlate antibody signals with mutation status.

  • ChIP-Seq Analysis: Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using HIST1H3A antibodies can map the genomic distribution of wild-type versus mutant histones, revealing altered binding patterns that may drive oncogenic processes.

  • Sequential ChIP (Re-ChIP): This technique allows researchers to determine whether mutant histones co-localize with specific histone modifications by performing successive immunoprecipitations with different antibodies (e.g., first with mutation-specific antibodies, then with modification-specific antibodies).

  • Proximity Ligation Assays (PLA): This method can detect interactions between mutant histones and chromatin-modifying enzymes in situ, helping to understand how mutations disrupt normal epigenetic regulation.

  • Mass Spectrometry: Following immunoprecipitation with HIST1H3A antibodies, mass spectrometry can identify post-translational modification patterns on wild-type versus mutant histones.

It's important to note that approximately 70-80% of pediatric gliomas are characterized by specific histone mutations, primarily in H3.3 (H3F3A) but also in H3.1 (HIST1H3B) . These mutations occur heterozygously, with one wild-type allele remaining. This understanding is crucial for interpreting experimental results involving histone antibodies in cancer samples.

What methodological approaches can resolve contradictory results when studying HIST1H3A post-translational modifications?

When faced with contradictory results in HIST1H3A post-translational modification (PTM) studies, researchers should implement a multi-faceted verification approach:

  • Antibody Validation Protocol:

    • Perform peptide competition assays with both modified and unmodified peptides

    • Compare results from at least two antibodies recognizing different epitopes of the same modification

    • Validate using samples with known modification status (e.g., after treatment with HDAC inhibitors for acetylation studies)

  • Technical Cross-Validation:

    • Complement antibody-based methods (Western blot, ChIP) with mass spectrometry

    • Use recombinant HIST1H3A protein (with and without specific modifications) as controls

    • Apply CRISPR-based approaches to generate cells with mutation of specific modification sites

  • Biological Context Assessment:

    • Evaluate cell cycle phase-specific dynamics, as histone modifications fluctuate during the cell cycle

    • Consider the impact of neighboring modifications (modification crosstalk)

    • Account for the balance between histone H3.1 (including HIST1H3A) and variant H3.3 in the cell type being studied

  • Data Integration Approach:

    • Combine genomic approaches (ChIP-seq) with proteomic methods

    • Correlate modification patterns with functional outcomes (e.g., transcription levels)

    • Use mathematical modeling to resolve apparently contradictory temporal dynamics

Additionally, researchers should be aware that HIST1H3A is primarily expressed during S phase, with expression decreasing as cell division slows during differentiation . This expression pattern can significantly impact experimental results when comparing rapidly dividing cells to differentiated tissues.

How can HIST1H3A antibodies be used to investigate the interplay between histone variants in genomic stability?

Investigating the interplay between histone H3.1 (HIST1H3A) and other histone variants (particularly H3.3) in maintaining genomic stability requires sophisticated methodological approaches:

  • Chromatin Fractionation Studies:

    • Use HIST1H3A antibodies in combination with variant-specific antibodies to isolate and characterize chromatin regions enriched for specific histone variants

    • Analyze these fractions for DNA damage markers or repair proteins to establish correlations between variant distribution and genomic stability

  • Live-Cell Imaging with Tagged Histones:

    • Complement antibody-based approaches with live-cell imaging using fluorescently tagged histone variants

    • Track the recruitment of DNA repair machinery to sites containing specific histone variants

  • Nascent Chromatin Capture:

    • Use HIST1H3A antibodies in nascent chromatin capture experiments to study the incorporation of H3.1 during DNA replication

    • Compare with H3.3 incorporation patterns, which occur in a replication-independent manner

  • Centromere and Telomere Stability Assessment:

    • Use co-immunoprecipitation of HIST1H3A with centromeric proteins like CENP-A

    • Study potential compensatory mechanisms between H3.1 and H3.3 at pericentromeric and telomeric regions

Research has demonstrated that partial loss of H3.3 function results in ectopic CENP-A foci formation, suggesting a compensatory gap-filling mechanism . Similar approaches can be applied to study the role of HIST1H3A in maintaining genomic stability. Studies in model organisms have shown that disruption of proper histone variant distribution leads to chromosomal abnormalities, aneuploidy, and defects in chromosome segregation .

