HIST1H3A (Ab-28) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HIST1H3A (Ab-28) Antibody

HIST1H3A (Ab-28) Antibody is a specialized reagent targeting phosphorylated serine 28 (pS28) on histone H3, a core nucleosomal protein critical for chromatin structure and gene regulation. This antibody is widely used to study mitotic processes, chromatin dynamics, and transcriptional activation. Its specificity for the phosphorylated form of histone H3 at serine 28 distinguishes it from antibodies targeting other histone modifications, such as phosphorylation at serine 10 (pS10) or acetylation at lysine residues .

Mechanism of Action and Biological Relevance

Phosphorylation of histone H3 at serine 28 (H3S28p) is tightly regulated by mitogen-activated protein kinase (MAPK) pathways, particularly via the kinase MSK1 . This modification is linked to:

  • Mitotic chromatin condensation: H3S28p is enriched during prophase and metaphase, correlating with chromosome condensation .

  • Transcriptional activation: Associated with active/competent chromatin regions, such as promoters of RNA polymerase III-transcribed genes .

  • Cellular stress responses: Induced by tumor promoters (e.g., UV, EGF) and oncoproteins (e.g., c-Myc) .

The antibody’s recognition of H3S28p is validated through peptide array assays, demonstrating specificity against the phosphorylated epitope and excluding cross-reactivity with unmodified or pS10 histone H3 .

Applications in Research

HIST1H3A (Ab-28) Antibody is employed across multiple experimental platforms:

ApplicationMethodologyKey FindingsSources
Flow CytometryIntracellular staining of nocodazole-treated cells (e.g., HeLa)Detects mitotic cells (G2/M phase) with high sensitivity .
Western Blot (WB)Analysis of histone lysates (e.g., HeLa, fruit fly embryos)Identifies a 15–17 kDa band corresponding to H3S28p; validated by peptide competition .
ImmunocytochemistryStaining of neural progenitor cells or HeLa cells (methanol/acetone fixation)Shows nuclear localization in mitotic cells (prophase to telophase) .
Chromatin FractionationImmunoprecipitation of active chromatin fractions (e.g., chicken erythrocytes)H3S28p is enriched in transcriptionally active DNA regions .

Mitotic Cell Detection

The HTA28 clone (a variant of HIST1H3A (Ab-28) Antibody) is validated as a robust marker for M-phase cells. In regenerating rat liver, HTA28 staining indices closely mirrored mitotic indices (MI) and bromodeoxyuridine (BrdU) labeling, with a time lag reflecting G1/S phase progression . Key observations:

  • Sensitivity: Detects all M-phase stages (prophase to telophase), unlike pS10 antibodies, which are restricted to early mitosis .

  • Fixation Sensitivity: Antigenicity is lost with delayed fixation, necessitating immediate sample processing .

Chromatin Dynamics

H3S28p is preferentially associated with active chromatin in chicken erythrocytes:

  • Active/Competent Fractions: H3S28p and H3K4me3 co-localize in DNAase I-sensitive regions .

  • H3.3 Variant: The H3.3 variant is preferentially phosphorylated at S28 in active chromatin .

Transcriptional Regulation

Phosphorylation at S28 is linked to RNA polymerase III transcription machinery activation, as demonstrated in UV- and EGF-stimulated cells .

Comparison of H3S28p Antibodies

Below is a comparison of commercially available antibodies targeting H3S28p, including HIST1H3A (Ab-28) variants:

AntibodyHostClonalityApplicationsReactivityCitations
HTA28 (ab10543)RatMonoclonalWB, ICC, IHCHuman, Mouse, Rat118
ab5169RabbitPolyclonalWB, ICC, PepArrHuman, Fruit fly17
ABIN7181536RabbitPolyclonalWB, ELISAHuman, Mouse, RatN/A

