Phospho-HIST1H3A (T3) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

Phospho-HIST1H3A (T3) antibodies are produced by immunizing hosts with synthetic phosphorylated peptides targeting human HIST1H3A (NP_003520.1). Key attributes include:

FeatureDetailsExample Products (SKU)
Host SpeciesRabbit (polyclonal or monoclonal)CABP0846 , CABP1152
ReactivityHuman, Mouse, Rat, others (predicted)84739-1-PBS , MAB12794
ApplicationsWestern blot (WB), Immunofluorescence (IF), Immunohistochemistry (IHC), ELISAGTX128116 , PAB16951
Recommended DilutionWB: 1:500–1:2000; IF/IHC: 1:50–1:500CABP1152 , CSB-RA010109A03phHU
ImmunogenSynthetic phosphopeptide around T3 (e.g., sequence: MART K)CABP0846 , MAB12794

These antibodies show minimal cross-reactivity with non-phosphorylated histones or other phosphorylated residues (e.g., H3S10ph) .

Chromatin Dynamics and Mitosis

  • Mitotic Chromosome Condensation: Phospho-H3T3 marks mitotic chromatin, aiding in Aurora kinase B recruitment and spindle assembly . Antibodies like CABP1152 detect this modification in nocodazole-synchronized cells (HeLa, NIH/3T3) .

  • Cell Cycle Studies: Elevated H3T3 phosphorylation correlates with G2/M phase transition, detectable via flow cytometry or immunofluorescence .

Disease Research

  • Cancer Biomarker: Phospho-H3T3 levels are prognostic in breast cancer, melanoma, and meningiomas. Immunostaining with pHH3 antibodies quantifies mitotic indices in tumors .

  • Epigenetic Dysregulation: Aberrant H3T3 phosphorylation is linked to transcriptional silencing and chromatin remodeling defects .

Technical Workflows

  • Chromatin Immunoprecipitation (ChIP): Recombinant antibodies (e.g., CSB-RA010109A03phHU) enable precise mapping of H3T3ph-associated genomic regions .

  • High-Throughput Screening: HTRF kits (e.g., revvity.com ) quantify H3T3 phosphorylation in cell-based assays without electrophoresis.

Specificity and Sensitivity

  • Western Blot: Distinct bands at ~15–17 kDa in HeLa, NIH/3T3, and 293T lysates . Pre-absorption with phosphorylated peptides abolishes signal .

  • Immunofluorescence: Nuclear localization in mitotic cells, colocalizing with DAPI-stained condensed chromosomes .

Validation MetricExample ResultsSource
Lot ConsistencyRecombinant antibodies show <10% variability in binding affinity .CSB-RA010109A03phHU
Cross-ReactivityNo binding to H3K4me3, H3K9me3, or H3S10ph .MAB12794

Key Research Findings

  1. Mitotic Regulation: Phospho-H3T3 recruits Aurora kinase B to centromeres, ensuring accurate chromosome segregation .

  2. Cancer Prognosis: High pHH3 indices correlate with aggressive tumor phenotypes and poor survival .

  3. Chromatin Signaling: H3T3 phosphorylation antagonizes H3K4 methylation, modulating transcriptional activity .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery information.
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, which wrap and compact DNA into chromatin. This compaction limits DNA accessibility to 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 system of post-translational modifications of histones, also known as the histone code. These modifications, along with nucleosome remodeling, contribute to the intricate control of gene expression.
Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer can be influenced by 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 helpful in determining whether the tumors are heterochronous. PMID: 29482987
  3. Recent findings demonstrate that JMJD5, a protein containing a Jumonji C (JmjC) domain, acts as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stressful conditions leading to a DNA damage response. PMID: 28982940
  4. Evidence suggests that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) offers an alternative as a proliferative marker. PMID: 29040195
  5. These results 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. This series of 47 diffuse midline gliomas revealed that the histone H3-K27M mutation was mutually exclusive with the IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with the BRAF-V600E mutation and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas... PMID: 26517431
  8. Data show 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 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 describes, 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). This mutation is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. The presence of the 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. The 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 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 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. 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 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. The 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 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 Phospho-HIST1H3A (T3) and what is its biological significance?

