Acetyl-HIST1H1E (K51) Antibody

Shipped with Ice Packs
In Stock

Description

Immunogen and Specificity

The antibody targets a peptide sequence surrounding acetylated lysine 51 on human histone H1.4 (UniProt ID: P10412). The immunogen is derived from the acetylated form of lysine 51, ensuring specificity for this modification. This acetylation is associated with gene activation, chromatin relaxation, and transcriptional regulation .

AttributeDetails
TargetAcetyl-HIST1H1E (K51)
ImmunogenPeptide sequence around Acetyl-Lys51 of human histone H1.4
HostRabbit
Species ReactivityHuman
ClonalityPolyclonal

Applications

The antibody is validated for multiple techniques, as summarized below:

ApplicationDescription
ELISADetects acetylated histone H1.4 in lysates or purified proteins .
ImmunocytochemistryVisualizes nuclear acetylation patterns in fixed cells .
ImmunofluorescenceLocalizes acetylated histone H1.4 in cellular compartments .
ChIPIdentifies genomic regions associated with acetylated histone H1.4 .

ChIP Experiments

  • Target Validation: The antibody successfully enriched DNA from the β-globin promoter in sodium butyrate-treated HeLa cells, confirming its specificity for acetylated chromatin regions .

  • Epigenetic Studies: Demonstrated utility in mapping histone acetylation patterns linked to transcriptional activation .

Cellular Localization

  • Immunofluorescence: Revealed nuclear staining in HeLa cells, with enhanced signal upon histone deacetylase inhibitor (e.g., sodium butyrate) treatment .

Suppliers and Availability

The antibody is available from multiple manufacturers:

SupplierProduct CodePriceApplications
CusabioCSB-PA010380PA51acHU$319ELISA, ICC, IF, ChIP .
Assay GeniePACO56594$214ELISA, IF, ChIP .
AFG ScientificA24760$225–$350ELISA, ICC, IF .
BiomatikCAC15228$319ELISA, ICC, IF .

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 timelines may vary depending on the mode of purchase and delivery location. Please consult your local distributors for specific delivery information.
Synonyms
H1 histone family member 4 antibody; H1.4 antibody; H14_HUMAN antibody; H1E antibody; H1F4 antibody; Hist1h1e antibody; Histone 1 H1e antibody; Histone cluster 1 H1e antibody; Histone H1 antibody; Histone H1.4 antibody; Histone H1B antibody; MGC116819 antibody
Target Names
HIST1H1E
Uniprot No.

Target Background

Function
Histone H1 protein is involved in the formation of the chromatin fiber, a macromolecular structure that comprises linker DNA between nucleosomes. Histones H1 are essential for the condensation of nucleosome chains into higher-order structured fibers. Furthermore, they play a regulatory role in individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research indicates the presence of histones H1.2 and H1.4 in MDA-MB-231 metastatic breast cancer cells. Notably, phosphorylation at S173 of histone H1.2 and S172, S187, T18, T146, and T154 of H1.4 demonstrates a significant increase during the M phase of the cell cycle, suggesting a cell cycle-dependent nature of these events. PMID: 26209608
  2. This study identified and confirmed changes in HIST1H1E protein levels within the postsynaptic density in schizophrenia. PMID: 25048004
  3. The N-terminal domain of H1 plays a crucial role in determining the affinity and specificity of H1-chromatin interactions. PMID: 22425985
  4. Phosphorylation of H1.4 at S35, mediated by PKA, results in the dissociation of H1.4 from mitotic chromatin. However, this phosphorylation is also suggested to be necessary for specific mitotic functions. PMID: 21852232
  5. Allele-specific underacetylation of histone H4 downstream from the promoter is associated with X-inactivation in human cells. PMID: 12498347
  6. The lysine residue adjacent to the phosphorylation site found on the serine residue on the H1.4 peptide KARKSAGAAKR was also shown to be methylated, raising the question of whether the hypothesized "methyl/phos" switch could be extended to linker histones. PMID: 15595731
  7. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins PMID: 19144645

Show More

Hide All

Database Links

HGNC: 4718

OMIM: 142220

KEGG: hsa:3008

STRING: 9606.ENSP00000307705

UniGene: Hs.248133

Involvement In Disease
Rahman syndrome (RMNS)
Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=Mainly localizes in heterochromatin. Dysplays a punctuate staining pattern in the nucleus.

