This antibody is utilized to investigate:
Chromatin Dynamics: Linker histones like H1.4 stabilize nucleosome structures and regulate DNA accessibility . Phosphorylation at Ser35 may modulate chromatin compaction during mitosis or transcriptional activation.
Disease Mechanisms: HIST1H1E mutations cause Rahman syndrome, a neurodevelopmental disorder . While this antibody does not target disease-associated variants, it aids in studying wild-type H1.4 regulation.
Epigenetic Signaling: PTMs of H1.4 influence interactions with chromatin remodelers and DNA repair proteins .
Immunofluorescence: Localizes phosphorylated H1.4 to heterochromatin regions in HeLa cells .
Western Blot: Detects a single band at ~22 kDa (predicted molecular weight: 21,865 Da) .
Phospho-HIST1H1E (S35) Antibody is pivotal for dissecting:
HIST1H1E (Histone H1.4) is a linker histone that binds to DNA between nucleosomes to form higher-order chromatin structures. Phosphorylation at serine 35 (S35) is a post-translational modification that plays a crucial role in regulating chromatin dynamics, particularly during cell cycle progression and transcriptional regulation. The phosphorylation status at S35 has been linked to changes in chromatin compaction and accessibility for transcription factors, making it an important epigenetic marker for studies of gene expression regulation .
The Phospho-HIST1H1E (S35) antibody is validated for multiple applications including:
ELISA (Enzyme-Linked Immunosorbent Assay)
ICC (Immunocytochemistry)
IF (Immunofluorescence)
Recommended dilutions for optimal results are:
ICC: 1:20-1:200
IF: 1:50-1:200
These applications allow researchers to detect and quantify phosphorylated H1.4 in various experimental settings, providing insights into epigenetic regulation mechanisms .
This polyclonal antibody is generated against a peptide sequence surrounding the phosphorylated serine 35 site of human Histone H1.4 (UniProt ID: P10412). It specifically recognizes the phosphorylated form of HIST1H1E at Ser35, enabling researchers to distinguish between the phosphorylated and non-phosphorylated states of the protein. This specificity is critical for studies investigating the functional consequences of this post-translational modification in various cellular processes .
For optimal immunofluorescence results with Phospho-HIST1H1E (S35) antibody:
Sample preparation:
Fixation: Use 4% paraformaldehyde for 15 minutes at room temperature
Permeabilization: 0.2% Triton X-100 for 10 minutes
Blocking: 5% BSA in PBS for 1 hour
Antibody incubation:
Primary antibody: Use Phospho-HIST1H1E (S35) at 1:50-1:200 dilution overnight at 4°C
Secondary antibody: Anti-rabbit IgG conjugated with fluorophore at 1:500 dilution for 1 hour at room temperature
Controls to include:
Negative control: Secondary antibody only
Dephosphorylation control: Treat samples with phosphatase before antibody incubation
Cell cycle synchronization: Compare G1, S, and G2/M phases as phosphorylation levels may vary
Counterstaining:
Effective nuclear protein extraction for HIST1H1E studies requires careful fractionation to maintain phosphorylation status:
Cell harvesting and lysis:
Collect cells by gentle trypsinization
Wash with ice-cold PBS containing phosphatase inhibitors (10mM NaF, 1mM Na3VO4)
Resuspend in hypotonic buffer (10mM HEPES pH 7.9, 10mM KCl, 1.5mM MgCl2, 0.34M sucrose, 10% glycerol, 1mM DTT, protease and phosphatase inhibitors)
Add Triton X-100 to 0.1% final concentration
Incubate on ice for 8 minutes
Nuclear isolation:
Centrifuge at 1,300×g for 5 minutes at 4°C
Wash nuclear pellet once with hypotonic buffer
Resuspend in high-salt extraction buffer (20mM HEPES pH 7.9, 420mM NaCl, 1.5mM MgCl2, 0.2mM EDTA, 25% glycerol, protease and phosphatase inhibitors)
Incubate on ice for 30 minutes with periodic vortexing
Quality control:
This protocol helps preserve the phosphorylation status of HIST1H1E for subsequent analysis while minimizing contamination from other cellular compartments.