Phospho-HIST1H3A (T3) refers to the phosphorylated form of Histone H3 at threonine 3. This specific histone modification plays critical roles in chromatin remodeling and gene transcription regulation. The phosphorylation of Histone H3 at threonine 3 (T3) is particularly implicated in essential cellular processes including DNA repair, cell cycle progression, and apoptosis . This post-translational modification is a key regulatory mechanism during mitosis, specifically in the condensation of chromosomes, making it an important marker for studying cell division dynamics and chromatin structure . Growth factor stimulation has been shown to result in phosphorylation of histone H3, which correlates with the expression of immediate early genes, suggesting this modification is associated with transcriptional activation mechanisms .

How do polyclonal and monoclonal Phospho-HIST1H3A (T3) antibodies differ in research applications?

Polyclonal and monoclonal Phospho-HIST1H3A (T3) antibodies offer different advantages depending on research needs:

FeaturePolyclonal Antibodies (e.g., CABP0846)Monoclonal Antibodies (e.g., CABP1152, MAB12794)
Epitope RecognitionMultiple epitopes on the antigenSingle epitope on the antigen
SpecificityBroader recognition but potential for cross-reactivityHigher specificity with reduced cross-reactivity
Batch-to-Batch VariationHigher variationMinimal variation
SensitivityOften higher due to multiple epitope bindingMay be less sensitive for low abundance targets
ApplicationsExcellent for initial detection and screeningPreferred for quantitative or reproducible assays

Both types are raised in rabbits and target the phosphorylated form of Histone H3 at T3. Polyclonal antibodies provide broader recognition but may exhibit increased cross-reactivity, while monoclonal antibodies offer higher specificity with minimal batch-to-batch variation . Monoclonal antibodies like clone RM159 demonstrate high specificity with no cross-reactivity with other phosphorylated histones, making them invaluable for discriminating between specific histone modifications in complex experimental systems .

What are the validated applications for Phospho-HIST1H3A (T3) antibodies and their recommended protocols?

Phospho-HIST1H3A (T3) antibodies have been validated for multiple research applications with specific recommended protocols:

ApplicationRecommended DilutionKey Protocol Considerations
Western Blot1:500 - 1:2000 (CABP0846) 1:500 - 1:1000 (CABP1152) 0.1 - 1 μg/mL (MAB12794) Best results obtained with acid extracts from cells; treatment with nocodazole enhances signal
ELISAStandard protocols0.2 - 1 μg/mL concentration recommended for optimal results
Immunocytochemistry0.5 - 2 μg/mL Works effectively on fixed HeLa cells; can be combined with cytoskeletal stains
Immunohistochemistry~1 μg/mL Validated on formalin-fixed paraffin-embedded tissue sections

For optimal Western blot results, acid extracts from HeLa cells treated with nocodazole (which arrests cells in mitosis) often provide the clearest bands at approximately 16kDa, corresponding to phosphorylated Histone H3 . For immunocytochemistry applications, these antibodies can be effectively combined with other cellular markers like fluorescein phalloidin to visualize actin filaments, enabling multiparameter analysis of cellular structures during different phases of the cell cycle .

How can Phospho-HIST1H3A (T3) antibodies be used to study mitotic progression in cell culture models?

Phospho-HIST1H3A (T3) antibodies provide powerful tools for studying mitotic progression in experimental models. The methodological approach involves:

  • Cell Synchronization: Treat cells with nocodazole or other mitotic arrest agents to enrich for populations in mitosis. This significantly increases the proportion of cells with phosphorylated H3-T3, as this modification is most prominent during chromosome condensation in early mitosis .

  • Detection Methods:

    • For quantitative assessment, use flow cytometry with anti-Phospho-HIST1H3A (T3) antibodies to measure the percentage of mitotic cells in a population

    • For visualization, employ immunofluorescence microscopy with these antibodies (0.5-2 μg/mL) to observe the spatial and temporal dynamics of H3-T3 phosphorylation during different mitotic phases

  • Co-staining Approaches: Combine Phospho-HIST1H3A (T3) antibody (red channel) with actin staining (green channel) and DNA counterstains (blue channel) to create comprehensive visualizations of cellular structures during mitosis . This triple-labeling approach enables researchers to correlate chromosome condensation with other cellular reorganization events during cell division.