Q&A

What is HIST1H1E and what role does it play in chromatin structure?

HIST1H1E (Histone H1.4) is a linker histone that binds to DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. Unlike core histones (H2A, H2B, H3, and H4) that form the nucleosome core particle, HIST1H1E belongs to the H1 histone family that stabilizes higher-order chromatin structures. This histone is necessary for the condensation of nucleosome chains into more compact fibers and acts as a regulator of individual gene transcription through mechanisms involving chromatin remodeling, nucleosome spacing, and DNA methylation .

The H1 family of histones, including HIST1H1E, functions as chromatin architects that influence accessibility of transcription factors to DNA, thereby controlling gene expression patterns in different cellular contexts. Research has shown that differential expression and post-translational modifications of HIST1H1E can significantly alter chromatin compaction states with downstream effects on transcriptional programs.

What is the significance of acetylation at lysine 51 (K51) in HIST1H1E?

Acetylation at lysine 51 (K51) of HIST1H1E represents a specific post-translational modification that alters the protein's function in chromatin organization. While histones are commonly known to undergo various modifications (including acetylation, methylation, phosphorylation, and ubiquitination), the K51 site in HIST1H1E appears to have particular significance in epigenetic regulation .

Acetylation generally neutralizes the positive charge of lysine residues, potentially weakening histone-DNA interactions. In the context of HIST1H1E, K51 acetylation may modulate the protein's binding affinity to linker DNA, influencing chromatin compaction and accessibility. This modification likely contributes to the dynamic regulation of chromatin structure during processes such as transcription, replication, and DNA repair.

What are the recommended applications for Acetyl-HIST1H1E (K51) antibodies?

Acetyl-HIST1H1E (K51) antibodies have been validated for multiple experimental applications crucial for epigenetic research. The primary recommended applications include:

ApplicationRecommended DilutionPurpose
ELISAAs per manufacturer protocolQuantitative detection of acetylated HIST1H1E
ICC (Immunocytochemistry)1:20-1:200Cellular localization studies
IF (Immunofluorescence)1:50-1:200Visualization of acetylated HIST1H1E in fixed cells
ChIP (Chromatin Immunoprecipitation)Application-specificIdentifying genomic regions associated with acetylated HIST1H1E

The appropriate application depends on the specific research question. For instance, ChIP experiments are valuable for identifying genomic regions where acetylated HIST1H1E is enriched, potentially revealing genes regulated by this modification. Meanwhile, ICC and IF provide spatial information about the distribution of acetylated HIST1H1E within cellular compartments .

How should Acetyl-HIST1H1E (K51) antibody be stored and handled to maintain its activity?

Proper storage and handling of the Acetyl-HIST1H1E (K51) antibody is essential to preserve its specificity and sensitivity. Based on manufacturer recommendations, the antibody should be stored at -20°C or -80°C upon receipt. Repeated freeze-thaw cycles should be avoided as they can degrade antibody quality and reduce binding efficiency .

The antibody is typically supplied in liquid form with a buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative. The glycerol serves as a cryoprotectant, while the preservative helps prevent microbial contamination. For short-term use (1-2 weeks), aliquots can be stored at 4°C, but for longer-term storage, maintaining the antibody at -20°C or below is recommended to prevent degradation of the protein structure that could compromise epitope recognition.

How can I optimize ChIP protocols specifically for Acetyl-HIST1H1E (K51) to improve signal-to-noise ratio?

Optimizing ChIP protocols for Acetyl-HIST1H1E (K51) requires careful consideration of several parameters to enhance specificity and reduce background. Unlike core histones, linker histones such as HIST1H1E have different binding dynamics and may require modified approaches:

  • Crosslinking optimization: Test different formaldehyde concentrations (0.5-2%) and incubation times (5-15 minutes) to find the optimal balance. Excessive crosslinking can mask epitopes, while insufficient crosslinking may not properly preserve protein-DNA interactions.

