HIST1H3A (Ab-4) Antibody

Shipped with Ice Packs
In Stock

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 method of purchase 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 H3A is a core component of the nucleosome, the fundamental unit of chromatin structure. Nucleosomes play a crucial role in packaging and compacting DNA, thus limiting DNA accessibility to cellular machineries that require DNA as a template. Consequently, histones, including H3A, are central to the regulation of transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is meticulously controlled by a complex interplay of post-translational modifications of histones, often referred to as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests a mechanism for 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 aid in determining whether tumors are heterochronous. PMID: 29482987
  3. JMJD5, a Jumonji C (JmjC) domain-containing protein, has been identified as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that induce a DNA damage response. PMID: 28982940
  4. Findings indicate that the Ki-67 antigen proliferative index has significant limitations, making phosphohistone H3 (PHH3) a viable alternative proliferative marker. PMID: 29040195
  5. These results point to cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. 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. 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, rarely co-occurring with BRAF-V600E mutation. It was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral, and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments revealed 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 presents, for the first time, 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 has been shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data shows 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. Research 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, 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. 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 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 Raji cells treated by 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. Research indicates 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 suggest 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. Histone H3 lysine methylation (H3K4me3) plays a critical 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. Findings indicate that lower-resolution mass spectrometry instruments can be effectively used for histone post-translational modifications (PTMs) analysis. 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. 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 why is it important in epigenetic research?

HIST1H3A encodes one of the histone H3 family members, which are small, highly basic proteins critical for nucleosome structure in eukaryotic chromosomal fibers. Histone H3 is one of the five main histones (H2A, H2B, H3, H4, and H1) responsible for packaging DNA into chromatin. Each nucleosome consists of approximately 146 bp of DNA wrapped around an octamer composed of two copies each of the four core histones (H2A, H2B, H3, and H4) . The HIST1H3A gene is located within the large histone gene cluster on chromosome 6p22-p21.3 . Due to its fundamental role in chromatin structure and epigenetic regulation, HIST1H3A is a critical target for researchers studying transcriptional regulation, cell differentiation, and disease mechanisms.

How do I select between monoclonal and polyclonal HIST1H3A antibodies for my research?

The selection between monoclonal and polyclonal antibodies depends on your specific research application and requirements:

Polyclonal HIST1H3A antibodies (e.g., 29200-1-AP, E-AB-53536):

  • Recognize multiple epitopes on the histone H3 protein

  • Generally provide higher sensitivity due to binding to multiple epitopes

  • Best for applications where maximum detection is prioritized

  • Ideal for proteins expressed at low levels or when protein conformation may be altered

  • Example applications: Initial protein detection studies, immunoprecipitation

Monoclonal HIST1H3A antibodies (e.g., M12477-1, M12477-4):

  • Recognize a single epitope with high specificity

  • Provide more consistent results between experiments and batches

  • Better for distinguishing between closely related proteins or specific modifications

  • Preferred for quantitative applications where reproducibility is critical

  • Example applications: Detection of specific histone modifications like H3K4me2 or H3R17me1

For applications requiring detection of specific histone modifications, monoclonal antibodies targeting the precise modification site (like M12477-1 for H3K4me2) are strongly recommended.

What are the critical factors for storing and handling HIST1H3A antibodies to maintain their activity?

Proper storage and handling of HIST1H3A antibodies is essential for maintaining their reactivity and specificity:

Storage ConditionRecommendationPurpose
Long-term storage-20°CMaintains antibody stability for up to one year
Short-term/frequent use4°CConvenient for up to one month of regular use
Freeze-thaw cyclesAvoid repeated cyclesPrevents protein denaturation and loss of activity
AliquotingRecommended for antibodies in frequent useMinimizes freeze-thaw damage
Buffer conditionsPBS with 0.02% sodium azide and 50% glycerolProvides optimal stability

For antibodies containing BSA (e.g., some 20µl formulations contain 0.1% BSA), special consideration should be given to potential background issues in certain applications .

What are the optimal dilution ranges for HIST1H3A antibodies in different experimental applications?