Optimizing ChIP for Phospho-HIST1H1E (S35) requires special considerations:
Crosslinking optimization:
Use 1% formaldehyde for 10 minutes at room temperature
For linker histones, consider dual crosslinking with 1.5mM EGS (ethylene glycol bis[succinimidylsuccinate]) for 30 minutes before formaldehyde
Chromatin fragmentation:
Sonicate to generate fragments of 200-500bp
Verify fragmentation by agarose gel electrophoresis
Immunoprecipitation:
Pre-clear chromatin with protein A/G beads
Use 3-5μg of Phospho-HIST1H1E (S35) antibody per 25μg of chromatin
Include phosphatase inhibitors (10mM NaF, 1mM Na3VO4) in all buffers
Extended incubation (overnight at 4°C) with gentle rotation
Washing stringency:
Perform sequential washes with increasing salt concentration
Include a LiCl wash to reduce non-specific binding
Controls:
Input chromatin (10%)
IgG negative control
Positive control using antibody against unmodified HIST1H1E
DNA purification and analysis:
Phosphorylation of HIST1H1E at S35 exhibits distinct patterns throughout the cell cycle:
| Cell Cycle Phase | Expected Phospho-HIST1H1E (S35) Pattern | Biological Significance |
|---|---|---|
| G0/G1 | Low levels, primarily at heterochromatic regions | Maintenance of stable chromatin structure |
| S phase | Gradual increase, appearing at replication foci | Facilitates chromatin decompaction for DNA replication |
| G2 | High levels, broadly distributed | Prepares chromatin for mitotic condensation |
| Mitosis | Maximum levels, associated with condensed chromosomes | Essential for proper chromosome condensation |
| Cytokinesis | Rapid dephosphorylation | Reestablishment of interphase chromatin structure |
When interpreting changes in Phospho-HIST1H1E (S35) signals:
Temporal context: Always consider the cell cycle stage of your samples
Spatial distribution: Note whether signals are broadly nuclear or localized to specific domains
Co-localization analysis: Compare with other cell cycle markers (e.g., Ki67, PCNA)
Quantitative assessment: Measure signal intensity relative to total HIST1H1E levels
Perturbation responses: Evaluate how treatments affecting CDKs or other kinases alter the phosphorylation pattern
Non-specific binding issues can compromise data quality when working with phospho-specific antibodies:
| Issue | Possible Causes | Solutions |
|---|---|---|
| High background signal | Insufficient blocking | Extend blocking time to 2 hours; use 5% BSA or 5% milk in TBS-T |
| Excessive antibody concentration | Perform titration experiments to determine optimal dilution | |
| Cross-reactivity with other phosphorylated histones | Pre-absorb antibody with non-phosphorylated peptide | |
| Cytoplasmic staining | Sample over-permeabilization | Reduce Triton X-100 concentration to 0.1% |
| Phosphatase activity during sample preparation | Double phosphatase inhibitor concentration | |
| Loss of signal over time | Repeated freeze-thaw cycles | Aliquot antibody upon receipt and store at -80°C |
| Improper storage | Follow manufacturer's storage recommendations (avoid repeated freeze) | |
| Inconsistent results between experiments | Variability in fixation conditions | Standardize fixation time and temperature |
| Cell culture conditions affecting phosphorylation | Control for cell density and serum levels |
For best results, always run appropriate controls, including:
Phosphatase-treated samples
Competitive blocking with phosphopeptide
When faced with discrepancies between antibody-based detection and mass spectrometry (MS) results for Phospho-HIST1H1E (S35):
Analytical considerations:
Antibody specificity: Verify using phospho-null mutants (S35A) and phospho-mimetic mutants (S35D/E)
MS sample preparation: Ensure preservation of phosphorylation using appropriate extraction buffers with phosphatase inhibitors
MS ionization efficiency: Phosphopeptides often have lower ionization efficiency; consider phospho-enrichment steps
Biological variables:
Dynamic range: MS may not detect low-abundance phosphorylation that antibodies can detect
Epitope accessibility: In some contexts, the S35 phosphorylation site may be masked in complexes
