HIST1H3A encodes a replication-dependent histone H3 variant essential for nucleosome assembly. Histones compact DNA into chromatin, regulating transcription, DNA repair, and replication. Post-translational modifications (PTMs) at specific residues, such as K45, modulate these processes .
The HIST1H3A (Ab-45) antibody is designed to detect PTMs at K45. While the exact modification type (e.g., methylation, acetylation) is not explicitly defined in all sources, available data suggest specificity for methylation states:
Detects methylation at K45, a modification linked to transcriptional activation or repression depending on context .
Validated in WB (1:500–1:2000 dilution) and ICC (1:200–1:1000) .
Aberrant histone methylation at K45 is implicated in cancers and developmental disorders .
Used to study chromatin remodeling in apoptosis and cell differentiation .
Dot Blot: No reactivity with unmodified H3K45 or other histone modifications (e.g., H3K27me3) .
ELISA: Strong signal for methylated K45 peptides (EC₅₀: ~1:73,200) .
| Assay | Sensitivity | Specificity |
|---|---|---|
| Western Blot | 15 kDa band | No cross-reactivity with H2A/H2B/H4 . |
| Immunofluorescence | Nuclear staining | Confirmed in HeLa cells . |
Methylation Dynamics: Di-methylation at K45 correlates with active transcription in gene promoters .
Apoptosis Link: PTMs near K45 (e.g., phosphorylation at T45) are associated with apoptotic signaling .
Further studies are needed to:
HIST1H3A is a gene encoding histone H3.1, one of the canonical histone H3 variants. Histone H3.1 is a 135-amino acid protein (after cleavage of the first methionine) containing a histone tail, four α-helices, and two loop domains . While H3.1 shares high sequence homology with other H3 variants, it has some unique features:
H3.1 contains a unique cysteine residue at position 96 that can function as a chromatin-embedded redox sensor
Unlike H3.3, H3.1 is predominantly synthesized and incorporated into chromatin during DNA replication in the S phase of the cell cycle
The tails of H3.1 and other H3 histones (amino acids 1-89) are identical except for residue 31, where H3.1 contains alanine while other variants contain serine
The specific molecular weight of HIST1H3A protein is approximately 15,404 Da .
Threonine 45 of histone H3 (H3T45) has been identified as a site of phosphorylation in vivo . This modification is part of the core histone domain rather than the histone tail, giving it the potential to directly modulate nucleosome structure and consequently DNA accessibility . Key characteristics of this modification include:
H3T45 phosphorylation (H3T45ph) can be detected at low levels in untreated cells but increases substantially following phosphatase inhibition
This phosphorylation appears to be linked to apoptotic processes, suggesting its role in cellular stress responses
The modification occurs within the core nucleosomal structure, potentially influencing DNA-histone interactions more directly than tail modifications
Antibodies targeting phosphorylated H3T45 provide essential tools for studying this specific modification in various research contexts. These antibodies enable:
Visualization and quantification of H3T45ph during different cellular processes
Mapping genomic locations where this modification occurs through chromatin immunoprecipitation (ChIP) experiments
Investigation of relationships between H3T45ph and other histone modifications or cellular events
Assessment of how H3T45ph changes in response to cellular stresses or signaling events
These antibodies are typically validated through multiple methods, including peptide competition assays, ELISA, and peptide dot blots to ensure specificity against the phosphorylated form of the residue .
Thorough validation of antibodies targeting H3T45ph is essential for reliable experimental results. Based on established methods, the following protocol is recommended:
Peptide competition assay: Pre-incubate the antibody with increasing concentrations of either phosphorylated or non-phosphorylated peptides corresponding to the region around H3T45, then perform western blotting or immunostaining to demonstrate specific blocking with the phosphorylated peptide only .
ELISA validation: Coat wells with H3 peptides (phosphorylated at T45 and non-phosphorylated controls), then test antibody binding using standard ELISA protocols. Quantitative measurement of binding affinity and specificity should show strong preference for the phosphorylated epitope .
Peptide dot blots: Spot varying concentrations of H3T45ph and control peptides on membranes, then probe with the antibody to visualize binding specificity .