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 shipping method and destination. Please consult your local distributors for specific delivery times.
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, which are fundamental structures responsible for wrapping and compacting DNA into chromatin. This process limits DNA accessibility to cellular machinery that requires DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is controlled through a complex system of post-translational modifications of histones, known as the histone code. This code involves modifications such as acetylation, methylation, phosphorylation, and ubiquitination, which regulate the interactions between DNA and histones. Nucleosome remodeling is another key process in DNA accessibility, and it involves the repositioning or removal of nucleosomes to allow for access to specific DNA regions.
Gene References Into Functions
  1. Studies suggest a mechanism for epigenetic regulation in cancer involving E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Elevated expression of H3K27me3 during a patient's clinical course may indicate the presence of heterochronous tumors. PMID: 29482987
  3. Research has shown that JMJD5, a Jumonji C (JmjC) domain-containing protein, functions as a Cathepsin L-type protease. This protease mediates histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Evidence suggests that the Ki-67 antigen proliferative index has limitations, and phosphohistone H3 (PHH3) is a viable alternative as a proliferative marker. PMID: 29040195
  5. These findings indicate that cytokine-induced histone 3 lysine 27 trimethylation serves as a mechanism to stabilize gene silencing in macrophages. PMID: 27653678
  6. Data suggests that in the early developing human brain, HIST1H3B represents the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This analysis of 47 diffuse midline gliomas revealed a mutually exclusive relationship between histone H3-K27M mutation and IDH1-R132H mutation and EGFR amplification. Furthermore, the K27M mutation was rarely co-occurring with BRAF-V600E mutation but commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Studies demonstrate that histone chaperone HIRA colocalizes with viral genomes. It binds to incoming viral and deposits histone H3.3 onto these, playing a role in viral replication. PMID: 28981850
  9. These experiments show that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl). It functions as a transcriptional co-regulator, influencing gene expression. 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 research 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 affect EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation is not uncommon in adult cerebellar HGG. PMID: 28547652
  14. Data 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 immediately after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Evidence 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 prevalent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. Conversely, 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 efficiently control the DHRS4 gene cluster. PMID: 26864944
  23. Research suggests that nuclear antigen Sp100C acts as a multifaceted sensor of histone H3 methylation and phosphorylation. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis. This alteration promotes 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, facilitating its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. In summary, the authors confirmed that histone H3 is a genuine 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. Data indicates that minichromosome maintenance protein 2 (MCM2) binding is not required for the incorporation of histone H3.1-H4 into chromatin but is essential for the stability of H3.1-H4. PMID: 26167883
  31. Studies suggest that histone H3 lysine methylation (H3K4me3) plays a critical 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. Evidence indicates that 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. The authors report that 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 HIST1H3A and why is phosphorylation at serine 28 important?

HIST1H3A (Histone Cluster 1, H3a) is a variant of histone H3, one of the core histones that form the nucleosome structure in eukaryotic chromatin. Phosphorylation at serine 28 (pSer28) of histone H3 is a critical post-translational modification associated with chromosome condensation during mitosis and is also involved in gene regulation processes. This modification is mediated by kinases such as MSK1 following activation of the MAP kinase signaling pathway in response to stimuli like UV radiation, EGF, and oncoproteins like c-Myc, c-Jun, and c-Fos . Understanding H3 pSer28 is crucial for investigating cellular processes like cell division, transformation, and transcriptional regulation.

How do HIST1H3A (Ab-28) antibodies differ from other histone H3 antibodies?

HIST1H3A (Ab-28) antibodies specifically recognize histone H3 phosphorylated at serine 28, distinguishing them from antibodies targeting other H3 modifications such as phosphorylation at serine 10 (pSer10), various lysine methylations (e.g., H3K4me3, H3K9me3, H3K27me3), or acetylations (e.g., H3K9ac, H3K27ac). The HTA28 monoclonal antibody, for example, specifically recognizes phosphorylated serine 28 of human, mouse, rat, bovine, and hamster histone H3 without cross-reactivity to phosphorylated serine 10 . This specificity makes these antibodies valuable tools for distinguishing between different phosphorylation states that may have distinct biological functions.

What are the typical applications for HIST1H3A (Ab-28) antibodies?

