Phospho-HIST1H3A (T80) 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 the products within 1-3 business days after receiving your order. Delivery times 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 H3A is a core component of nucleosomes. Nucleosomes are responsible for wrapping and compacting DNA into chromatin, restricting DNA accessibility to the cellular machinery that requires DNA as a template. Therefore, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests a mechanism for epigenetic regulation in cancer through 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 can be helpful in determining whether the tumors are heterochronous. PMID: 29482987
  3. A study found 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 that induce a DNA damage response. PMID: 28982940
  4. Data indicate that Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) is a viable alternative proliferative marker. PMID: 29040195
  5. These findings indicate that cytokine-induced histone 3 lysine 27 trimethylation acts as 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. This series of 47 diffuse midline gliomas revealed that the histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Research demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, and deposits histone H3.3 onto these. PMID: 28981850
  9. 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), functioning 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. For the first time, researchers described 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 outcomes and has been shown to influence EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data suggest 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. The 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. This 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 the 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. On the other hand, 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, 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. Collectively, the authors verified that histone H3 is a real substrate for GzmA in vivo in the 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 incorporation of histone H3.1-H4 into chromatin but is essential for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays 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. Lower-resolution mass spectrometry instruments can be utilized 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. 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 is the biological significance of HIST1H3A T80 phosphorylation?

HIST1H3A (Histone H3.1) functions as a core component of the nucleosome, fundamentally organizing and compacting DNA into chromatin. Phosphorylation at threonine 80 (T80) represents one of several post-translational modifications that collectively constitute the "histone code." This specific modification contributes to the regulation of DNA accessibility to cellular machinery involved in transcription, DNA repair, DNA replication, and maintenance of chromosomal stability . The phosphorylation state at this residue can dynamically change during cellular processes such as the cell cycle, differentiation, and stress responses, affecting downstream chromatin-dependent processes.

How does Phospho-HIST1H3A (T80) antibody specificity compare to other histone H3 modification antibodies?

Phospho-HIST1H3A (T80) antibodies are generated using immunogens specifically designed around the threonine 80 phosphorylation site of human Histone H3.1 . This specificity distinguishes them from antibodies targeting other histone H3 modifications, such as phosphorylation at serine 28 (S28) or serine 10 (S10). When selecting an antibody, researchers should verify cross-reactivity profiles, as some antibodies may detect similar modifications across multiple histone variants. For instance, while Phospho-HIST1H3A (T80) antibodies are designed to be specific for T80 phosphorylation, validation experiments such as peptide competition assays and immunoblotting with phosphatase-treated samples can confirm specificity against related modifications or unphosphorylated states.

What are the recommended applications for Phospho-HIST1H3A (T80) antibodies?

Based on validated technical specifications, Phospho-HIST1H3A (T80) polyclonal antibodies have proven efficacy in multiple experimental applications:

ApplicationRecommended DilutionKey Considerations
ELISAVaries by manufacturerUseful for quantitative detection of phosphorylated protein
Immunofluorescence (IF)Varies by manufacturerVisualizes cellular localization patterns
Chromatin Immunoprecipitation (ChIP)Varies by manufacturerIdentifies genomic regions associated with the modification

These applications enable comprehensive investigation of T80 phosphorylation in diverse experimental contexts . For optimal results, antibody concentrations should be empirically determined for each specific experimental system, as sensitivity can vary between cell types and experimental conditions.

What is the optimal sample preparation protocol for detecting Phospho-HIST1H3A (T80) in various applications?

For effective detection of Phospho-HIST1H3A (T80), proper sample preparation is critical to preserve phosphorylation status:

For immunofluorescence applications:

  • Fix cells using fresh 3.7% formaldehyde in PBS for 10 minutes at room temperature

  • Permeabilize using either cold methanol (-20°C) for 5 minutes or 0.1% Triton X-100 for 5 minutes

  • Block with 3% FBS in PBS for 30 minutes

  • Apply diluted primary antibody according to manufacturer recommendations

  • Counter-stain nuclei with DNA-specific dyes like Hoechst

For ChIP applications:

  • Cross-link protein-DNA complexes with 1% formaldehyde

  • Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) in all buffers to prevent dephosphorylation

  • Optimize sonication conditions to generate DNA fragments of 200-500 bp

  • Use appropriate blocking reagents to minimize non-specific binding

For all applications, it's crucial to maintain samples at 4°C and include phosphatase inhibitors throughout processing to prevent loss of phosphorylation signal .