Mass spectrometry correlation: Treat cells with phosphatase inhibitors like calyculin A for 15 minutes, extract histones, and perform parallel analyses using both the antibody of interest and MS/MS to confirm detection of the same modification .
Knockout/knockdown controls: When possible, validate antibody specificity using genetic models where the target histone or modifying enzyme has been depleted.
For optimal western blotting results with anti-HIST1H3A antibodies, the following protocol parameters are recommended:
Sample preparation:
Gel electrophoresis:
Antibody dilution and incubation:
Detection methods:
HRP-conjugated secondary antibodies with enhanced chemiluminescence (ECL) detection
Expected molecular weight band: approximately 15-17 kDa
| Sample Type | Protein Loading | Gel Percentage | Antibody Dilution | Blocking Solution | Incubation Time |
|---|---|---|---|---|---|
| Cell lysates | 20-40μg | 15-18% | 1:500-1:1000 | 5% BSA in TBST | 16h at 4°C |
| Tissue extracts | 30-50μg | 15-18% | 1:500 | 5% BSA in TBST | 16h at 4°C |
| Purified histones | 2-5μg | 15% | 1:1000 | 3% BSA in TBST | 2h at RT or 16h at 4°C |
For immunohistochemistry applications with anti-HIST1H3A antibodies, follow these optimized protocol guidelines:
Tissue preparation:
Fix tissues in 10% neutral buffered formalin
Embed in paraffin and section at 4-6μm thickness
For phospho-specific epitopes, minimize dephosphorylation by rapid processing
Antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes
Alternative: Tris-EDTA buffer (pH 9.0) for some applications
Antibody dilution and incubation:
Detection system:
Use a polymer-HRP based detection system for optimal sensitivity
DAB (3,3'-diaminobenzidine) substrate for visualization
Counterstain with hematoxylin
Controls:
Investigating the role of H3T45 phosphorylation in apoptosis requires sophisticated experimental approaches:
Time-course analysis during apoptosis:
Induce apoptosis using established triggers (e.g., staurosporine, TNF-α with cycloheximide)
Collect cells at regular intervals (0, 1, 2, 4, 8, 12, 24 hours)
Perform western blotting with anti-H3T45ph antibody to track phosphorylation dynamics
In parallel, measure apoptotic markers (cleaved caspase-3, PARP cleavage) to correlate with H3T45ph timing
Chromatin immunoprecipitation (ChIP) during apoptosis:
Perform ChIP-seq with anti-H3T45ph antibody at key timepoints during apoptosis
Analyze genomic distribution of H3T45ph to identify regions affected early in apoptosis
Correlate with gene expression changes using RNA-seq
Identify transcription factors that co-localize with H3T45ph regions
Mass spectrometry-based quantification:
Inhibitor studies:
This multi-method approach enables comprehensive understanding of how H3T45 phosphorylation contributes to apoptotic processes.
Identifying the enzymatic regulators of H3T45 phosphorylation requires systematic screening approaches:
Kinase inhibitor screening:
Treat cells with a panel of selective kinase inhibitors
Measure H3T45ph levels by western blotting
Narrow down candidate kinase families based on inhibitor specificity
Validate using genetic approaches (siRNA, CRISPR-Cas9)
In vitro kinase assays:
Express and purify recombinant H3 as substrate
Test candidate kinases in vitro with γ-32P-ATP or use antibody-based detection
Confirm specificity using H3 variants with T45A mutation
Determine kinetic parameters (Km, Vmax) for positive hits
Phosphatase identification:
Treat cells with phosphatase inhibitors of varying specificity
Analyze H3T45ph levels to identify phosphatase families involved
Perform phosphatase substrate-trapping experiments
Validate candidates using phosphatase overexpression and knockdown
Proximity labeling proteomics:
Generate H3.1-BioID or H3.1-TurboID fusion constructs
Identify proteins in proximity to H3.1 using streptavidin pulldown and mass spectrometry
Focus on kinases/phosphatases among proximity interactors
Compare interactomes between wild-type and T45A mutant H3.1
These approaches collectively provide a comprehensive strategy for identifying the enzymatic regulators of H3T45 phosphorylation.