HIST1H3A (Ab-28) antibodies are commonly used in:

  • Flow cytometric analysis to detect mitotic cells

  • Immunohistochemistry (IHC) for visualizing M-phase cells in tissue sections

  • Western blotting (WB) to detect H3 pSer28 levels

  • Immunofluorescence (IF) for cellular localization studies

  • Chromatin immunoprecipitation (ChIP) to identify genomic regions associated with H3 pSer28

  • ELISA for quantitative detection

These applications enable researchers to investigate the dynamics of histone H3 phosphorylation during various cellular processes, particularly during cell division and transcriptional regulation.

What are the optimal fixation protocols for preserving H3 pSer28 antigenicity in tissue samples?

Preserving H3 pSer28 antigenicity requires careful attention to fixation protocols. Research indicates that H3 pSer28 antigenicity is highly labile and can be rapidly lost during sample processing. Key methodological considerations include:

  • Immediate fixation is crucial - a 30% reduction in HTA28 staining index was observed after soaking tissues in PBS for only 2 hours before fixation

  • 10% neutral buffered formalin is recommended as the fixation agent

  • Optimal fixation time is 24 hours at room temperature; extended fixation periods (48-168 hours) result in progressive loss of antigenicity

  • Avoid repeat freeze-thaw cycles when storing antibody solutions

  • For flow cytometry applications, use Protocol A (two-step protocol for cytoplasmic proteins) or Protocol C (two-step protocol with fixation/methanol) rather than Protocol B

These findings emphasize that surgeons and pathologists should immediately and precisely fix samples if clinical specimens are to be examined for H3 pSer28 .

How can I optimize western blotting protocols for HIST1H3A (Ab-28) antibody?

For optimal western blotting results with HIST1H3A (Ab-28) antibody:

ParameterRecommended Condition
Dilution Range1:500-1:2000
Blocking SolutionPBS containing 0.5% BSA
Sample PreparationInclude phosphatase inhibitors in lysis buffers to prevent dephosphorylation
Positive ControlsNocodazole-treated cells (increases mitotic cells and H3 pSer28)
Detection MethodEnhanced chemiluminescence (ECL)
Expected Molecular Weight15-17 kDa

To verify specificity, consider using:

  • Dephosphorylated samples as negative controls

  • Comparing with other H3 modification-specific antibodies to confirm band identity

What factors affect the reproducibility of flow cytometry results with HIST1H3A (Ab-28) antibody?

When using HIST1H3A (Ab-28) antibody for flow cytometry:

  • Pre-titrate the antibody; optimal concentration is ≤0.5 μg per test (where a test refers to the amount needed to stain a cell sample in 100 μL)

  • Cell number should be empirically determined but typically ranges from 10^5 to 10^8 cells/test

  • For optimal detection, use Protocol A (two-step protocol for cytoplasmic proteins) or Protocol C (two-step protocol with fixation/methanol)

  • Include both positive controls (e.g., nocodazole-treated cells with high mitotic index) and negative controls

  • Use freshly prepared samples, as phosphorylation status can change during storage

  • Ensure consistency in permeabilization conditions, as insufficient permeabilization can limit nuclear antigen access

How can HIST1H3A (Ab-28) antibody be used to study the relationship between histone phosphorylation and chromatin states?

Studies using H3 pSer28 antibodies have revealed important insights into chromatin regulation:

Research has shown that H3 phosphorylation at serine 28 preferentially partitions with chromatin enriched in transcriptionally active/competent regions. When chromatin was fractionated, H3S28p preferentially partitioned into fractions enriched in active and competent DNA sequences (F1 and F2), while H3S10p was equally distributed between all fractions . This suggests distinct roles for these two phosphorylation sites despite their proximity in the histone tail.

To investigate these relationships:

  • Perform chromatin fractionation to separate active vs. inactive chromatin regions

  • Use HIST1H3A (Ab-28) antibody in combination with antibodies against other histone marks in ChIP-seq experiments

  • Correlate H3 pSer28 distribution with gene expression data to identify regulatory relationships

  • Compare patterns of H3 pSer28 with other histone marks (H3K4me3, H3K27ac) associated with active chromatin

This approach enables researchers to map the genome-wide distribution of H3 pSer28 and correlate it with transcriptional activity and other chromatin features.