How do HIST1H3A and H3.3 differ in their chromatin assembly pathways and what methodologies can detect these differences?

HIST1H3A (H3.1) and H3.3 utilize distinct chromatin assembly pathways that can be studied using different methodological approaches:

  • Chaperone-Specific Analysis:

    • HIST1H3A (H3.1) is incorporated into chromatin through the CAF-1 (Chromatin Assembly Factor 1) complex in a replication-dependent manner

    • H3.3 is incorporated via two separate pathways: the HIRA complex for genic/euchromatic regions (replication-independent) and the DAXX/ATRX complex for pericentromeric and telomeric heterochromatin

To investigate these differences, researchers can employ:

  • Co-Immunoprecipitation Studies:

    • Use HIST1H3A antibodies to pull down associated chaperone complexes

    • Compare with H3.3 immunoprecipitation to identify differential chaperone associations

  • Proximity Ligation Assays:

    • Visualize the in situ interaction between HIST1H3A and CAF-1 components

    • Compare with H3.3 interactions with HIRA or DAXX/ATRX

  • Cell Cycle-Specific Analysis:

    • Synchronize cells at different cell cycle stages and analyze HIST1H3A incorporation patterns

    • Compare with the replication-independent incorporation of H3.3

  • Pulse-Chase Experiments:

    • Use tagged histones or metabolic labeling to track the incorporation kinetics of newly synthesized HIST1H3A versus H3.3

    • Analyze incorporation patterns following replication inhibition (which should primarily affect HIST1H3A but not H3.3 incorporation)

Understanding these differential assembly pathways is crucial for interpreting experimental results, particularly in studies involving cell cycle perturbations or DNA damage responses. Research has shown that disruption of HIRA, the major chaperone for H3.3 deposition, causes defects in early embryogenesis, while loss of ATRX results in aneuploidy and defects in chromosomal segregation .

What is the significance of HIST1H3A post-translational modifications in transcriptional regulation?

HIST1H3A post-translational modifications (PTMs) establish a complex "histone code" that regulates gene expression through various mechanisms:

  • Key Regulatory Modifications:

    • H3K4 methylation: Associated with active transcription, particularly H3K4me3 at promoters

    • H3K27 methylation: H3K27me3 is associated with gene silencing and is catalyzed by the Polycomb Repressive Complex 2 (PRC2)

    • H3K9 methylation: H3K9me3 is associated with heterochromatin formation

    • H3K27 acetylation: Counteracts the repressive H3K27me3 mark and is associated with active enhancers

  • Methodological Approaches:

    • ChIP-seq Analysis: Use modification-specific antibodies in combination with HIST1H3A antibodies to map the genomic distribution of specific modifications

    • Sequential ChIP: Determine co-occurrence of different modifications on the same histone molecule

    • Mass Spectrometry: Identify combinations of modifications on individual histone molecules after immunoprecipitation with HIST1H3A antibodies

  • Functional Impact Assessment:

    • Reporter Assays: Assess the impact of specific HIST1H3A modifications on gene expression using reporter constructs

    • CRISPR-based Approaches: Generate mutations at specific modification sites to determine their functional importance

    • Inhibitor Studies: Use small molecule inhibitors of histone-modifying enzymes to manipulate specific modifications

What are the most common technical challenges when using HIST1H3A antibodies in ChIP applications and how can they be addressed?

Chromatin immunoprecipitation (ChIP) with HIST1H3A antibodies presents several technical challenges that require specific methodological solutions:

  • Cross-Reactivity with Other H3 Variants:

    • Challenge: HIST1H3A (H3.1) shares high sequence similarity with other H3 variants, making specific detection challenging.