  • Time-course Experiments: Track the appearance and disappearance of the H3-T3 phosphorylation signal at different time points after releasing cells from synchronization, which provides insights into the kinetics of this modification during mitotic progression.

This methodology allows researchers to precisely track mitotic cells and evaluate the effects of experimental treatments on cell cycle progression and chromosome dynamics .

How can researchers address inconsistent Phospho-HIST1H3A (T3) antibody staining patterns in immunocytochemistry?

Inconsistent staining patterns with Phospho-HIST1H3A (T3) antibodies may result from several factors. Here's a methodological approach to troubleshooting:

  • Cell Cycle Dependence: Phosphorylation of H3-T3 is highly cell cycle-dependent, primarily occurring during mitosis. Unsynchronized cell populations will show heterogeneous staining with only mitotic cells displaying strong signals. To obtain more consistent results:

    • Synchronize cells using nocodazole treatment (recommended for HeLa, NIH/3T3, and C6 cell lines)

    • Perform time-course experiments to identify optimal harvest times after synchronization

  • Fixation Protocol Optimization:

    • For formaldehyde fixation: Use freshly prepared 4% paraformaldehyde for 10-15 minutes at room temperature

    • For methanol fixation: Ice-cold methanol for 10 minutes preserves phospho-epitopes more effectively

    • Avoid overfixation, which can mask antibody epitopes

  • Antibody Concentration Titration:

    • Perform a dilution series ranging from 0.1-5 μg/mL to identify optimal concentration

    • For challenging samples, consider signal amplification systems like tyramide signal amplification

  • Phosphatase Inhibition:

    • Include phosphatase inhibitors in all buffers until fixation is complete

    • Consider adding sodium fluoride (10mM) and sodium orthovanadate (1mM) to preservation buffers

  • Background Reduction:

    • Use appropriate blocking with 1-5% BSA (found in storage buffer of antibody preparations)

    • Include 0.1-0.3% Triton X-100 in blocking buffer to improve antibody penetration

When optimized, immunocytochemistry with Phospho-HIST1H3A (T3) antibodies should yield distinct nuclear staining specifically in mitotic cells, with particularly intense signals at condensed chromosomes .

What controls should be included when validating Phospho-HIST1H3A (T3) antibody specificity for chromatin immunoprecipitation (ChIP) experiments?

When validating Phospho-HIST1H3A (T3) antibodies for ChIP experiments, implementing rigorous controls is essential:

  • Positive Controls:

    • Include nocodazole-treated cell samples, which are enriched for mitotic cells with H3-T3 phosphorylation

    • Use known genomic regions associated with mitotic H3-T3 phosphorylation

  • Negative Controls:

    • Perform ChIP with IgG from the same species as the primary antibody (rabbit IgG for these antibodies)

    • Include samples treated with phosphatase to remove phosphorylation modifications

    • Use quiescent cells (G0 phase) where H3-T3 phosphorylation should be minimal

  • Specificity Validation:

    • Peptide competition assay using the immunogen peptide (synthetic phosphorylated peptide around T3 of human HIST1H3A)

    • Western blot analysis in parallel with ChIP to confirm antibody specificity to the 16kDa band corresponding to phosphorylated H3

  • Technical Validations:

    • Input chromatin controls (typically 1-5% of starting material)

    • Serial dilution of antibody to determine optimal concentration

    • Sequential ChIP with antibodies against different histone modifications to assess co-occurrence

  • Biological Validations:

    • Use haspin kinase inhibitors (the enzyme responsible for H3-T3 phosphorylation) to create negative control samples

    • Compare results between multiple antibodies targeting the same modification (e.g., both monoclonal and polyclonal anti-Phospho-HIST1H3A)

These controls ensure that the observed chromatin enrichment is specific to the Phospho-HIST1H3A (T3) modification and not due to experimental artifacts or cross-reactivity with other histone modifications.

