HIST1H3A (Ab-10) Antibody

Shipped with Ice Packs
In Stock

Description

Target and Structure

Target Protein: HIST1H3A (UniProt ID: P68431), a core component of nucleosomes that plays a critical role in chromatin structure and gene regulation .
Epitope: Peptide sequence around Ser10 of human histone H3.1 .
Immunogen: Synthetic peptide derived from human histone H3.1, ensuring specificity for the unmodified or post-translationally modified Ser10 region .

Immunogen DetailsValue
SpeciesHuman
Modification SiteSerine 10 (Ser10)
Sequence OriginHistone H3.1 (HIST1H3A)

Host Species and Clonality

  • Host Species: Rabbit .

  • Clonality: Polyclonal, enabling recognition of multiple epitopes within the target region .

Applications and Performance

Validated applications include:

ApplicationDilution RangeValidated Cell Lines/Tissues
Western Blot1:500–5,000 , 1:100–1,000 Jurkat, HEK293, Hela, HepG2
IHC (Paraffin)1:20–200 , 1:1–100 Human colon, lung, prostate cancer, glioma
IF/ICC1:50–200 Hela cells
ChIP5 µg per assay Chromatin from Hela cells

Key Findings:

  • ChIP Validation: Demonstrated specificity in chromatin immunoprecipitation assays using Hela cells, with robust enrichment compared to control IgG .

  • Cancer Research: Successfully detected histone H3.1 in paraffin-embedded human cancer tissues, including colon, lung, and prostate carcinomas .

  • Cross-Reactivity: No reported cross-reactivity with non-histone proteins, though optimal performance requires validation in user-specific conditions .

Specificity and Validation

  • Specificity: Confirmed via peptide competition assays and reactivity across human, mouse, and rat samples .

  • Western Blot: Detects a single band at ~17 kDa corresponding to histone H3 in acid-extracted cell lysates .

  • IF/ICC: Localizes to the nucleus in Hela cells, consistent with histone function .

Research Limitations

  • Exclusively for Research: Not approved for diagnostic, therapeutic, or clinical use .

  • Batch Variability: As with all polyclonal antibodies, performance may vary between lots; independent validation is recommended .

Comparative Insights from Literature

  • Antibody Validation: Peer-reviewed studies emphasize the importance of rigorous validation for histone antibodies. The HIST1H3A (Ab-10) Antibody aligns with ENCODE standards for specificity in ChIP and WB .

  • Contextual Caveats: While some histone modification antibodies exhibit cross-reactivity (e.g., H3K27me3 vs. H3K4me3) , HIST1H3A (Ab-10) targets unmodified H3.1, reducing such risks .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery time estimates.
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 play a crucial role in packaging and compacting DNA into chromatin, which limits accessibility of DNA to cellular machinery requiring it as a template. Histones, therefore, play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated by a complex interplay of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies have elucidated the mechanism of epigenetic regulation in cancer through the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course can be a useful indicator for determining the heterochronous nature of tumors. PMID: 29482987
  3. Research has demonstrated that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions inducing a DNA damage response. PMID: 28982940
  4. Data suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) is a viable alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, histone H3-K27M mutation was found to be mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with BRAF-V600E mutation and was commonly 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 and deposits histone H3.3 onto these. PMID: 28981850
  9. Experiments have demonstrated that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl) where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This study provides, for the first time, a detailed 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) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. The presence of H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest whereas the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. Conversely, histone 3 mutations do not seem to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis and repression of APAK. PMID: 25961932
  27. Taken together, the authors verified that histone H3 is a real substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) serves a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Data indicate that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays is 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 is HIST1H3A and what role does it play in cellular processes?

HIST1H3A encodes Histone H3.1, one of the core histone proteins involved in chromatin structure in eukaryotic cells. Nucleosomes, which consist of DNA wrapped around histone octamers, compact DNA into chromatin, limiting DNA accessibility to cellular machinery. Histone H3.1 plays a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability . Unlike the constitutively expressed H3.3, H3.1 is primarily expressed during the S-phase of the cell cycle, making it particularly important during DNA replication .