Investigating potential crosstalk between H3T45 phosphorylation and the Cys96-mediated redox sensing function of H3.1 represents an advanced research question that integrates multiple histone functions:
Simultaneous modification analysis:
Treat cells with oxidative stress inducers (H₂O₂, menadione)
Analyze both H3T45ph and Cys96 oxidation status using antibodies and mass spectrometry
Determine if these modifications co-occur on the same histone molecules
Analyze temporal dynamics to establish potential sequential ordering
Structural impact analysis:
Perform molecular dynamics simulations of nucleosomes with various modification states:
Unmodified H3.1
H3T45ph only
Cys96 oxidation only
Both modifications simultaneously
Analyze impacts on nucleosome stability and DNA accessibility
Mutational studies:
Generate cell lines expressing H3.1 variants:
Wild-type
T45A (phospho-null)
T45E (phospho-mimetic)
C96S (redox-null)
Combined mutations
Analyze cellular responses to oxidative stress and apoptotic stimuli
Perform ChIP-seq to map genomic distribution changes
Chromatin exchange dynamics:
Investigate whether H3T45 phosphorylation affects the H3.1-to-H3.3 exchange process that occurs following Cys96 oxidation
Use FRAP (Fluorescence Recovery After Photobleaching) with fluorescently tagged histones to measure exchange rates
Determine if phosphatase inhibition (to increase H3T45ph) affects oxidation-induced histone variant exchange
This integrated approach would illuminate potential functional connections between these two distinct regulatory modifications in H3.1.
Non-specific binding is a common challenge when working with histone antibodies. Here are the most frequent causes and their solutions:
Cross-reactivity with other H3 variants:
Problem: High sequence homology between H3 variants leads to cross-reactivity
Solution: Use peptide competition assays with specific peptides from different H3 variants
Validation: Test antibody against recombinant H3.1, H3.2, and H3.3 proteins
Epitope masking by nearby modifications:
Insufficient blocking:
Problem: High background due to inadequate blocking
Solution: Extend blocking time (2-3 hours) and optimize blocking agent (5% BSA often works better than milk for phospho-epitopes)
Alternative: Test synthetic blocking peptides specific to the antibody epitope region
Sample preparation issues:
Problem: Dephosphorylation during sample preparation
Solution: Include phosphatase inhibitors in all buffers and keep samples cold
Optimization: Minimize time between sample collection and fixation/extraction
| Issue | Likely Cause | Diagnostic Test | Solution |
|---|---|---|---|
| No signal | Epitope degradation | Test with positive control sample | Include phosphatase inhibitors |
| Multiple bands | Cross-reactivity | Peptide competition | Use more stringent washing |
| High background | Insufficient blocking | Test different blocking agents | Increase blocking time, optimize antibody dilution |
| Inconsistent results | Batch variability | Test antibody lot with standard sample | Purchase larger antibody lots when possible |
Optimizing ChIP-seq experiments for H3T45ph requires specific adjustments to standard protocols:
Crosslinking optimization:
Test multiple formaldehyde concentrations (0.5-2%) and incubation times (5-20 minutes)
For some applications, try dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde
Ensure rapid quenching with glycine to prevent over-crosslinking
Chromatin fragmentation:
Sonication parameters must be carefully optimized for histone PTM ChIP
Target fragment size: 150-300bp (slightly smaller than standard ChIP)
Verify fragmentation efficiency by agarose gel electrophoresis
Consider using micrococcal nuclease (MNase) digestion instead of or in addition to sonication
Antibody specificity measures:
Pre-clear chromatin with protein A/G beads to reduce non-specific binding
Include IgG control and input normalization
Include spike-in controls with chromatin from another species
Perform parallel ChIP with total H3 antibody for normalization
Washing stringency:
Increase salt concentration in wash buffers (up to 500mM NaCl) for more stringent washing
Add non-ionic detergents (0.1% NP-40 or Triton X-100) to reduce non-specific binding
Increase number of washes (5-6 washes) to improve signal-to-noise ratio
Library preparation considerations:
Start with more immunoprecipitated material than standard ChIP-seq
Minimize PCR cycles during library amplification to reduce bias
Include library preparation controls to ensure quality
These optimizations collectively enhance the signal-to-noise ratio in H3T45ph ChIP-seq experiments, enabling more accurate mapping of this modification across the genome.