  • Sonication parameters: Linker histones require careful optimization of sonication conditions. Start with milder sonication (e.g., fewer cycles or lower power settings) compared to standard ChIP protocols, as over-sonication may disproportionately remove linker histones from chromatin.

  • Antibody validation: Perform preliminary IP experiments followed by western blotting to confirm the antibody's specificity for acetylated HIST1H1E before proceeding with full ChIP experiments.

  • Blocking optimization: Include additional blocking agents such as BSA (0.1-0.5%) and specific competitors (like salmon sperm DNA) to reduce non-specific binding.

  • Sequential ChIP considerations: For studying co-occurrence with other modifications, sequential ChIP (re-ChIP) protocols may need extensive optimization due to the relatively lower abundance of HIST1H1E compared to core histones.

A successful ChIP experiment should include appropriate controls, such as IgG negative controls, input DNA controls, and positive controls targeting known regions where acetylated HIST1H1E is expected to bind .

What are the implications of HIST1H1E in cancer research, particularly in relation to acetylation status?

Research has revealed significant connections between HIST1H1E expression, its post-translational modifications (including acetylation), and cancer development. In acute myeloid leukemia (AML), high expression of HIST1H1E has been associated with poor patient prognosis . The altered expression and modification patterns of HIST1H1E likely contribute to dysregulated chromatin structure and aberrant gene expression in cancer cells.

Studies using AML cell lines have shown that knockdown of HIST1H1E promoted cell proliferation, suggesting a potential tumor suppressor role . This presents an interesting contradiction, as high expression correlates with poor prognosis while knockdown promotes proliferation in cell models. This apparent discrepancy may be explained by context-dependent functions of HIST1H1E in different cellular environments or by post-translational modifications like acetylation at K51.

The acetylation status of HIST1H1E may influence:

  • Chromatin accessibility: Altered acetylation patterns may change chromatin compaction states, affecting access to oncogenes or tumor suppressor genes.

  • Transcriptional networks: Modified HIST1H1E may interact differently with transcription factors and chromatin remodeling complexes.

  • DNA damage response: HIST1H1E plays roles in DNA repair pathways, and its acetylation status may affect repair efficiency, potentially contributing to genomic instability in cancer.

Researchers investigating cancer epigenetics should consider examining both expression levels and acetylation status of HIST1H1E at specific residues like K51 to gain a more complete understanding of its role in oncogenesis .

How can I design experiments to investigate the interplay between HIST1H1E acetylation and other histone modifications?

Investigating the interplay between HIST1H1E acetylation at K51 and other histone modifications requires a multi-faceted experimental approach:

  • Sequential ChIP (re-ChIP): This technique allows detection of co-occurrence of multiple modifications on the same chromatin regions.

    • First ChIP with anti-Acetyl-HIST1H1E (K51)

    • Elute the chromatin complexes under mild conditions

    • Perform a second ChIP with antibodies against other modifications of interest

    • Analyze enriched regions by qPCR or sequencing

  • Mass spectrometry-based approaches:

    • Purify nucleosomes or chromatin fragments

    • Perform quantitative MS to identify co-occurring modifications

    • Use targeted MS/MS to focus on specific histone residues

    • Implement SILAC or TMT labeling for quantitative comparisons between conditions

  • Proximity ligation assays (PLA):

    • Use pairs of antibodies (anti-Acetyl-HIST1H1E (K51) and antibodies against other modifications)

    • PLA signals will be generated only when modifications are in close proximity

    • Analyze by microscopy to detect nuclear localization patterns

  • CRISPR-based approaches:

    • Generate K51R mutants (preventing acetylation) using CRISPR-Cas9

    • Perform ChIP-seq for other modifications to detect changes in their patterns

    • Compare with wild-type cells to identify modifications dependent on K51 acetylation

A comprehensive experimental design should include perturbations of enzymes responsible for installing or removing acetylation at K51 (such as specific HATs or HDACs) to determine causality in the relationships between different modifications .

What are the technical challenges in differentiating between HIST1H1E and other H1 histone family members in experimental settings?