How reliable is HIST1H3A (Ab-28) antibody for quantifying mitotic cells compared to traditional methods?

The HTA28 monoclonal antibody has been extensively validated for detecting mitotic cells:

  • In a study using regenerating rat liver after partial hepatectomy, the HTA28 staining index (SI) closely paralleled the mitotic index (MI) determined by morphological assessment, but was consistently higher at all time points

  • HTA28 immunoreactivity was maintained throughout all stages of M-phase (prophase through telophase), making it more comprehensive than morphological assessment alone

  • The spatial distribution of HTA28-positive cells corresponded with those identified by other proliferative cell markers

For optimal quantification:

  • Include both positive controls (tissue with known high mitotic activity) and negative controls

  • Consider dual staining with other cell cycle markers to confirm specificity

  • Compare results with established methods (e.g., phospho-histone H3 Ser10, Ki-67) for validation

  • Be aware that telophase cells may stain less intensely than earlier mitotic phases

What is the relationship between H3 pSer28 and H3 variant incorporation in chromatin remodeling?

Recent research has revealed complex relationships between histone phosphorylation and histone variant dynamics:

Studies show that histone variants H3.1 and H3.3 are differentially regulated and can be subject to distinct post-translational modifications. H3.1 serves as a chromatin redox sensor that is engaged by mitochondrial H2O2 signaling . The replacement of H3.1 by H3.3 has been correlated with increased chromatin accessibility and gene expression, particularly for EMT genes .

To investigate these relationships:

  • Use HIST1H3A (Ab-28) antibody in combination with antibodies specific for H3.1 or H3.3 variants

  • Perform ChIP-seq or CUT&RUN to map genome-wide distribution of pSer28 on specific H3 variants

  • Correlate phosphorylation patterns with histone variant dynamics during processes like cell division or differentiation

  • Use sequential ChIP (re-ChIP) to determine if pSer28 co-occurs with other modifications on the same histone molecule

This approach can provide insights into how phosphorylation at serine 28 might differentially affect the function of distinct histone H3 variants.

How can I verify the specificity of HIST1H3A (Ab-28) antibody in my experimental system?

To confirm antibody specificity:

  • Peptide competition assay: Pre-incubate the antibody with a synthetic phosphopeptide containing pSer28 (e.g., CKKAARKpSAPATGGV) before applying to your sample; this should eliminate specific signal

  • Comparison with non-phosphorylated controls: Include samples treated with phosphatase or phosphorylation inhibitors

  • Timing experiments: Since H3 pSer28 is cell cycle-dependent, synchronize cells and test at different cycle stages; the signal should increase during mitosis

  • Cross-reactivity testing: Test against closely related modifications like H3 pSer10 to ensure specificity

  • Knockout/knockdown validation: Where possible, use cells lacking the target or with reduced expression

It's worth noting that HTA28 specifically recognizes H3 phosphorylated at serine 28 but not serine 10, providing a useful tool for distinguishing between these modifications .

What are common pitfalls when using HIST1H3A (Ab-28) antibody and how can they be avoided?

Common challenges and their solutions include:

ChallengeSolution
Loss of phosphorylation during sample processingInclude phosphatase inhibitors in all buffers; process samples rapidly
Low signal in IHC/IFEnsure proper antigen retrieval; optimize fixation time (24 hrs optimal)
Non-specific bindingIncrease blocking time/concentration; optimize antibody dilution
High backgroundUse fresh blocking reagents; include 0.5% BSA in antibody diluent
Batch-to-batch variabilityInclude consistent positive controls with each experiment
False positives in apoptotic cellsUse dual staining with apoptotic markers to exclude these cells

Additionally, always validate new lots of antibody against a known standard, and consider using recombinant antibodies when available for greater consistency.

How do I interpret conflicting results between HIST1H3A (Ab-28) antibody and other histone modification markers?

When facing discrepancies between H3 pSer28 and other histone modification patterns:

  • Consider biological timing: Different modifications occur at distinct cell cycle phases or in response to different stimuli. H3S28 phosphorylation is mediated by MSK1 following activation of the MAP kinase signaling pathway , while other modifications may follow different pathways.