How does Phospho-HIST1H3A (T3) relate to other histone H3 modifications in the context of the histone code?

Phospho-HIST1H3A (T3) operates within the complex network of histone modifications known as the "histone code." Its relationships with other modifications reveal important regulatory mechanisms:

  • Functional Relationships with Other Modifications:

    • H3-T3 phosphorylation often occurs concurrently with H3-S10 phosphorylation during mitosis, but they are catalyzed by different kinases and serve distinct functions

    • H3-T3 phosphorylation has been shown to disrupt the binding of certain methyl-binding proteins to H3K4 methylation, creating a "phospho-methyl switch" mechanism

    • The interplay between H3-T3 phosphorylation and other modifications like H3K9 methylation contributes to the regulation of heterochromatin formation

  • Temporal Dynamics During Cell Cycle:

    • Unlike H3-S10 phosphorylation, which can occur during both interphase (associated with gene activation) and mitosis, H3-T3 phosphorylation is primarily observed during mitosis

    • H3-T3 phosphorylation appears early in prophase, reaches maximum levels in metaphase, and decreases during anaphase and telophase

  • Spatial Distribution on Chromatin:

    • H3-T3 phosphorylation is found along the entire chromosome arms during mitosis but is particularly enriched at centromeric regions

    • In contrast, other mitotic modifications like H3-S28 phosphorylation show different distribution patterns

  • Crosstalks with DNA Methylation:

    • Emerging evidence suggests complex interactions between histone phosphorylation events, including H3-T3, and DNA methylation patterns

    • These interactions may be particularly relevant in cancer research contexts where epigenetic dysregulation is common

Understanding these relationships is essential for developing comprehensive models of chromatin regulation and for identifying potential therapeutic targets in diseases with epigenetic components .

What is the role of Phospho-HIST1H3A (T3) in cancer biology, and how can researchers apply these antibodies in oncology studies?

Phospho-HIST1H3A (T3) has emerging significance in cancer biology, with several applications in oncology research:

  • Prognostic Marker Potential:

    • While studies have yielded mixed results regarding Phospho-Histone H3 (pHH3) as a prognostic marker, research indicates it may have utility in specific cancer contexts

    • Approximately 60% of patients exhibiting progression of non-functioning pituitary adenomas demonstrated pHH3 immunoreactivity, suggesting potential value as a biomarker that warrants further investigation in prospective studies

  • Correlation with Proliferation Markers:

    • Interestingly, pHH3 immunoreactivity did not correlate with established proliferation markers like MIB-1/Ki-67 or p53 in some studies

    • This suggests pHH3 may provide independent and complementary information to traditional proliferation markers

  • Methodological Applications in Cancer Research:

    • Tumor Heterogeneity Assessment: Using these antibodies for immunohistochemistry on tumor sections can reveal spatial heterogeneity in mitotic activity

    • Treatment Response Monitoring: Changes in H3-T3 phosphorylation patterns following treatment with anti-mitotic drugs can serve as pharmacodynamic markers

    • Cancer Cell Line Authentication: Distinct patterns of histone modifications, including H3-T3 phosphorylation, can help characterize and authenticate cancer cell lines

  • Technical Approaches for Cancer Studies:

    • Immunohistochemical staining of formalin-fixed paraffin-embedded cancer tissue sections (e.g., breast cancer) at 1 μg/mL concentration

    • Combined analysis with other markers: While pHH3 alone may not predict time to progression, multiparameter analysis including MIB-1/Ki-67 and p53 may enhance prognostic value

    • Development of quantitative image analysis algorithms to objectively assess pHH3 immunoreactivity in tissue sections

These applications highlight the importance of Phospho-HIST1H3A (T3) antibodies as tools for investigating the complex role of histone modifications in cancer development, progression, and treatment response .

How can Phospho-HIST1H3A (T3) antibodies be integrated into multi-parameter flow cytometry panels for cell cycle analysis?