Differentiating between HIST1H1E (H1.4) and other H1 histone family members presents significant technical challenges due to their high sequence similarity and structural conservation. Researchers should consider these approaches to ensure specificity:

  • Antibody validation strategies:

    • Perform western blots using recombinant H1 variants to confirm antibody specificity

    • Use HIST1H1E knockout or knockdown cells as negative controls

    • Test cross-reactivity with other H1 variants using peptide competition assays

    • Validate with orthogonal detection methods

  • Sequence-specific detection:

    • Design primers or probes targeting unique regions of HIST1H1E for qPCR or RNA-seq

    • Focus on the regions surrounding K51, which may have sequence variations between H1 variants

    • Use isoform-specific siRNAs or shRNAs for knockdown experiments

  • Mass spectrometry approaches:

    • Analyze tryptic peptides containing K51 and surrounding regions

    • Look for variant-specific peptides that can differentiate between H1 family members

    • Use targeted MS/MS to focus on discriminating peptides

  • Genomic targeting:

    • When performing ChIP-seq, analyze genomic distribution patterns that may be unique to HIST1H1E

    • Compare with publicly available datasets for other H1 variants to identify HIST1H1E-specific binding regions

The challenge is particularly relevant when studying acetylation, as modification sites may be conserved across H1 variants. Researchers should always validate their findings using multiple approaches and clearly acknowledge potential cross-reactivity limitations in their experimental design and data interpretation .

How does HIST1H1E acetylation at K51 impact disease states beyond cancer, such as HIST1H1E syndrome?

Recent clinical findings have identified HIST1H1E syndrome as a distinct genetic disorder caused by pathogenic variants in the HIST1H1E gene. While the acetylation status at K51 has not been specifically studied in this syndrome, understanding this modification may provide insights into the molecular mechanisms of the condition .

HIST1H1E syndrome is characterized by:

  • Mild intellectual disability

  • Distinctive facial features

  • Endocrine abnormalities (including type 2 diabetes in at least one case)

  • Growth abnormalities

  • Possible cardiac manifestations (such as patent ductus arteriosus)

The impact of K51 acetylation in this context remains to be fully elucidated, but several research directions could be pursued:

  • Developmental impact: Investigating how K51 acetylation affects HIST1H1E function during embryonic development and neurogenesis could help explain the neurodevelopmental aspects of the syndrome.

  • Metabolic connections: The reported association with type 2 diabetes suggests potential roles in metabolic regulation. Researchers could examine how K51 acetylation affects HIST1H1E binding to regulatory regions of genes involved in glucose metabolism.

  • Tissue-specific effects: Comparing K51 acetylation patterns across different tissues could reveal why certain organ systems are particularly affected in HIST1H1E syndrome.

  • Therapeutic implications: Understanding how K51 acetylation affects HIST1H1E function could potentially identify targets for therapeutic intervention, such as specific histone deacetylase inhibitors or acetylation modulators.

Experimental approaches might include generating patient-derived induced pluripotent stem cells (iPSCs) and differentiating them into relevant cell types to study how HIST1H1E mutations affect K51 acetylation and downstream cellular processes .

What controls should be included when using Acetyl-HIST1H1E (K51) antibody in ChIP experiments?

Robust experimental design for ChIP experiments using Acetyl-HIST1H1E (K51) antibody requires comprehensive controls to ensure validity and interpretability of results:

  • Technical controls:

    • Input DNA control: A small portion (typically 5-10%) of pre-immunoprecipitated chromatin should be processed in parallel to account for differences in starting material.

    • IgG negative control: Non-specific IgG from the same species as the primary antibody (rabbit) should be used to establish background signal levels.

    • No-antibody control: Processing samples without antibody helps identify background associated with beads or other reagents.

  • Biological controls:

    • Positive genomic regions: Include analysis of regions known to be associated with HIST1H1E binding (such as specific gene promoters or heterochromatic regions).

    • Negative genomic regions: Include analysis of regions expected to lack HIST1H1E binding.

    • Treatment controls: Where applicable, include cells treated with HDAC inhibitors (which should increase acetylation) or HAT inhibitors (which should decrease acetylation).