Localized vs. global changes: Antibody-based imaging detects localized changes that may be diluted in whole-cell MS samples
Resolution strategies:
Perform fractionation to enrich for HIST1H1E before MS analysis
Use parallel reaction monitoring (PRM) for targeted MS quantification
Apply phosphopeptide enrichment (TiO2 or IMAC) before MS
Validate with orthogonal methods like Phos-tag gels
Control for potential confounding factors like cell cycle synchronization
Investigating the relationship between HIST1H1E phosphorylation and chromatin remodeling requires multi-modal approaches:
Chromatin accessibility analysis:
Combine Phospho-HIST1H1E (S35) ChIP-seq with ATAC-seq or DNase-seq
Compare phosphorylated vs. non-phosphorylated HIST1H1E distribution relative to accessible regions
Analyze temporal changes following signaling pathway activation
Nucleosome dynamics assessment:
Perform MNase-seq in conjunction with Phospho-HIST1H1E (S35) ChIP
Map nucleosome positioning changes relative to phosphorylation status
Use live-cell imaging with fluorescently tagged HIST1H1E phospho-mimetics
Protein interaction studies:
Conduct Phospho-HIST1H1E (S35) antibody-based co-IP followed by mass spectrometry
Identify differential interactors between phosphorylated and non-phosphorylated forms
Validate key interactions using proximity ligation assays
Functional genomics approaches:
Studying Phospho-HIST1H1E (S35) in development and differentiation requires specialized approaches:
Temporal sampling strategy:
Define critical developmental timepoints for analysis
Consider both rapid transitions (hours) and long-term changes (days)
Include matched controls at each timepoint
Cell heterogeneity management:
Implement single-cell approaches (CyTOF, scRNA-seq with protein detection)
Use FACS to isolate pure populations based on differentiation markers
Apply tissue-specific markers for in vivo studies
Quantification methods:
Develop ratiometric imaging approaches (Phospho-HIST1H1E/total HIST1H1E)
Use in situ proximity ligation assays for tissue samples
Apply phospho-flow cytometry for quantitative population analysis
Perturbation strategies:
Design stage-specific inhibition of relevant kinases/phosphatases
Create conditional phospho-mutant models using CRISPR/Cas9
Utilize inducible expression systems for temporal control
Data integration approaches:
Investigating cross-talk between HIST1H1E phosphorylation and other epigenetic modifications requires sophisticated experimental designs:
Sequential ChIP (Re-ChIP) approach:
First round: Immunoprecipitate with Phospho-HIST1H1E (S35) antibody
Second round: IP with antibodies against other histone modifications (H3K27me3, H3K4me3, etc.)
Compare enrichment patterns to single ChIP results
Include appropriate controls for each round of IP
Proximity-based detection methods:
Proximity ligation assays (PLA) to detect co-occurrence of modifications
FRET-based approaches using labeled antibodies
Mass spectrometry of intact nucleosomes to identify co-occurring modifications
Perturbation studies:
Selective inhibition of writers/erasers of specific modifications
Monitor consequent changes in HIST1H1E phosphorylation
Track reciprocal effects on other modifications when S35 phosphorylation is manipulated
Genomic distribution analysis:
Generate genome-wide maps of Phospho-HIST1H1E (S35) and other modifications
Perform correlation analyses at different genomic features
Identify domains with synergistic or antagonistic modification patterns
Functional readouts:
For accurate quantification of Phospho-HIST1H1E (S35) levels:
Western blot quantification:
Always normalize phospho-signal to total HIST1H1E
Include standard curves using recombinant phosphorylated protein
Use infrared or chemiluminescent detection systems with verified linear range
Apply appropriate statistical tests for multiple comparisons
Immunofluorescence quantification:
Collect images under identical acquisition parameters
Measure nuclear intensity in at least 100 cells per condition
Use automated segmentation to eliminate selection bias
Apply background subtraction using nuclear-free areas
Express results as phospho/total HIST1H1E ratio