    • Solution: Validate antibody specificity using recombinant H3.1 and H3.3 proteins. For variant-specific ChIP, consider using epitope-tagged H3.1 in cell models or complementing antibody-based approaches with mass spectrometry validation.

  • Epitope Masking by Post-Translational Modifications:

    • Challenge: PTMs near the antibody epitope can interfere with antibody binding.

    • Solution: Use antibodies raised against unmodified peptides or select antibodies whose epitopes are in regions less prone to modifications. Alternatively, use multiple antibodies recognizing different epitopes of HIST1H3A.

  • Sonication Efficiency and Chromatin Fragmentation:

    • Challenge: Insufficient chromatin fragmentation can lead to high background and poor resolution.

    • Solution: Optimize sonication conditions for each cell type. Aim for fragments between 200-500 bp for standard ChIP and 100-300 bp for ChIP-seq. Verify fragmentation by agarose gel electrophoresis before proceeding.

  • Fixation Parameters:

    • Challenge: Excessive crosslinking can mask epitopes, while insufficient crosslinking may not preserve protein-DNA interactions.

    • Solution: Optimize formaldehyde concentration (typically 0.75-1% for histones) and fixation time (typically 10 minutes for histones). Consider dual crosslinking with disuccinimidyl glutarate (DSG) followed by formaldehyde for improved capture of protein-protein interactions.

  • Quantification and Normalization:

    • Challenge: Determining appropriate normalization methods for ChIP-qPCR or ChIP-seq data.

    • Solution: For ChIP-qPCR, use multiple normalization strategies, including percent input, IgG control normalization, and normalization to a housekeeping gene region. For ChIP-seq, include spike-in controls with exogenous chromatin from another species.

A refined protocol for HIST1H3A ChIP includes:

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

  • Quench with 125 mM glycine for 5 minutes

  • Isolate nuclei and sonicate to generate 200-500 bp fragments

  • Pre-clear chromatin with protein A/G beads

  • Incubate with HIST1H3A antibody (3-5 μg) overnight at 4°C

  • Capture antibody-chromatin complexes with protein A/G beads

  • Perform stringent washes to reduce background

  • Reverse crosslinks and purify DNA for analysis

How can researchers ensure specificity when studying HIST1H3A versus other histone H3 variants?

Ensuring specificity when studying HIST1H3A versus other histone H3 variants requires a strategic methodological approach:

  • Variant-Specific Region Targeting:

    • HIST1H3A (H3.1) and H3.3 differ at only five amino acid positions (positions 31, 87, 89, 90, and 96)

    • Select antibodies that target these variant-specific regions or verify that the antibody epitope includes these distinctive residues

  • Combinatorial Approach:

    • Genetic Manipulation: Use CRISPR/Cas9 to tag endogenous HIST1H3A with a small epitope tag

    • Expression Analysis: Correlate protein detection with mRNA expression data (H3.1 is primarily expressed during S phase )

    • Cell Cycle Synchronization: Analyze synchronized cell populations, as HIST1H3A is predominantly expressed during S phase

  • Mass Spectrometry Validation:

    • Immunoprecipitate histones with the antibody

    • Analyze by mass spectrometry to confirm variant identity

    • Quantify the ratio of HIST1H3A to other H3 variants in the immunoprecipitated material

  • Recombinant Protein Controls:

    • Use recombinant HIST1H3A and H3.3 proteins to determine antibody specificity by Western blot

    • Perform peptide competition assays with variant-specific peptides

  • Functional Approaches:

    • Correlate antibody signals with replication timing (HIST1H3A incorporation is replication-dependent)

    • Use nascent DNA labeling (e.g., EdU) to identify replication sites and correlate with HIST1H3A signal

When publishing results, researchers should explicitly state which histone H3 variant they are studying and provide evidence for the specificity of their detection methods. This is particularly important given the different functional roles of H3.1 and H3.3 in chromatin regulation and their distinct patterns of post-translational modifications.

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.