Phospho-HIST1H3A (T3) antibodies are produced by immunizing hosts with synthetic phosphorylated peptides targeting human HIST1H3A (NP_003520.1). Key attributes include:
These antibodies show minimal cross-reactivity with non-phosphorylated histones or other phosphorylated residues (e.g., H3S10ph) .
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 .
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 .
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.
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 .
Mitotic Regulation: Phospho-H3T3 recruits Aurora kinase B to centromeres, ensuring accurate chromosome segregation .
Cancer Prognosis: High pHH3 indices correlate with aggressive tumor phenotypes and poor survival .
Chromatin Signaling: H3T3 phosphorylation antagonizes H3K4 methylation, modulating transcriptional activity .
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 .
Polyclonal and monoclonal Phospho-HIST1H3A (T3) antibodies offer different advantages depending on research needs:
| Feature | Polyclonal Antibodies (e.g., CABP0846) | Monoclonal Antibodies (e.g., CABP1152, MAB12794) |
|---|---|---|
| Epitope Recognition | Multiple epitopes on the antigen | Single epitope on the antigen |
| Specificity | Broader recognition but potential for cross-reactivity | Higher specificity with reduced cross-reactivity |
| Batch-to-Batch Variation | Higher variation | Minimal variation |
| Sensitivity | Often higher due to multiple epitope binding | May be less sensitive for low abundance targets |
| Applications | Excellent for initial detection and screening | Preferred 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 .
Phospho-HIST1H3A (T3) antibodies have been validated for multiple research applications with specific recommended protocols:
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 .
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 .
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:
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:
When optimized, immunocytochemistry with Phospho-HIST1H3A (T3) antibodies should yield distinct nuclear staining specifically in mitotic cells, with particularly intense signals at condensed chromosomes .
When validating Phospho-HIST1H3A (T3) antibodies for ChIP experiments, implementing rigorous controls is essential:
Positive Controls:
Negative Controls:
Specificity Validation:
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.
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:
Understanding these relationships is essential for developing comprehensive models of chromatin regulation and for identifying potential therapeutic targets in diseases with epigenetic components .
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:
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 .
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.
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 .