  • Antibody validation controls:

    • Peptide competition: Pre-incubation of the antibody with the acetylated peptide should abrogate specific binding.

    • Use of cells with K51R mutation: If available, cells expressing a K51R mutant of HIST1H1E (preventing acetylation at this site) should show reduced signal.

  • Data analysis controls:

    • Normalization strategies: When performing ChIP-seq, appropriate normalization to input or spike-in controls should be employed.

    • Replicate concordance: Multiple biological replicates should show consistent enrichment patterns.

Implementing these controls helps distinguish genuine biological signals from technical artifacts and provides confidence in the specificity of the observed patterns of Acetyl-HIST1H1E (K51) binding .

How can I integrate ChIP-seq data for Acetyl-HIST1H1E (K51) with other omics datasets to gain functional insights?

Integrating ChIP-seq data for Acetyl-HIST1H1E (K51) with other omics datasets enables comprehensive understanding of the functional impact of this histone modification. Here's a methodological framework:

  • Integration with transcriptomic data:

    • Correlate Acetyl-HIST1H1E (K51) binding patterns with RNA-seq data

    • Analyze differential expression between regions with high versus low K51 acetylation

    • Perform time-course experiments to determine causality between changes in acetylation and transcriptional responses

    • Implementation: Use tools like BETA (Binding and Expression Target Analysis) or GSEA (Gene Set Enrichment Analysis)

  • Integration with other epigenomic data:

    • Overlay with maps of other histone modifications (H3K27ac, H3K4me3, etc.)

    • Compare with chromatin accessibility data (ATAC-seq, DNase-seq)

    • Correlate with DNA methylation patterns (WGBS, RRBS)

    • Implementation: Use tools like ChromHMM, EpiExplorer, or custom R/Python scripts with GenomicRanges

  • Integration with protein interaction data:

    • Identify proteins that preferentially interact with acetylated versus non-acetylated HIST1H1E

    • Use techniques like BioID or RIME (Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins)

    • Implementation: Use STRING, BioGRID, or other protein interaction databases to contextualize findings

  • Integration with 3D genome organization data:

    • Correlate Acetyl-HIST1H1E (K51) binding with Hi-C or ChIA-PET data

    • Analyze associations with topologically associating domains (TADs) and chromatin loops

    • Implementation: Use tools like HiCExplorer, Juicer, or FAN-C

  • Machine learning approaches:

    • Train predictive models using Acetyl-HIST1H1E (K51) ChIP-seq and other omics data

    • Identify genomic and epigenomic features that predict acetylation patterns

    • Implementation: Use scikit-learn, TensorFlow, or specialized tools like ChromImpute

This integrated approach can reveal how Acetyl-HIST1H1E (K51) contributes to higher-order chromatin organization, transcriptional regulation, and ultimately cellular phenotypes in both normal and disease contexts .

What are the recommended methods for quantifying changes in HIST1H1E acetylation at K51 in response to experimental treatments?

Accurate quantification of changes in HIST1H1E acetylation at K51 in response to experimental treatments requires multiple complementary approaches:

  • Western blotting with quantitative analysis:

    • Extract histones using acid extraction to ensure complete recovery

    • Separate by SDS-PAGE and probe with Acetyl-HIST1H1E (K51) antibody

    • Normalize to total HIST1H1E levels using a modification-insensitive antibody

    • Quantify using densitometry with appropriate statistical analysis

    • Use H3 or H4 as loading controls

  • Mass spectrometry-based quantification:

    • Perform histone extraction followed by propionylation to block unmodified lysines

    • Digest with trypsin and analyze by LC-MS/MS

    • Use labeled internal standards for absolute quantification

    • Calculate the ratio of acetylated to total HIST1H1E K51-containing peptides

    • Compare ratios across experimental conditions

  • ELISA-based approaches:

    • Use sandwich ELISA with capture antibody against HIST1H1E and detection antibody against acetyl-lysine

    • Alternatively, use a direct ELISA with the Acetyl-HIST1H1E (K51) antibody

    • Generate standard curves using recombinant acetylated and non-acetylated peptides