How can researchers validate the specificity of Phospho-HIST1H3A (T80) antibody signals?

Rigorous validation ensures experimental results accurately reflect T80 phosphorylation status:

  • Peptide competition assays: Pre-incubate antibody with phosphorylated vs. non-phosphorylated peptides corresponding to the T80 region before application to samples

  • Phosphatase treatment controls: Compare signals from samples treated with or without lambda phosphatase

  • Genetic approaches: Use CRISPR/Cas9-engineered T80A (phospho-null) mutant cell lines as negative controls

  • Western blot verification: Confirm antibody detects a single band at the expected molecular weight (~15.4 kDa) for histone H3

  • Cross-reactivity testing: Verify specificity against related histone variants and modifications

These validation steps are essential for distinguishing genuine T80 phosphorylation signals from potential artifacts or cross-reactivity with other histone modifications.

What storage conditions maintain optimal antibody performance?

To preserve antibody functionality:

  • Store unopened antibody at -20°C to -80°C for long-term storage

  • After initial use, aliquot into small volumes to avoid repeated freeze-thaw cycles

  • For working solutions, store at 4°C for short-term use (typically 1-2 weeks)

  • Include carrier proteins (e.g., BSA) at 1-5 mg/ml in storage buffers to prevent adsorption to container surfaces

  • Check manufacturer specifications for specific storage recommendations, as some formulations may include glycerol (50%) and stabilizers

Proper storage significantly extends antibody shelf-life and maintains consistent performance across experiments.

How should researchers optimize Chromatin Immunoprecipitation (ChIP) protocols for Phospho-HIST1H3A (T80) antibodies?

ChIP optimization for phospho-specific histone antibodies requires careful attention to several parameters:

  • Cross-linking conditions: Optimize formaldehyde concentration (typically 1%) and incubation time (8-10 minutes) to adequately capture histone-DNA interactions without overfixing

  • Chromatin fragmentation: Adjust sonication parameters to generate fragments of 200-500 bp, verifying by agarose gel electrophoresis

  • Antibody amount: Titrate antibody concentration (typically 2-5 μg per ChIP reaction) to determine optimal signal-to-noise ratio

  • Washing stringency: Balance between removing non-specific interactions while preserving specific binding

  • Phosphatase inhibitors: Include comprehensive phosphatase inhibitor cocktails in all buffers

  • Negative controls: Include IgG control and, when possible, samples depleted of the modification (e.g., kinase inhibitor-treated cells)

These optimizations enhance the specificity and sensitivity of ChIP experiments targeting T80 phosphorylation, allowing more accurate mapping of genomic regions associated with this modification .

What cell cycle considerations should influence experimental design when studying T80 phosphorylation?

Histone phosphorylation states often fluctuate throughout the cell cycle, affecting experimental interpretation:

  • Synchronize cell populations using methods appropriate for your cell type:

    • Double thymidine block for G1/S boundary enrichment

    • Nocodazole treatment for mitotic enrichment

    • Serum starvation for G0/G1 enrichment

  • Verify synchronization efficiency using flow cytometry with propidium iodide or other DNA content markers

  • Consider the half-life of T80 phosphorylation and potential rapid dynamic changes during cell cycle transitions

  • Include cell cycle markers (e.g., cyclin levels, other histone modifications like H3S10ph) as internal controls

  • For heterogeneous cell populations, consider dual staining approaches combining T80 phosphorylation detection with cell cycle markers

Understanding the cell cycle dependency of T80 phosphorylation provides crucial context for interpreting experimental results, particularly when comparing different treatment conditions or cell types.

How can researchers integrate Phospho-HIST1H3A (T80) analysis with other epigenetic modifications?

Modern epigenetic research requires multi-parameter approaches to understand the combinatorial nature of histone modifications:

  • Sequential ChIP (Re-ChIP): Perform successive immunoprecipitations to identify genomic regions carrying both T80 phosphorylation and another modification of interest

  • Mass spectrometry approaches:

    • Use targeted approaches to quantify T80 phosphorylation in conjunction with other modifications on the same histone tail

    • Employ middle-down or top-down proteomics to preserve information about co-occurrence of modifications

  • Multiplexed imaging:

    • Combine antibodies against different modifications using spectrally distinct fluorophores

    • Implement iterative fluorescence labeling techniques for higher multiplexing capacity

  • Correlation analyses:

    • Compare ChIP-seq profiles of T80 phosphorylation with datasets for other modifications

    • Identify statistically significant patterns of co-occurrence or mutual exclusion

These integrative approaches reveal how T80 phosphorylation functions within the broader histone code context, providing insights into combinatorial regulation mechanisms .