Optimal dilutions vary by application and specific antibody. Below is a comprehensive guide based on validated antibodies:

ApplicationPolyclonal Antibody (29200-1-AP)Polyclonal Antibody (E-AB-53536)Monoclonal Antibodies (M12477 series)
Western Blot (WB)1:500-1:20001:500-1:20001:500
Immunohistochemistry (IHC)1:50-1:5001:25-1:100Validated
Immunofluorescence (IF)ValidatedNot specifiedValidated
Immunocytochemistry (ICC)Not specifiedNot specifiedValidated
Flow CytometryNot specifiedNot specifiedValidated for M12477-1
Immunoprecipitation (IP)Not specifiedNot specifiedValidated for M12477-1

It is strongly recommended to titrate these antibodies in your specific testing system to determine optimal conditions, as results can be sample-dependent . For Western blot applications, consider the cell or tissue type being analyzed, as protein expression levels may vary significantly.

What is the recommended protocol for antigen retrieval when using HIST1H3A antibodies in immunohistochemistry?

Effective antigen retrieval is critical for successful immunohistochemical detection of histone proteins, as their tight association with DNA can mask epitopes:

Recommended antigen retrieval methods:

  • Heat-mediated antigen retrieval with TE buffer (pH 9.0)

    • Primary recommendation for HIST1H3A detection in tissues such as mouse testis

  • Alternative method: Citrate buffer (pH 6.0)

    • Alternative option when TE buffer doesn't provide optimal results

  • Heat-mediated retrieval duration:

    • 20 minutes at controlled temperature

For paraffin-embedded tissues, a validated protocol involves:

  • Heat-mediated antigen retrieval in citrate buffer (pH 6, epitope retrieval solution) for 20 minutes

  • Blocking with 10% goat serum

  • Overnight incubation with primary antibody at 4°C (1μg/ml)

  • Detection using biotinylated secondary antibodies and Strepavidin-Biotin-Complex with DAB as chromogen

Different tissue types may require optimization of these conditions for optimal signal-to-noise ratio.

How can I validate the specificity of a HIST1H3A antibody for my particular experimental system?

  • Positive controls:

    • Use cell lines with known expression: HeLa, HepG2, 293T, CACO-2, NIH/3T3, RAW264.7 cells have been validated for various HIST1H3A antibodies

    • Use tissues with known expression: Mouse testis, human liver have been validated

  • Expected molecular weight verification:

    • Histone H3 should appear at approximately 15-18 kDa on Western blots

    • The observed molecular weight for HIST1H3A is typically around 15 kDa for modified forms and 18 kDa for unmodified forms

  • Peptide competition assay:

    • Pre-incubate the antibody with the immunizing peptide before application

    • Signal should be significantly reduced or eliminated if the antibody is specific

  • Knockout/knockdown validation:

    • Compare signal between wild-type and HIST1H3A-depleted samples

    • Significant signal reduction confirms specificity

  • Cross-reactivity assessment:

    • Test the antibody against related histone variants to ensure it does not cross-react

Why might I observe multiple bands or unexpected molecular weights when using HIST1H3A antibodies in Western blot?

Multiple bands or unexpected molecular weights can occur for several reasons when using histone H3 antibodies:

  • Post-translational modifications (PTMs):

    • Histone H3 undergoes numerous PTMs including methylation, acetylation, phosphorylation, and ubiquitination

    • These modifications can alter migration patterns on SDS-PAGE gels

    • Different cell types or conditions may show varying modification patterns

  • Histone variants:

    • The H3 family includes variants (H3.1, H3.2, H3.3, CENP-A) with slightly different mobility

    • Antibodies may detect multiple variants depending on epitope conservation

  • Proteolytic degradation:

    • Histone proteins are susceptible to degradation during sample preparation

    • Degradation products may appear as lower molecular weight bands

  • Technical factors affecting mobility:

    • As noted in the Elabscience antibody information: "The mobility is affected by many factors, which may cause the observed band size to be inconsistent with the expected size"

    • These factors include gel percentage, running buffer composition, and voltage

For accurate interpretation, it's important to note that the expected molecular weight for histone H3 is approximately 15-18 kDa, but the actual observed band may differ based on these factors .

What are the most common pitfalls when working with HIST1H3A antibodies and how can they be addressed?