H3T45 phosphorylation exists within a complex landscape of histone modifications that change during the cell cycle. Understanding these correlations requires integrated analysis:
Cell synchronization approaches:
Synchronize cells at different cell cycle phases:
G1/S boundary: Double thymidine block
S phase: Thymidine-aphidicolin sequential block
G2/M: Nocodazole treatment
Collect synchronized populations for histone PTM profiling
Multi-modification analysis:
Perform Western blotting with antibodies against multiple PTMs (H3T45ph, H3S10ph, H3K9me3, H3K27me3)
Use MS/MS to quantify modification levels on the same histone peptides
Calculate correlation coefficients between modification levels
Determine if specific modifications tend to co-occur or are mutually exclusive
Immunofluorescence co-localization:
Perform dual immunofluorescence staining for H3T45ph and other modifications
Use confocal microscopy to assess spatial co-localization
Quantify degree of overlap using Pearson's correlation coefficient
Sequential ChIP (Re-ChIP):
Perform ChIP with anti-H3T45ph antibody followed by a second ChIP with antibodies against other modifications
Identify genomic regions containing both modifications
Compare with single-modification ChIP profiles
This integrated approach reveals how H3T45 phosphorylation fits into the broader epigenetic landscape across cell cycle phases.
Cancer cells often exhibit altered histone modification patterns. Understanding H3T45ph in this context has important implications:
Cancer tissue analysis:
Relationship to oncogenic signaling:
Drug resistance mechanisms:
Therapeutic targeting strategies:
Identify druggable enzymes that regulate H3T45ph
Test combination treatments targeting both H3T45ph regulatory pathways and standard chemotherapies
Develop phospho-H3T45 levels as potential biomarkers for treatment response
This research direction connects basic epigenetic mechanisms to potential clinical applications in cancer diagnosis and treatment.
Several cutting-edge technologies show particular promise for advancing our understanding of H3T45 phosphorylation and other HIST1H3A modifications:
Single-cell epigenomics:
Single-cell ChIP-seq and CUT&Tag approaches enable analysis of H3T45ph heterogeneity
Integration with single-cell transcriptomics reveals functional consequences at individual cell level
Spatial epigenomics techniques allow mapping histone modifications within tissue architecture
Live-cell imaging of histone dynamics:
FRET-based biosensors for real-time monitoring of H3T45 phosphorylation
Engineered antibody fragments (Fabs) for tracking histone modifications in living cells
Correlation with chromatin accessibility measured by live-cell DNA-binding dyes
Cryo-EM of modified nucleosomes:
High-resolution structural analysis of nucleosomes containing H3T45ph
Investigation of structural impacts on DNA-histone interactions
Comparison with other core modifications to build comprehensive structural models
CRISPR epigenome editing:
Targeted modulation of H3T45 phosphorylation at specific genomic loci
Development of CRISPR-based readers and writers for T45 phosphorylation
Precise control of modification timing using optogenetic or chemical induction approaches
These emerging technologies will provide unprecedented insights into the dynamic regulation and functional consequences of H3T45 phosphorylation in diverse biological contexts.
Despite significant advances, several fundamental questions about H3T45 phosphorylation remain unanswered:
Enzymatic regulation:
What are the specific kinases and phosphatases that regulate H3T45 phosphorylation?
How is their activity coordinated across different cellular contexts?
What upstream signaling pathways control these enzymes?
Functional consequences:
How does H3T45 phosphorylation directly affect nucleosome structure and stability?
What is the relationship between H3T45ph and DNA accessibility?
Does H3T45ph coordinate with other histone modifications to form a "histone code"?
Evolutionary conservation:
Is the regulation and function of H3T45 phosphorylation conserved across species?
How does this modification contribute to species-specific chromatin functions?
What are the evolutionary drivers for maintaining this site of regulation?
Disease relevance:
How do disease-associated mutations in H3.1 affect T45 phosphorylation?
Can H3T45ph serve as a biomarker for specific pathological conditions?
Is targeted modulation of this modification a viable therapeutic strategy?
Addressing these questions will require interdisciplinary approaches combining structural biology, biochemistry, genomics, and cell biology to fully understand the biological significance of H3T45 phosphorylation.