What are the recommended strategies for quantitative analysis of Phospho-HIST1H3A (T80) levels?

Accurate quantification enables meaningful comparisons across experimental conditions:

  • Western blot quantification:

    • Include loading controls (total H3 or other stable proteins)

    • Establish linear range of detection for densitometry

    • Use ratio of phospho-H3(T80)/total H3 for normalization

  • ELISA-based approaches:

    • Develop standard curves using recombinant phosphorylated peptides

    • Implement sandwich ELISA formats for improved specificity

    • Consider competitive ELISA designs for challenging samples

  • Flow cytometry:

    • Establish compensation controls for multi-parameter analyses

    • Use median fluorescence intensity rather than mean values

    • Normalize to cell cycle distribution when appropriate

  • Image-based quantification:

    • Implement automated segmentation of nuclear regions

    • Measure nuclear intensity values across statistically robust cell numbers (>100 cells)

    • Control for variations in antibody accessibility and nuclear volume

Rigorous quantification approaches with appropriate controls enable detection of subtle changes in T80 phosphorylation levels under varying experimental conditions.

How do different fixation methods affect the detection of Phospho-HIST1H3A (T80)?

Fixation protocols significantly impact phospho-epitope preservation and accessibility:

Fixation MethodAdvantagesLimitationsRecommendations
Formaldehyde (3.7-4%)Preserves nuclear architecture, Good for most applicationsMay mask some epitopesOptimal for IF and ChIP applications
Methanol (-20°C)Excellent for phospho-epitope preservation, Provides additional permeabilizationCan distort some cellular structuresRecommended for phospho-histone detection in IF
Paraformaldehyde + Triton X-100Maintains cellular architecture while allowing antibody accessMay require optimization of permeabilization timeUse 0.1% Triton X-100 for 5 minutes after PFA fixation
Glyoxal-based fixativesAlternative for some phospho-epitopes resistant to aldehyde fixationLess common in histone researchConsider for challenging epitopes

Researchers should empirically test multiple fixation protocols when establishing a new experimental system to identify conditions that optimally preserve T80 phosphorylation while maintaining sample integrity and allowing antibody accessibility .

What strategies can address weak or inconsistent Phospho-HIST1H3A (T80) antibody signals?

When encountering suboptimal signals:

  • Antibody optimization:

    • Titrate antibody concentration across a wider range

    • Extend primary antibody incubation time (overnight at 4°C)

    • Try alternative buffer compositions (varying detergents, salt concentrations)

  • Sample preparation improvements:

    • Verify and enhance phosphatase inhibition throughout sample handling

    • Consider alternative fixation methods that better preserve phospho-epitopes

    • Optimize permeabilization conditions for nuclear access

  • Signal amplification approaches:

    • Implement tyramide signal amplification for immunofluorescence

    • Try biotin-streptavidin systems for signal enhancement

    • Consider more sensitive detection reagents

  • Biological considerations:

    • Verify that your experimental conditions actually induce T80 phosphorylation

    • Include positive controls (e.g., cells treated with phosphatase inhibitors)

    • Consider cell cycle synchronization to enrich for the modification

Systematic troubleshooting of these parameters typically resolves weak signal issues while maintaining specificity.

How can researchers distinguish between specific and non-specific binding in Phospho-HIST1H3A (T80) experiments?

Differentiating genuine signals from artifacts requires rigorous controls:

  • Primary controls:

    • Omit primary antibody while maintaining all other conditions

    • Use non-specific IgG from the same species at equivalent concentration

    • Include samples from cells with experimentally reduced T80 phosphorylation

  • Competitive inhibition:

    • Pre-incubate antibody with phosphorylated vs. non-phosphorylated peptides

    • Titrate competing peptide to demonstrate concentration-dependent inhibition

  • Signal validation:

    • Verify expected subcellular localization patterns

    • Confirm molecular weight in western blot applications

    • Test parallel antibodies from different suppliers or clones

  • Biological validation:

    • Manipulate T80 phosphorylation using kinase inhibitors or genetic approaches

    • Correlate signal with expected biological contexts (e.g., cell cycle stages)

These approaches establish confidence in the specificity of observed signals and provide essential controls for publication-quality research.