Common PitfallPossible CausesSolutions
Weak or no signalInsufficient antibody concentration, degraded antibody, low target expressionOptimize antibody dilution, verify storage conditions, use fresh antibody, include positive controls
High backgroundNon-specific binding, excessive antibody concentration, inadequate blockingIncrease blocking time/concentration, optimize antibody dilution, add detergent to wash buffers
Inconsistent resultsAntibody batch variation, protocol inconsistencies, sample degradationUse the same antibody lot when possible, standardize protocols, ensure proper sample handling
False positivesCross-reactivity with similar histones or modified formsValidate with additional approaches (e.g., mass spectrometry), use highly specific monoclonal antibodies
Signal variability between cell typesDifferential expression or modification of H3Include appropriate cell type-specific controls, normalize to total H3 when studying modifications

For applications targeting specific modifications (like the M12477-1 antibody for H3K4me2), additional validation is critical to ensure the antibody recognizes only the intended modification state .

How do I properly interpret variation in HIST1H3A signals across different cell and tissue types?

Interpreting variation in HIST1H3A signals requires consideration of several biological and technical factors:

  • Biological variation factors:

    • Cell cycle stage: Histone modifications fluctuate throughout the cell cycle

    • Differentiation state: Stem cells vs. differentiated cells show distinct histone modification patterns

    • Tissue-specific expression: Different tissues exhibit varying levels of histone variants and modifications

    • Disease states: Pathological conditions can alter histone modification patterns

  • Normalization approaches:

    • Always normalize modification-specific signals to total H3 levels

    • Include housekeeping proteins as loading controls (though these may vary by tissue type)

    • Consider using multiple antibodies targeting different epitopes for verification

  • Validated cell/tissue types:
    The following cell and tissue types have been experimentally validated for various HIST1H3A antibodies and can serve as reference points:

    • Cell lines: HeLa, HepG2, 293T, CACO-2, PC-12, C6, RAW264.7, NIH/3T3, HUVEC

    • Tissues: Mouse testis, human liver, human esophagus cancer

When comparing signals across different samples, it's essential to maintain consistent experimental conditions including sample preparation, antibody concentrations, and detection methods.

How can I optimize ChIP protocols when using HIST1H3A antibodies to study genome-wide binding patterns?

Optimizing ChIP (Chromatin Immunoprecipitation) protocols with HIST1H3A antibodies requires attention to several critical parameters:

For ChIP-seq applications, additional considerations include library preparation methods that work well with limited material, as histone modification ChIPs may yield lower amounts of DNA than transcription factor ChIPs.

What are the best approaches for studying the dynamics of histone H3 modifications in response to cellular stimuli?

Studying dynamic changes in histone H3 modifications requires temporal resolution and quantitative approaches:

  • Time-course experimental design:

    • Establish appropriate time points based on the kinetics of your cellular response

    • Include both early (minutes to hours) and late (hours to days) time points

    • Maintain synchronized cell populations when possible

  • Quantitative detection methods:

    • Western blot with fluorescent secondary antibodies for linear quantification

    • ELISA-based approaches for high-throughput quantification

    • Mass spectrometry for unbiased profiling of multiple modifications simultaneously

  • Single-cell approaches:

    • Flow cytometry using HIST1H3A modification-specific antibodies (e.g., M12477-1) can reveal population heterogeneity

    • Mass cytometry (CyTOF) allows simultaneous detection of multiple histone modifications

    • Imaging approaches with fluorescently-labeled antibodies for spatial information

  • Genome-wide temporal dynamics:

    • ChIP-seq at multiple time points to track genome-wide redistribution

    • CUT&RUN or CUT&Tag for higher sensitivity with lower cell numbers

    • Integration with transcriptomics data to correlate modification changes with gene expression

When studying rapid changes in histone modifications, it's critical to rapidly halt cellular processes during sample collection. This can be achieved by direct addition of fixatives to culture media or rapid cooling to prevent artifactual changes during processing.

How can HIST1H3A antibodies be used to investigate the relationship between histone modifications and disease progression?