How does HIST1H3A differ from other histone H3 variants?

In humans, there are several histone H3 variants, each with distinct expression patterns and functions. H3.1 (encoded by HIST1H3A and other genes) and H3.2 are expressed primarily during S-phase and are incorporated into chromatin during DNA replication. In contrast, H3.3 is expressed constitutively throughout the cell cycle . Some H3 variants have tissue-restricted expression patterns (H3.5, H3.X, H3.Y), while others like CENP-A are deposited only at centromeres . These differences in expression timing and patterns contribute to their specialized functions in chromatin regulation.

What post-translational modifications occur on HIST1H3A?

Histone H3.1 undergoes extensive post-translational modifications, particularly on its N-terminal tail that protrudes from the globular nucleosome core . These modifications include:

  • Acetylation (at various lysine residues including K23, K27)

  • Methylation (at lysine residues including K4, K27, K36)

  • Phosphorylation (at serine residues including S10)

  • Other modifications including ubiquitination and SUMOylation

These modifications constitute part of the "histone code" that regulates DNA accessibility and recruits specific protein complexes to chromatin .

What is the significance of HIST1H3A as a redox sensor?

Recent research has identified histone H3.1 as a chromatin-embedded redox sensor. According to a 2024 study, H3.1 can sense changes in hydrogen peroxide levels within the nucleus, with Cysteine 96 (Cys96) playing a crucial role in this sensing mechanism . When oxidized, H3.1 undergoes changes that lead to its replacement, potentially with H3.3, resulting in chromatin decompaction and transcriptional activation . This function connects cellular redox state directly to chromatin structure and gene expression regulation.

What types of HIST1H3A antibodies are available for research?

Researchers can choose from several types of HIST1H3A antibodies:

  • Polyclonal antibodies: Derived from multiple B cell lineages, recognizing multiple epitopes on H3.1

  • Monoclonal antibodies: Derived from a single B cell lineage, recognizing a single epitope (e.g., clones 27F2, 6F6)

  • Modification-specific antibodies: Recognizing specific post-translational modifications on H3.1, such as acetylation at Lys23 (acLys23), acetylation at Lys27 (acLys27), di-methylation at Lys4 (H3K4me2), di-methylation at Lys27 (H3K27me2), and tri-methylation at various lysine residues

How should I validate the specificity of a HIST1H3A antibody?

Antibody validation is crucial for reliable research results. Consider these approaches:

  • Western blot analysis with positive and negative controls to confirm band size (approximately 15.4 kDa for Histone H3)

  • Peptide competition assays to verify epitope specificity

  • Knockout/knockdown experiments to confirm antibody specificity

  • Cross-reactivity tests against other histone variants, particularly H3.2 and H3.3, which have high sequence homology with H3.1

  • Dot blot or ELISA testing with modified and unmodified peptides for modification-specific antibodies

What considerations should guide antibody selection for specific applications?

When selecting a HIST1H3A antibody, consider:

  • The specific post-translational modification you're studying (if applicable)

  • Application compatibility - check if the antibody has been validated for your application (Western blot, ChIP, immunofluorescence, etc.)

  • Species reactivity - ensure the antibody recognizes HIST1H3A in your experimental organism

  • Clonality - polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variation; monoclonal antibodies provide consistent specificity but may be more sensitive to epitope masking

  • Host species - consider secondary antibody compatibility and potential cross-reactivity issues in your experimental system

What are the optimal conditions for using HIST1H3A antibodies in Western blotting?