How can researchers incorporate Phospho-HIST1H3A (T80) analysis in single-cell epigenetic studies?

Emerging single-cell technologies offer new insights into T80 phosphorylation heterogeneity:

  • Single-cell CUT&Tag/CUT&RUN adaptations:

    • Optimize antibody concentration for single-cell applications

    • Implement carrier DNA strategies to improve recovery

    • Develop rigorous computational approaches for sparse data analysis

  • Mass cytometry (CyTOF) approaches:

    • Develop metal-conjugated antibodies against T80 phosphorylation

    • Combine with other histone modification and cellular markers

    • Implement high-dimensional data analysis techniques (e.g., UMAP, tSNE)

  • Imaging innovations:

    • Super-resolution microscopy to visualize subnuclear distribution

    • Live-cell compatible approaches using nanobodies or other alternatives

    • Spatial correlation with transcriptional activity markers

  • Microfluidic approaches:

    • Develop microfluidic immunocapture for sorting based on modification status

    • Create integrated platforms for phosphorylation analysis in limited samples

These single-cell approaches reveal cellular heterogeneity in T80 phosphorylation patterns that may be masked in bulk analyses, providing deeper insights into epigenetic regulation at the individual cell level.

What are the emerging roles of HIST1H3A T80 phosphorylation in disease processes?

Research connecting T80 phosphorylation to pathological states is evolving:

  • Cancer biology:

    • Altered T80 phosphorylation patterns in specific cancer types

    • Correlation with treatment response and patient outcomes

    • Potential therapeutic targeting of enzymes regulating T80 phosphorylation

  • Neurological disorders:

    • Dysregulation in neurodegenerative diseases

    • Role in neuronal differentiation and plasticity

    • Potential biomarker applications

  • Inflammatory conditions:

    • Modulation during cellular stress responses

    • Relationship to inflammatory signaling cascades

    • Dynamic changes during immune cell activation

  • Developmental disorders:

    • Function in embryonic development and cellular differentiation

    • Consequences of genetic alterations affecting T80 regulation

    • Implications for regenerative medicine applications

Understanding the involvement of T80 phosphorylation in these contexts may identify novel therapeutic targets and diagnostic approaches based on this specific histone modification.

What are the current knowledge gaps regarding HIST1H3A T80 phosphorylation?

Despite advances in histone modification research, several key questions about T80 phosphorylation remain:

  • Enzymatic regulation:

    • Identity of specific kinases and phosphatases acting on T80

    • Signaling pathways controlling these enzymes

    • Temporal dynamics of modification/demodification

  • Functional consequences:

    • Direct effects on chromatin structure and DNA accessibility

    • Reader proteins recognizing T80 phosphorylation

    • Crosstalk with other histone modifications

  • Genomic distribution:

    • Comprehensive mapping across cell types and conditions

    • Association with specific genomic features

    • Changes during differentiation and development

  • Evolutionary conservation:

    • Functional significance across species

    • Species-specific variations in regulation

    • Evolutionary relationship to other phosphorylation sites

These knowledge gaps represent promising areas for future research that will enhance our understanding of T80 phosphorylation's role in chromatin biology.

What best practices should researchers follow when publishing studies involving Phospho-HIST1H3A (T80) antibodies?

To ensure reproducibility and data quality:

  • Antibody validation reporting:

    • Document lot number and source

    • Describe all validation experiments performed

    • Include images of positive and negative controls

    • Report exact experimental conditions

  • Methodology transparency:

    • Provide detailed protocols including buffer compositions

    • Specify exact fixation and permeabilization conditions

    • Report antibody dilutions and incubation parameters

    • Describe quantification methods in detail

  • Data presentation:

    • Include representative images with scale bars

    • Show uncropped western blots with molecular weight markers

    • Present quantitative data with appropriate statistical analysis

    • Provide raw data in supplementary materials when possible

  • Biological context:

    • Clearly describe cell types, treatments, and time points

    • Report cell cycle distribution when relevant

    • Consider wider epigenetic context when interpreting results

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.