Integrating Phospho-HIST1H3A (T3) antibodies into multi-parameter flow cytometry provides powerful insights into cell cycle dynamics:

  • Panel Design Considerations:

    • Core Markers: Combine Phospho-HIST1H3A (T3) antibody with DNA content dyes (DAPI or propidium iodide) for basic cell cycle positioning

    • Complementary Cell Cycle Markers: Include additional markers such as cyclin B1 (G2/M), cyclin E (G1/S), and cyclin A (S/G2)

    • Proliferation Markers: Add Ki-67 to distinguish cycling from quiescent cells

    • DNA Damage Response Markers: Incorporate γH2AX to assess DNA damage during different cell cycle phases

  • Protocol Optimization:

    • Fixation and Permeabilization: Use methanol or formaldehyde fixation followed by Triton X-100 permeabilization to preserve phospho-epitopes while allowing antibody access

    • Signal Amplification: Consider tyramide signal amplification for detecting low abundance phosphorylation events

    • Fluorophore Selection: Choose fluorophores with minimal spectral overlap to reduce compensation requirements

    • Titration: Determine optimal antibody concentration using positive control samples (nocodazole-treated cells)

  • Advanced Analysis Approaches:

    • High-Dimensional Analysis: Apply tSNE or UMAP dimensionality reduction to visualize complex cell cycle transitions

    • Trajectory Analysis: Use algorithms like Wanderlust or FLOW-MAP to map pseudotime trajectories through the cell cycle

    • Machine Learning Classification: Train models to identify subtle cell cycle perturbations based on multi-parameter data

  • Application Examples:

    • Drug Response Profiling: Measure cell cycle arrest patterns induced by various compounds

    • Checkpoint Activation Analysis: Assess checkpoint activation by correlating H3-T3 phosphorylation with other markers

    • Cell Heterogeneity Characterization: Identify subpopulations with distinct cell cycle kinetics within complex samples

This integrated approach provides substantially more information than traditional DNA content analysis alone, enabling researchers to dissect complex cell cycle perturbations in response to experimental manipulations or disease states.

What emerging techniques combine Phospho-HIST1H3A (T3) detection with genome-wide approaches to understand chromatin dynamics?

Several cutting-edge methodologies now combine Phospho-HIST1H3A (T3) detection with genome-wide approaches:

  • CUT&RUN and CUT&Tag with Phospho-HIST1H3A (T3) Antibodies:

    • These techniques offer higher resolution and lower background than traditional ChIP-seq

    • Protocol modifications: Use unfixed cells and include phosphatase inhibitors throughout the procedure

    • Advantages: Requires fewer cells and provides cleaner signal-to-noise ratio for phospho-histone modifications

    • Application: Mapping precise genomic locations of H3-T3 phosphorylation during mitotic progression

  • Single-Cell Multi-Omics Approaches:

    • scCUT&Tag: Detect H3-T3 phosphorylation patterns in individual cells to reveal cell-to-cell heterogeneity

    • CITE-seq adaptations: Combine cell surface protein detection with intracellular Phospho-HIST1H3A (T3) detection

    • Multi-modal integration: Correlate H3-T3 phosphorylation with transcriptome or other epigenetic modifications at single-cell resolution

  • Super-Resolution Microscopy Techniques:

    • STORM/PALM imaging: Visualize nanoscale distribution of H3-T3 phosphorylation on chromatin

    • Live-cell imaging: Use antibody fragments or nanobodies to track H3-T3 phosphorylation dynamics in living cells

    • Correlative light-electron microscopy (CLEM): Connect H3-T3 phosphorylation patterns with chromatin ultrastructure

  • Mass Spectrometry-Based Approaches:

    • Targeted proteomics: Develop selective reaction monitoring (SRM) assays for quantitative assessment of H3-T3 phosphorylation

    • PTM crosstalk analysis: Use middle-down or top-down proteomics to analyze co-occurrence of H3-T3 phosphorylation with other modifications

    • Proximity labeling: Identify proteins associated with H3-T3 phosphorylated chromatin regions using BioID or APEX2 systems

These emerging techniques are expanding our understanding of the dynamic role of H3-T3 phosphorylation in chromatin biology and opening new avenues for investigating its function in both normal cellular processes and disease states .

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.