    • Normalize to total protein or specifically to total HIST1H1E

  • ChIP-qPCR at sentinel genomic regions:

    • Identify regions with stable HIST1H1E binding across conditions

    • Perform ChIP with Acetyl-HIST1H1E (K51) antibody

    • Calculate enrichment at these regions by qPCR

    • Compare enrichment across experimental conditions

    • Normalize to total HIST1H1E ChIP at the same regions

  • Immunofluorescence with quantitative image analysis:

    • Perform IF with Acetyl-HIST1H1E (K51) antibody

    • Co-stain with total HIST1H1E antibody

    • Acquire images under identical settings across conditions

    • Quantify nuclear signal intensity using software like ImageJ or CellProfiler

    • Calculate the ratio of acetylated to total HIST1H1E signal per nucleus

Each method has advantages and limitations, and combining multiple approaches provides the most robust quantification of changes in acetylation status .

How can Acetyl-HIST1H1E (K51) antibody be used to investigate epigenetic mechanisms in cellular differentiation models?

Acetyl-HIST1H1E (K51) antibody can be instrumental in unraveling epigenetic mechanisms during cellular differentiation through the following experimental approaches:

  • Temporal profiling during differentiation:

    • Track changes in K51 acetylation at different time points during differentiation

    • Correlate with expression of lineage-specific genes

    • Compare with other epigenetic marks to establish temporal hierarchies of epigenetic changes

    • Implementation: Perform ChIP-seq or ChIP-qPCR at defined stages of differentiation

  • Manipulating K51 acetylation levels:

    • Overexpress or inhibit specific HATs or HDACs that target K51

    • Assess impact on differentiation efficiency and lineage specification

    • Use CRISPR/Cas9 to create K51R (acetylation-deficient) or K51Q (acetylation-mimetic) mutations

    • Implementation: Combine with differentiation assays and gene expression analysis

  • Single-cell approaches:

    • Perform single-cell CUT&Tag or single-cell chromatin profiling

    • Identify heterogeneity in K51 acetylation within differentiating populations

    • Correlate with single-cell transcriptomics data

    • Implementation: Use computational methods to reconstruct differentiation trajectories based on epigenetic states

  • Spatial organization analysis:

    • Perform immunofluorescence to track changes in nuclear localization of acetylated HIST1H1E

    • Assess correlation with nuclear compartments (e.g., lamina, nucleolus, speckles)

    • Monitor changes in chromosome territory organization

    • Implementation: Use super-resolution microscopy techniques like STORM or PALM

  • Functional validation experiments:

    • Perform rescue experiments in cells with HIST1H1E knockdown using wild-type versus K51R mutants

    • Assess effects on differentiation markers and cellular phenotypes

    • Use inducible systems to modulate acetylation at specific differentiation stages

    • Implementation: Combine with phenotypic assays relevant to the differentiation model

These approaches can reveal how dynamic changes in HIST1H1E K51 acetylation contribute to the establishment and maintenance of cell type-specific gene expression programs during development and differentiation .

What troubleshooting approaches should be considered when Acetyl-HIST1H1E (K51) antibody yields inconsistent results in ChIP experiments?

When encountering inconsistent results with Acetyl-HIST1H1E (K51) antibody in ChIP experiments, systematic troubleshooting is essential:

  • Antibody-specific issues:

    • Lot-to-lot variability: Test multiple antibody lots and standardize using a reference sample

    • Antibody age/storage: Prepare fresh aliquots and avoid repeated freeze-thaw cycles

    • Specificity verification: Perform peptide competition assays to confirm specificity

    • Antibody concentration: Titrate antibody concentration (typically 1-10 μg per ChIP reaction)

  • Sample preparation issues:

    • Fixation parameters: Test different formaldehyde concentrations (0.5-2%) and times (5-15 minutes)

    • Cell number optimization: Ensure sufficient cell numbers (typically 1-5 million cells per ChIP)

    • Chromatin shearing: Verify fragment size distribution (200-500 bp is optimal)

    • Acetylation preservation: Add HDAC inhibitors (e.g., sodium butyrate) to all buffers

  • Technical variables:

    • Wash stringency: Adjust salt concentration in wash buffers (150-500 mM NaCl)

    • Blocking conditions: Test different blocking agents (BSA, milk, salmon sperm DNA)

    • Incubation times: Optimize antibody incubation (overnight at 4°C is typical)

    • Bead type: Compare different bead types (Protein A, Protein G, or a mixture)

  • Biological variables:

    • Cell cycle effects: Synchronize cells or account for cell cycle distribution

    • Cell culture conditions: Standardize passage number, confluence, and medium composition

    • Treatment timing: Optimize timing for treatments that affect acetylation levels

  • Control experiments and validation:

    • Positive control regions: Include genomic regions known to be enriched for K51 acetylation

    • Parallel ChIP: Perform simultaneous ChIP with antibodies against well-characterized marks

    • Alternative detection methods: Validate findings with orthogonal approaches (e.g., mass spectrometry)

Troubleshooting ParameterSuggested Optimization RangeNotes
Antibody amount1-10 μg per reactionStart with manufacturer's recommendation
Chromatin amount10-30 μg per reactionAdjust based on target abundance
Formaldehyde fixation0.5-2%, 5-15 minutesLower for linker histones
Sonication cycles10-30 cycles (30s on/30s off)Verify fragment size by gel
Wash buffer stringency150-500 mM NaClHigher stringency reduces background

Systematic optimization of these parameters can significantly improve reproducibility and sensitivity of ChIP experiments using Acetyl-HIST1H1E (K51) antibody .

How can I design experiments to investigate the functional consequences of HIST1H1E K51 acetylation in gene regulation?

Designing experiments to elucidate the functional consequences of HIST1H1E K51 acetylation in gene regulation requires a multi-faceted approach:

  • Genetic engineering approaches:

    • Generate CRISPR/Cas9-mediated K51R (acetylation-deficient) mutations

    • Create K51Q (acetylation-mimetic) mutations for comparison

    • Develop inducible systems to modulate acetylation levels temporally

    • Implementation: Use site-directed mutagenesis followed by stable transfection or homology-directed repair

  • Enzymatic modulation:

    • Identify and manipulate specific HATs that acetylate K51

    • Target specific HDACs that remove acetylation at K51

    • Use chemical inhibitors or genetic approaches (siRNA, CRISPR) to modulate these enzymes

    • Implementation: Perform enzyme screens using in vitro assays followed by cellular validation

  • Genomic binding analysis:

    • Compare genomic binding profiles of wild-type versus K51R mutant HIST1H1E

    • Identify differentially bound regions and associated genes

    • Correlate with changes in chromatin accessibility and other histone modifications

    • Implementation: ChIP-seq followed by differential binding analysis

  • Transcriptional impact assessment:

    • Perform RNA-seq in cells expressing wild-type versus mutant HIST1H1E

    • Analyze direct targets by integrating binding and expression data

    • Measure transcription rates using nascent RNA sequencing (GRO-seq, PRO-seq)

    • Implementation: Differential expression analysis with pathway enrichment

  • Chromatin structure analysis:

    • Assess impact on nucleosome positioning using MNase-seq

    • Evaluate effects on higher-order chromatin organization using Hi-C

    • Measure chromatin compaction using imaging approaches

    • Implementation: Compare structural features between wild-type and mutant conditions

  • Mechanistic studies:

    • Identify differential protein interactions of acetylated versus non-acetylated HIST1H1E

    • Assess impact on recruitment of chromatin remodeling complexes

    • Evaluate effects on phase separation properties of chromatin

    • Implementation: IP-MS, proximity labeling, or FRAP experiments

These complementary approaches can establish causal relationships between K51 acetylation and specific aspects of gene regulation, revealing the molecular mechanisms by which this modification influences chromatin function .

What emerging technologies might enhance our understanding of HIST1H1E K51 acetylation in chromatin biology?

Several cutting-edge technologies are poised to revolutionize our understanding of HIST1H1E K51 acetylation in chromatin biology:

  • Single-molecule approaches:

    • Single-molecule imaging: Techniques like PALM/STORM can visualize individual acetylated HIST1H1E molecules in situ

    • Single-molecule tracking: Using photoactivatable fluorescent proteins fused to HIST1H1E to track dynamics

    • Single-molecule force spectroscopy: Measuring how K51 acetylation affects binding strength to DNA

    • Implementation timeline: These techniques are currently available but require specialized equipment and expertise

  • Spatial epigenomics:

    • Spatial transcriptomics combined with protein detection: Techniques like MERFISH with antibody detection

    • In situ ChIP-seq: Methods that preserve tissue architecture while mapping histone modifications

    • Implementation timeline: These methods are rapidly developing and should become more accessible in the next 2-3 years

  • Multi-omics integration:

    • Single-cell multi-omics: Simultaneous measurement of K51 acetylation, transcription, and chromatin accessibility in single cells

    • Spatial multi-omics: Adding spatial dimension to integrated analyses

    • Implementation timeline: Commercial platforms are becoming available, with custom methods continuing to develop

  • AI-driven approaches:

    • Deep learning models: Predicting functional consequences of K51 acetylation based on sequence and structural context

    • AlphaFold-derived structural predictions: Modeling how K51 acetylation affects HIST1H1E structure and interactions

    • Implementation timeline: These computational resources are currently available and continuing to improve

  • Genome-wide engineering:

    • Base editing or prime editing: Precisely modifying K51 across the genome without double-strand breaks

    • Epigenetic editing: Targeted modification of K51 acetylation at specific genomic loci

    • Implementation timeline: These techniques are rapidly advancing and should be widely applicable within 1-2 years

These technologies will help resolve current contradictions in our understanding, such as the apparent discrepancy between high HIST1H1E expression correlating with poor prognosis in AML while knockdown promotes cell proliferation in experimental models .

How might understanding HIST1H1E K51 acetylation contribute to therapeutic approaches for diseases like cancer or HIST1H1E syndrome?

Understanding HIST1H1E K51 acetylation holds significant potential for developing novel therapeutic strategies for diseases including cancer and HIST1H1E syndrome:

  • Cancer therapeutic applications:

    • Biomarker development: Acetylation at K51 may serve as a prognostic or predictive biomarker, particularly in AML where HIST1H1E expression correlates with outcome

    • Target identification: Enzymes specifically modifying K51 could become therapeutic targets

    • Combination therapies: Understanding how K51 acetylation affects response to existing epigenetic drugs (HDAC inhibitors, BET inhibitors) could inform combination strategies

    • Synthetic lethality approaches: Identifying genes that become essential in the context of altered K51 acetylation

  • HIST1H1E syndrome interventions:

    • Acetylation modulation: If K51 acetylation is dysregulated in HIST1H1E syndrome, targeted approaches to normalize this modification might ameliorate symptoms

    • Compensatory approaches: Identifying and targeting pathways that can compensate for HIST1H1E dysfunction

    • Gene therapy strategies: For mutations affecting K51 acetylation, gene editing approaches might restore normal function

  • Technological development pathways:

    • High-throughput screening: Identifying small molecules that specifically modulate K51 acetylation

    • Structure-based drug design: Using structural information about acetylated HIST1H1E to design inhibitors or activators

    • Deliverable development: Creating methods to target therapies to specific cell types affected in disease

  • Translational considerations:

    • Patient stratification: Using K51 acetylation status to identify patients likely to respond to specific therapies

    • Resistance mechanisms: Understanding how changes in K51 acetylation might contribute to treatment resistance

    • Monitoring strategies: Developing methodologies to track K51 acetylation in patient samples as a biomarker of response

  • Broader implications:

    • Developmental disorders: Insights from HIST1H1E syndrome may extend to other neurodevelopmental conditions

    • Metabolic disorders: The association with type 2 diabetes suggests potential applications in metabolic disease

    • Aging-related conditions: Given the role of epigenetic changes in aging, understanding K51 acetylation might inform interventions for age-related disorders

The development of such therapeutic approaches requires continued basic research to fully characterize the molecular mechanisms and cellular consequences of HIST1H1E K51 acetylation in both normal physiology 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 2024 Thebiotek. All Rights Reserved.