Flow cytometry approaches:
Fix cells with 4% paraformaldehyde followed by 90% methanol permeabilization
Perform dual staining for phospho and total HIST1H1E
Include isotype and phosphatase-treated controls
Gate on specific cell populations if analyzing heterogeneous samples
ELISA-based quantification:
A comprehensive ChIP-seq protocol for Phospho-HIST1H1E (S35):
Sample preparation:
Crosslink cells with 1% formaldehyde for 10 minutes
Consider dual crosslinking for linker histones (1.5mM EGS for 30 minutes before formaldehyde)
Quench with 125mM glycine
Isolate nuclei using hypotonic lysis
Chromatin preparation:
Sonicate chromatin to 200-500bp fragments
Include phosphatase inhibitors in all buffers
Verify fragmentation by agarose gel electrophoresis
Pre-clear chromatin with protein A/G beads
Immunoprecipitation:
Use 5μg Phospho-HIST1H1E (S35) antibody per 25μg chromatin
Include appropriate controls (IgG, input, total HIST1H1E)
Incubate overnight at 4°C with rotation
Wash with progressively stringent buffers
Library preparation considerations:
Repair DNA ends and adapt for sequencing
Size select for fragments of 150-300bp
Use minimum PCR cycles to avoid amplification bias
Include spike-in controls for normalization
Data analysis pipeline:
To validate Phospho-HIST1H1E (S35) antibody specificity:
Genetic approaches:
Generate HIST1H1E knockout cells as negative controls
Create S35A mutant (non-phosphorylatable) and S35D/E (phospho-mimetic) constructs
Compare antibody reactivity across these genetic models
Biochemical validations:
Perform peptide competition assays using phosphorylated and non-phosphorylated peptides
Treat samples with lambda phosphatase to remove phosphorylation
Use CDK inhibitors to reduce S35 phosphorylation in vivo
Perform immunodepletion experiments
Orthogonal methods:
Compare results with mass spectrometry
Use Phos-tag gel electrophoresis to separate phosphorylated species
Validate with multiple antibodies targeting the same modification
Compare reactivity across cell cycle stages (phosphorylation increases during mitosis)
Cross-reactivity assessments:
Phospho-HIST1H1E (S35) antibody can provide valuable insights into cancer epigenetics:
Comparative profiling approaches:
Compare phosphorylation patterns between tumor and adjacent normal tissues
Analyze phosphorylation across cancer progression stages
Correlate with clinical outcomes and treatment responses
Mechanistic studies:
Investigate altered CDK activity effects on HIST1H1E phosphorylation
Map phosphorylation changes to oncogene activation or tumor suppressor loss
Identify cancer-specific interaction partners of phosphorylated HIST1H1E
Functional consequences assessment:
Compare chromatin accessibility changes at cancer-relevant loci
Analyze transcriptional consequences using CRISPR-engineered phospho-mutants
Evaluate effects on DNA damage response and genomic stability
Therapeutic implications:
Monitor HIST1H1E phosphorylation changes in response to kinase inhibitors
Correlate phosphorylation status with drug resistance mechanisms
Investigate combination strategies targeting both histone modifications and signaling pathways
Biomarker development:
Studying Phospho-HIST1H1E (S35) in neurodegenerative contexts requires specialized approaches:
Tissue-specific considerations:
Use fresh-frozen tissue sections for optimal phospho-epitope preservation
Compare phosphorylation patterns in affected vs. unaffected brain regions
Consider laser capture microdissection for cell-type-specific analysis
Disease model approaches:
Compare transgenic models with human post-mortem samples
Evaluate changes in induced pluripotent stem cell (iPSC)-derived neurons
Utilize brain organoids for developmental aspects
Technical adaptations:
Optimize fixation protocols to preserve phospho-epitopes in post-mortem tissue
Use signal amplification methods for detection in small samples
Implement multiplexed IHC/IF to correlate with disease markers
Functional correlations:
Analyze relationship with DNA damage accumulation
Investigate heterochromatin maintenance
Assess impact on cellular senescence and aging pathways
Intervention strategies: