HIST1H1E (Histone Cluster 1 H1e) is a member of the H1 histone family, which stabilizes chromatin structure by binding linker DNA between nucleosomes. Acetylation at lysine 33 (K33) modulates chromatin accessibility and transcriptional regulation .
Property | Detail |
---|---|
UniProt ID | P10412 |
Protein Mass | ~21.9 kDa |
Localization | Nucleus, chromosomes |
Biological Role | Chromatin condensation, transcriptional regulation, DNA methylation |
The Acetyl-HIST1H1E (K33) Antibody is a rabbit-derived polyclonal IgG antibody validated for specificity and performance in multiple assays .
The antibody demonstrates robust performance across platforms:
Immunofluorescence (IF): Clear nuclear staining in MCF-7 cells, enhanced with sodium butyrate treatment .
ChIP: Effectively enriches acetylated HIST1H1E-bound chromatin fragments .
Specificity: No cross-reactivity with non-acetylated HIST1H1E or other histones .
Chromatin Dynamics: Mapping acetylation sites in chromatin remodeling studies .
Gene Regulation: Investigating histone acetylation's role in transcription .
Disease Models: Epigenetic studies in cancer, neurodegeneration, and developmental disorders.
Application | Recommended Dilution | Validation Source |
---|---|---|
ICC/IF | 1:20–1:200 | MCF-7 cell lines |
ChIP | 1:10–1:50 | Chromatin extracts |
ELISA | 1:1000–1:5000 | Peptide-coated plates |
Multiple vendors offer this antibody with slight variations in formulation and pricing:
Supplier | Product Code | Size | Price | Conjugate |
---|---|---|---|---|
Cusabio | CSB-PA010380PA33acHU | 50–100 µl | $200–$319 | Unconjugated |
Biomatik | CAC15245 | 50 µl | $225 | Unconjugated |
Qtonics | QA24755 | 50–100 µl | $200–$350 | Unconjugated |
Acetylation at K33 is associated with transcriptional activation and chromatin decompaction. This antibody enables:
Epigenetic Profiling: Correlating acetylation patterns with gene expression .
Drug Discovery: Screening for histone acetyltransferase (HAT) or deacetylase (HDAC) inhibitors .
Acetyl-HIST1H1E (K33) Antibody is a polyclonal antibody that specifically recognizes the acetylation of lysine 33 on histone H1.4 (HIST1H1E). This antibody targets a peptide sequence surrounding the acetylated lysine 33 site derived from human Histone H1.4 protein . The antibody enables researchers to investigate specific post-translational modifications (PTMs) on this important histone variant, allowing for detailed analysis of epigenetic regulation mechanisms in chromatin structure and gene expression .
Acetyl-HIST1H1E (K33) Antibody has been validated for multiple experimental applications critical to epigenetic research, including ELISA (Enzyme-Linked Immunosorbent Assay), ICC (Immunocytochemistry), IF (Immunofluorescence), and ChIP (Chromatin Immunoprecipitation) . The antibody is particularly valuable in ChIP experiments, where it can identify genomic regions associated with this specific histone modification, enabling researchers to map acetylation patterns across the genome and correlate them with gene expression profiles .
Specificity is crucial when studying histone modifications because each specific modification represents a unique signal for gene expression regulation. Different modifications, even on the same residue, can have dramatically different biological outcomes. For example, while studying one histone mark like H3K9, acetylation (H3K9ac) is associated with gene activation, whereas methylation of the same residue (H3K9me) can signal gene repression . The specificity of antibodies like Acetyl-HIST1H1E (K33) ensures that researchers can distinguish between different modification states, preventing erroneous interpretation of epigenetic signals .
Histone H1.4 (HIST1H1E) functions as a linker histone that binds to DNA between nucleosomes, forming the higher-order chromatin fiber structure. Acetylation at K33 likely affects the protein's interaction with DNA, potentially loosening chromatin structure. This modification plays a role in regulating gene transcription through chromatin remodeling, nucleosome spacing, and influencing DNA methylation patterns . The Acetyl-HIST1H1E (K33) mark therefore represents a specific epigenetic signal that researchers can use to investigate dynamic changes in chromatin accessibility and gene regulation.
While the common histone modifications on core histones (H2A, H2B, H3, H4) have been extensively studied, the specific functions of linker histone H1 modifications remain less characterized. Acetylation of HIST1H1E at K33 occurs in a region distinct from other acetylation sites like K16, K51, and K63, each potentially having different impacts on chromatin structure . Current research suggests that K33 acetylation may uniquely affect the globular domain interactions of H1.4 with the nucleosome, potentially altering chromatin compaction in a manner distinct from modifications at other lysine residues . Comparative studies using antibodies against different acetylation sites (such as HIST1H1E K16, K51, and K63) are necessary to fully elucidate these functional differences.
Cross-reactivity is a significant concern when using histone modification antibodies. For accurate interpretation of results, researchers must confirm that the Acetyl-HIST1H1E (K33) Antibody does not react with other acetylated lysines on HIST1H1E or similar motifs on other histone variants . Cross-reactivity validation requires testing the antibody against peptide arrays containing different acetylated histone sequences, similar to the ELISA approach demonstrated for H3K9me2 specificity testing . When designing experiments with this antibody, researchers should include appropriate controls, such as peptide competition assays or samples where the modification has been enzymatically removed, to confirm signal specificity.
The Acetyl-HIST1H1E (K33) Antibody can be employed in sequential ChIP (Re-ChIP) experiments to investigate the co-occurrence of this acetylation mark with other histone modifications or chromatin-associated proteins. This approach involves performing ChIP with the Acetyl-HIST1H1E (K33) Antibody, followed by a second immunoprecipitation using antibodies against other modifications . Additionally, combining ChIP-seq data using this antibody with other epigenomic datasets (DNA methylation, chromatin accessibility, other histone marks) can reveal correlations between HIST1H1E K33 acetylation and broader epigenetic landscapes. Such integrative analyses can elucidate how this specific modification interacts with other epigenetic mechanisms to regulate gene expression and chromatin structure .
For optimal ChIP experiments with Acetyl-HIST1H1E (K33) Antibody, researchers should follow these methodological guidelines:
Sample preparation: Cross-link protein-DNA complexes using 1% formaldehyde for 10 minutes at room temperature, followed by quenching with glycine.
Chromatin preparation: Sonicate chromatin to fragments of approximately 200-500 bp.
Immunoprecipitation: Use the antibody at optimized concentrations (starting with manufacturer recommendations) and incubate overnight at 4°C .
Washing and elution: Perform stringent washing steps to remove non-specific binding.
Cross-link reversal and DNA purification: Reverse cross-links at 65°C and purify DNA for downstream analysis.
For high-quality results, it's critical to validate the antibody's specificity and optimize conditions for each cell type or tissue being studied . Including appropriate controls, such as IgG and input chromatin, is essential for accurate data interpretation.
Based on manufacturer specifications, the following dilutions are recommended for Acetyl-HIST1H1E (K33) Antibody:
Application | Recommended Dilution | Optimization Notes |
---|---|---|
ICC | 1:20 - 1:200 | Start with 1:100 and adjust based on signal intensity |
IF | 1:50 - 1:200 | Lower dilutions may be needed for weakly expressed targets |
ChIP | Varies by protocol | Typically 2-5 μg per ChIP reaction |
ELISA | Varies by protocol | Titration required for optimal signal-to-noise ratio |
These recommendations serve as starting points and should be optimized for specific experimental conditions . Storage conditions (-20°C or -80°C) must be maintained to preserve antibody activity, and repeated freeze-thaw cycles should be avoided .
While not specifically mentioned for this antibody in the search results, Western blotting for histone modifications requires special considerations:
Sample preparation: Use acid extraction methods (e.g., 0.2N HCl or 0.4N H₂SO₄) to effectively isolate histones.
Gel selection: Use high-percentage (15-18%) or specialized gradient gels that resolve low molecular weight proteins (HIST1H1E is approximately 22 kDa).
Transfer conditions: Optimize transfer conditions for small proteins, potentially using PVDF membranes and adding SDS to transfer buffer.
Blocking: Use 5% BSA rather than milk to block membranes, as milk contains proteins that can cross-react with phospho-specific antibodies.
Antibody incubation: Incubate with Acetyl-HIST1H1E (K33) Antibody overnight at 4°C with gentle rocking .
Including proper controls, such as unmodified histone H1.4 and loading controls, is essential for accurate interpretation of Western blot results.
When encountering issues with non-specific binding or weak signals, consider the following troubleshooting approaches:
For non-specific binding:
Increase washing stringency (higher salt concentration or detergent)
Pre-clear lysates with protein A/G beads before immunoprecipitation
Optimize blocking conditions with different blocking agents
Validate antibody specificity using peptide competition assays
For weak signals:
Increase antibody concentration within recommended ranges
Extend incubation time (overnight at 4°C)
Ensure proper sample preparation to expose epitopes
Verify that the target modification is present in your experimental conditions
Optimize detection methods (enhanced chemiluminescence for Western blot or amplification systems for immunofluorescence)
Analyzing multiple biological replicates and quantifying signal intensities relative to appropriate controls can help distinguish genuine signals from background noise.
For robust quantitative analysis of ChIP-seq data using Acetyl-HIST1H1E (K33) Antibody:
Generate high-quality sequencing data with sufficient depth (minimum 20 million uniquely mapped reads).
Include input controls and IgG controls for normalization and background subtraction.
Use specialized ChIP-seq analysis pipelines (e.g., MACS2, HOMER) for peak calling, with parameters optimized for histone modifications (broader peaks than transcription factors).
Validate key findings using orthogonal methods such as ChIP-qPCR.
Analyze the genomic distribution of Acetyl-HIST1H1E (K33) signal relative to gene features (promoters, enhancers, gene bodies).
Integrate with gene expression data to correlate acetylation patterns with transcriptional outcomes.
Consider differential binding analysis across conditions to identify biologically significant changes in acetylation .
Visualization tools like IGV or UCSC Genome Browser can help interpret peak distributions, while pathway analysis of genes associated with Acetyl-HIST1H1E (K33) peaks can reveal biological processes regulated by this modification.
To validate the specificity of ChIP signals:
Perform peptide competition assays: Pre-incubate the antibody with acetylated and non-acetylated peptides before ChIP to demonstrate specific blocking with the acetylated peptide only.
Use genetic approaches: Employ cells with mutations at the K33 site (K33R) that prevent acetylation or knockdown/knockout of the acetyltransferases responsible for K33 acetylation.
Employ histone deacetylase (HDAC) inhibitors: Treatment with HDAC inhibitors should increase the ChIP signal if it genuinely represents acetylation.
Perform sequential ChIP: Combining ChIP with antibodies recognizing the unmodified H1.4 backbone followed by Acetyl-HIST1H1E (K33) Antibody can confirm signal specificity.
Compare ChIP-seq profiles: Correlation with datasets for other active chromatin marks can support the biological relevance of identified binding sites .
Proper validation ensures that experimental findings accurately reflect the biological distribution of the K33 acetylation mark rather than antibody artifacts.
Acetyl-HIST1H1E (K33) Antibody can be deployed in time-course ChIP-seq experiments to map dynamic changes in this histone modification during cellular differentiation processes. By collecting samples at key differentiation timepoints and performing ChIP-seq, researchers can generate temporal maps of K33 acetylation changes that correlate with developmental gene expression programs. This approach can reveal how HIST1H1E acetylation contributes to cell fate decisions through altered chromatin accessibility at lineage-specific genes . The data can be integrated with other epigenetic marks and transcription factor binding patterns to construct comprehensive models of the epigenetic reorganization driving differentiation.
Studying Acetyl-HIST1H1E (K33) in disease models, particularly cancer and neurodegenerative disorders, can provide valuable insights into disease mechanisms:
In cancer research: Aberrant histone acetylation patterns are hallmarks of many cancers. ChIP-seq using Acetyl-HIST1H1E (K33) Antibody can identify genome-wide changes in this modification between normal and cancer cells, potentially revealing novel oncogenic mechanisms related to altered chromatin structure .
In neurodegenerative disorders: Changes in histone acetylation have been implicated in conditions like Alzheimer's and Huntington's diseases. Mapping K33 acetylation patterns in disease models can reveal how epigenetic dysregulation contributes to pathogenesis.
In drug development: Monitoring changes in Acetyl-HIST1H1E (K33) patterns following treatment with epigenetic modulators (HDAC inhibitors, HAT inhibitors) can help evaluate drug efficacy and mechanism of action .
These applications require careful experimental design with appropriate disease models and controls to establish causative relationships between acetylation changes and disease phenotypes.
Several cutting-edge techniques can be combined with Acetyl-HIST1H1E (K33) Antibody research:
CUT&RUN and CUT&Tag: These techniques offer higher resolution and lower background than traditional ChIP, requiring fewer cells and less antibody while providing more precise mapping of modification sites.
Single-cell ChIP-seq: Analyzing K33 acetylation patterns at the single-cell level can reveal cell-to-cell heterogeneity in epigenetic regulation within seemingly homogeneous populations.
CRISPR-based epigenome editing: Coupling dCas9-acetyltransferase fusions with Acetyl-HIST1H1E (K33) ChIP allows researchers to establish causal relationships between site-specific K33 acetylation and functional outcomes.
Proximity labeling: BioID or APEX2 fusions to histone modifying enzymes, combined with Acetyl-HIST1H1E (K33) ChIP, can identify proteins associated with this modification.
Mass spectrometry-ChIP: Combining ChIP using Acetyl-HIST1H1E (K33) Antibody with mass spectrometry enables identification of protein complexes associated with K33-acetylated regions .