  • Examine cross-talk between modifications: Modifications on neighboring residues can affect antibody recognition. For example, acetylation at K27 might impact recognition of pS28.

  • Validate with multiple antibodies: Use antibodies from different sources or that recognize different epitopes containing the same modification.

  • Consider technical factors: Different antibodies may require distinct fixation and antigen retrieval protocols for optimal performance.

  • Employ complementary approaches: Combine antibody-based detection with mass spectrometry to resolve discrepancies.

Research has shown that H3 pSer28 and H3 pSer10 can have distinct distributions and functions despite their proximity in the histone tail , so apparent conflicts might reflect genuine biological differences rather than technical artifacts.

How can HIST1H3A (Ab-28) antibody be used to study the role of H3 pSer28 in disease processes?

Emerging applications of H3 pSer28 antibodies in disease research include:

  • Cancer research: H3 pSer28 has been linked to chromosome condensation during mitosis, cell transformation, and regulation of RNA polymerase III transcription machinery . Aberrant phosphorylation patterns may contribute to genomic instability in cancer.

  • Cell senescence studies: Histone modifications change dramatically during senescence. Research has identified distinct histone H3 tail proteolytic processing in models of both oncogene-induced and replicative senescence . HIST1H3A (Ab-28) antibody can help track changes in phosphorylation patterns during senescence induction.

  • Neural development: Recent research has discovered that histone H3 monoaminylation dynamics regulate neural transcriptional programs, with H3Q5 identified as a primary site of modification . Studying the interplay between these modifications and phosphorylation at serine 28 could provide insights into neuronal differentiation and function.

  • Therapeutic response monitoring: Changes in histone phosphorylation patterns can serve as biomarkers for response to treatment, particularly for drugs targeting kinase pathways that affect histone phosphorylation.

What are the considerations for multiplexing HIST1H3A (Ab-28) antibody with other antibodies in imaging or flow cytometry?

For successful multiplexing experiments:

  • Antibody compatibility:

    • Choose antibodies raised in different host species to avoid cross-reactivity

    • For same-species antibodies, use directly conjugated formats or sequential staining protocols

    • Consider using Zenon labeling technology for antibodies from the same species

  • Fluorophore selection:

    • When using Alexa Fluor 488-conjugated HIST1H3A (Ab-28) antibody, select complementary fluorophores with minimal spectral overlap

    • For the Alexa Fluor 488 conjugate: Excitation: 488 nm; Emission: 519 nm

    • Pair with far-red fluorophores for maximum separation

  • Staining protocol optimization:

    • Test antibodies individually before combining

    • Optimize concentration of each antibody separately

    • Consider sequential staining if antibodies require different fixation/permeabilization conditions

  • Controls for multiplexing:

    • Include single-stained controls for compensation

    • Use fluorescence-minus-one (FMO) controls to set proper gates

    • Include isotype controls for each antibody

How does the binding of HIST1H3A (Ab-28) antibody affect the interpretation of chromatin dynamics studies?

When using HIST1H3A (Ab-28) antibody in chromatin studies, consider these advanced interpretational factors:

  • Antibody accessibility issues: The compact nature of chromatin might limit antibody access to the H3 pSer28 epitope in certain chromatin states. This could lead to underestimation of modification levels in highly condensed regions.

  • Epitope masking by protein complexes: Protein complexes that recognize H3 pSer28 might compete with antibody binding, potentially resulting in false negatives in regions where the modification is functionally engaged.

  • Temporal dynamics considerations: H3 pSer28 is a dynamic modification whose levels change rapidly during cell cycle progression. Time-course studies with synchronized cells are essential for accurate interpretation.

  • Single-cell vs. population heterogeneity: Flow cytometry or imaging approaches with HIST1H3A (Ab-28) antibody can reveal cell-to-cell variability that might be masked in bulk chromatin studies like ChIP-seq.

  • Relationship to chromatin accessibility: Studies have shown that H3 phosphorylation at serine 28 preferentially partitions with chromatin enriched in transcriptionally active/competent regions . This association should be considered when interpreting genomic distribution patterns.

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.