For successful Western blotting with HIST1H3A antibodies:

  • Sample preparation: Use specialized histone extraction protocols to efficiently isolate histones from nuclear fractions

  • Loading control: Consider total H3 or other stable nuclear proteins as loading controls

  • Expected size: Look for a band at approximately 15.4 kDa corresponding to Histone H3

  • Dilution: Typical working dilutions range from 1:5000 to 1:10000, but optimize for your specific antibody

  • Blocking: Use 5% non-fat dry milk or BSA in TBST, depending on the antibody specifications

  • Membrane choice: PVDF membranes are generally preferred for histone detection

  • Transfer conditions: Use low methanol concentrations and adjust transfer time to prevent small proteins like histones from transferring through the membrane

How should HIST1H3A antibodies be optimized for ChIP applications?

For Chromatin Immunoprecipitation (ChIP) applications:

  • Crosslinking: Standard formaldehyde crosslinking (1% for 10 minutes) works for most histone ChIP experiments

  • Sonication: Optimize sonication conditions to generate chromatin fragments of 200-500 bp

  • Antibody amount: Typically use 2-5 μg of antibody per ChIP reaction

  • Positive controls: Include primers for regions known to be enriched for your specific histone modification

  • Negative controls: Include IgG antibody controls and primers for regions not expected to have your modification

  • Wash stringency: Adjust wash buffers based on antibody specificity and background levels

  • Validation: Confirm enrichment through qPCR before proceeding to sequencing

What considerations are important for immunofluorescence with HIST1H3A antibodies?

For optimal immunofluorescence results:

  • Fixation: 4% paraformaldehyde is commonly used; avoid methanol fixation which may affect some histone epitopes

  • Permeabilization: Use 0.1-0.5% Triton X-100 to access nuclear antigens

  • Antigen retrieval: May be necessary for some fixed tissues; citrate buffer (pH 6.0) is often effective

  • Blocking: 5% normal serum from the same species as the secondary antibody

  • Antibody dilution: Start with 1:500 dilution and optimize as needed

  • Controls: Include no primary antibody controls and, if possible, cells with known H3.1 modification states

  • Counterstaining: DAPI for nuclear visualization to confirm nuclear localization of H3.1 signal

How can HIST1H3A antibodies be used to study chromatin dynamics during cell cycle?

To study chromatin dynamics during the cell cycle:

  • Synchronize cells using methods appropriate for your cell type (double thymidine block, serum starvation/refeeding, etc.)

  • Collect cells at different cell cycle stages (confirmed by flow cytometry or cyclin expression)

  • Perform ChIP-seq or immunofluorescence with HIST1H3A antibodies to track H3.1 incorporation into chromatin

  • Compare with replication timing data to correlate H3.1 deposition with DNA synthesis

  • Use pulse-chase experiments with tagged histones to differentiate new deposition from existing H3.1

  • Consider dual immunofluorescence with cell cycle markers (e.g., PCNA for S-phase) to correlate H3.1 patterns with cell cycle stages

How can HIST1H3A antibodies be used to study the relationship between histone modifications and gene expression?

To investigate relationships between H3.1 modifications and gene expression:

  • Perform ChIP-seq with modification-specific H3.1 antibodies (e.g., H3K27me3, H3K4me3, acLys23)

  • Integrate with RNA-seq data from the same cell type/condition

  • Use sequential ChIP (re-ChIP) to identify genomic regions with co-occurring modifications

  • Compare ChIP-seq profiles before and after perturbations (e.g., inhibitors of histone-modifying enzymes)

  • Correlate with chromatin accessibility data (ATAC-seq, DNase-seq)

  • Perform reporter assays with mutated histone H3.1 to confirm functional relationships

  • Utilize CUT&RUN or CUT&Tag as alternative approaches for higher signal-to-noise ratio in mapping histone modifications

What approaches can be used to study HIST1H3A as a redox sensor in chromatin regulation?