HIST1H3A antibodies are powerful tools for investigating the epigenetic basis of disease:

  • Cancer research applications:

    • Compare histone modification patterns between normal and tumor tissues

    • Track changes in global and gene-specific histone modifications during cancer progression

    • Correlate modification patterns with clinical outcomes

    • Validated in cancer models: Human liver cancer, human esophagus cancer tissues have been successfully used with HIST1H3A antibodies

  • Neurodegenerative disease research:

    • Investigate histone modification changes in models of neurodegeneration

    • Compare patterns in affected vs. unaffected brain regions

    • Study the impact of disease-associated mutations on histone modification landscapes

  • Inflammatory and autoimmune conditions:

    • Monitor dynamic changes in histone modifications during inflammatory responses

    • Study the effect of anti-inflammatory treatments on the epigenome

    • Investigate the role of environmental factors in altering histone modification patterns

  • Therapeutic response monitoring:

    • Assess the impact of epigenetic drugs (HDAC inhibitors, HMT inhibitors) on histone modification profiles

    • Use modification-specific antibodies (like M12477-1 for H3K4me2) to monitor target engagement

    • Identify predictive biomarkers for treatment response based on baseline histone modification patterns

  • Methodological considerations for disease studies:

    • Always include appropriate disease-stage matched controls

    • Consider cell type heterogeneity in complex tissues

    • Normalize to total H3 levels, which may themselves change in disease contexts

    • Combine global approaches (Western blot) with locus-specific methods (ChIP-qPCR) for comprehensive analysis

By carefully selecting the appropriate antibodies and experimental approaches, researchers can gain valuable insights into the epigenetic mechanisms underlying disease pathogenesis and identify potential therapeutic targets.

How can I effectively combine HIST1H3A antibody-based approaches with other epigenetic techniques for comprehensive analysis?

Creating an integrated epigenetic analysis framework provides deeper insights than single-method approaches:

  • Multi-omics integration strategies:

    • Combine ChIP-seq using HIST1H3A modification-specific antibodies with:

      • RNA-seq to correlate histone modifications with gene expression

      • ATAC-seq to relate chromatin accessibility to histone modification states

      • DNA methylation analysis (WGBS, RRBS) to examine the interplay between histone marks and DNA methylation

      • Proteomics to identify readers, writers, and erasers of histone modifications

  • Sequential ChIP (Re-ChIP) approaches:

    • Use two different HIST1H3A modification antibodies sequentially to identify genomic regions carrying both modifications

    • Example: First immunoprecipitate with H3K4me2 antibody (M12477-1) , then with H3R17me1 antibody (M12477-4)

  • Functional validation techniques:

    • Combine antibody-based detection with targeted epigenome editing (CRISPR-dCas9 fused to epigenetic modifiers)

    • Correlate modification changes with functional outcomes in reporter assays

    • Manipulate writers/erasers of specific modifications and monitor effects using HIST1H3A antibodies

  • Computational integration:

    • Develop computational pipelines to integrate ChIP-seq data from multiple histone marks

    • Use machine learning approaches to identify combinatorial patterns predictive of gene regulation

    • Integrate with public databases like ENCODE and Roadmap Epigenomics

When designing integrated approaches, consider the technical limitations of each method and confirm key findings using orthogonal techniques whenever possible.

What controls and normalization strategies are essential when quantifying histone modifications using HIST1H3A antibodies?

Proper controls and normalization are critical for accurate quantification of histone modifications:

Control/Normalization TypePurposeImplementation
Input controlAccounts for biases in chromatin preparationProcess a portion of pre-immunoprecipitation material through all steps except immunoprecipitation
IgG controlMeasures non-specific bindingPerform parallel immunoprecipitation with matched IgG isotype
Total H3 normalizationAccounts for differences in nucleosome occupancyNormalize modification-specific signal to total H3 signal from parallel experiment
Spike-in normalizationControls for technical variation between samplesAdd known amount of exogenous chromatin (e.g., Drosophila) to each sample before processing
Housekeeping gene normalizationVerifies ChIP efficiencyQuantify recovery of regions with known modification status
Peptide competitionConfirms antibody specificityCompare signal with and without pre-incubation with immunizing peptide
Biological replicatesAccounts for biological variabilityPerform minimum of 3 independent biological replicates
Technical replicatesMeasures technical variabilityPerform duplicate or triplicate measurements of each biological sample

For Western blot quantification, additional considerations include:

  • Using validated loading controls (though standard loading controls may not be appropriate for all experimental conditions)

  • Ensuring detection is in the linear range of the assay

  • Using fluorescent secondary antibodies rather than chemiluminescence for more accurate quantification

How should I approach cross-species comparisons when using HIST1H3A antibodies?

When conducting cross-species studies with HIST1H3A antibodies, consider these important factors:

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.