Based on recent findings about H3.1's role as a redox sensor , researchers can:

  • Utilize oxidation-resistant H3.1 mutants (e.g., H3.1(C96S)) to study the impact of redox sensing on chromatin structure

  • Use DCP-Bio1 or similar reagents to label oxidized Cys-SOH residues in H3.1 following oxidative stress

  • Perform ChIP-seq before and after oxidative stress to map changes in H3.1 occupancy

  • Compare with H3.3 ChIP-seq to assess histone variant exchange during redox responses

  • Combine with transmission electron microscopy to visualize changes in chromatin compaction

  • Utilize fluorescence microscopy with nuclear size measurements to quantify chromatin decompaction

  • Perform RNA-seq to correlate transcriptional changes with histone oxidation and replacement

How can HIST1H3A antibodies be used to investigate the role of histone mutations in cancer?

To study cancer-associated histone H3 mutations:

  • Use mutation-specific antibodies (if available) or tagged mutant histones to track their incorporation

  • Perform ChIP-seq to map genome-wide distribution of mutant histones

  • Compare chromatin accessibility (ATAC-seq) in cells with wild-type versus mutant H3.1

  • Investigate changes in other histone modifications that may be affected by H3 mutations

  • Study protein interactions using immunoprecipitation followed by mass spectrometry

  • Assess impact on gene expression using RNA-seq

  • Correlate findings with cancer patient data to establish clinical relevance

What techniques can be used to study histone H3.1 turnover and dynamics?

To investigate H3.1 turnover and dynamics:

  • SNAP-tag or CLIP-tag labeling of H3.1 for pulse-chase experiments

  • FRAP (Fluorescence Recovery After Photobleaching) with fluorescently tagged H3.1

  • Stable isotope labeling with amino acids in cell culture (SILAC) combined with mass spectrometry

  • Inducible expression systems for temporal control of tagged H3.1 variants

  • ChIP-seq time courses following perturbations that affect histone deposition

  • Single-molecule tracking of tagged histones to measure diffusion rates and binding dynamics

  • Targeted degradation approaches (e.g., auxin-inducible degron system) to study rapid H3.1 depletion effects

What are common challenges when working with HIST1H3A antibodies and how can they be addressed?

Common challenges and solutions include:

  • Cross-reactivity with other H3 variants: Use highly specific antibodies or epitope tags; validate with peptide arrays

  • High background in Western blots: Increase blocking time/concentration; optimize antibody dilution; use more stringent washes

  • Poor signal in ChIP: Optimize chromatin fragmentation; increase antibody amount; ensure epitope is not masked by other modifications

  • Inconsistent results across experiments: Use lot-controlled antibodies; standardize protocols; include positive controls

  • Non-specific bands in Western blots: Use recombinant H3.1 as positive control; perform peptide competition assays

  • Epitope masking by adjacent modifications: Consider using alternative antibodies targeting different regions of H3.1

  • Poor reproducibility in ChIP-seq: Implement rigorous quality control; use spike-in controls; standardize bioinformatic pipelines

How should results from different HIST1H3A antibody applications be integrated for comprehensive analysis?

For integrative analysis:

  • Start with Western blotting to confirm antibody specificity and H3.1 modification levels

  • Perform immunofluorescence to determine cellular and subcellular localization patterns

  • Use ChIP-seq to map genome-wide distribution of H3.1 and its modifications

  • Integrate with gene expression data (RNA-seq) to correlate chromatin states with transcriptional output

  • Add chromatin accessibility data (ATAC-seq, DNase-seq) to understand functional consequences

  • Validate key findings with orthogonal approaches (e.g., CUT&RUN/CUT&Tag to complement ChIP)

  • Use computational approaches to integrate multiple datasets and identify statistically significant patterns

What controls should be included when conducting experiments with HIST1H3A antibodies?

Essential controls include:

  • Input controls for ChIP experiments to normalize for DNA abundance

  • IgG controls from the same species as the primary antibody to assess non-specific binding

  • Peptide competition controls to confirm antibody specificity

  • Positive controls: genomic regions known to be enriched for your histone modification

  • Negative controls: regions not expected to have your modification

  • Technical replicates to assess experimental variability

  • Biological replicates to account for natural biological variation

  • Treatment controls (e.g., histone modification enzyme inhibitors) to validate antibody specificity

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.