Di-methyl-HIST1H1E (K16) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Definition and Target

  • Target Protein: HIST1H1E (also known as Histone H1.4) is a linker histone that stabilizes nucleosome structure by binding to DNA between nucleosomes .

  • Epitope: The antibody specifically recognizes the di-methylation state of lysine 16 (K16) on HIST1H1E .

  • Immunogen: Peptide sequences surrounding the di-methylated lysine 16 residue derived from human Histone H1.4 .

Validated Applications

The antibody is optimized for multiple experimental techniques:

ApplicationDescriptionSources
ChIP/ChIP-seqDetects chromatin-bound di-methylated HIST1H1E in nuclear DNA-protein complexes .
ELISAQuantifies di-methylated HIST1H1E in lysates or purified histones .
Immunocytochemistry (ICC)Visualizes nuclear localization of di-methylated HIST1H1E in fixed cells .
Immunofluorescence (IF)Confocal microscopy studies confirm nuclear staining patterns .
Western Blot (WB)Detects di-methylated HIST1H1E in denatured samples (select products) .

Research Findings and Biological Relevance

  • Gene Regulation: Di-methylation of HIST1H1E K16 is associated with transcriptional repression, as linker histones modulate chromatin accessibility .

  • Cancer Research: Aberrant histone methylation, including H1E K16 di-methylation, has been linked to oncogenic processes .

  • Epigenetic Studies: This antibody enables mapping of H1E K16 di-methylation across the genome via ChIP-seq, aiding in chromatin dynamics research .

Key Considerations for Use

  • Validation: Ensure the antibody’s methylation state specificity matches experimental goals .

  • Cross-reactivity: Verify reactivity with non-human samples if applicable .

  • Optimization: Follow vendor-recommended dilutions and protocols for consistent results .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 working days of receipt. Delivery time may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
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 a crucial component of chromatin, binding to the linker DNA between nucleosomes. Its role is essential in the formation of the macromolecular structure known as the chromatin fiber. Histone H1 proteins are required for the condensation of nucleosome chains into higher-order structured fibers. Furthermore, they act as regulators of individual gene transcription by influencing chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research indicates that histones H1.2 and H1.4 are present in MDA-MB-231 metastatic breast cancer cells. Phosphorylation at S173 of histone H1.2 and S172, S187, T18, T146, and T154 of H1.4 significantly increases during the M phase of the cell cycle, suggesting that these events are cell cycle-dependent. PMID: 26209608
  2. This study identified and confirmed changes in HIST1H1E protein within the postsynaptic density in schizophrenia. PMID: 25048004
  3. The N-terminal domain of H1 is a significant determinant of the affinity and specificity of H1-chromatin interactions. PMID: 22425985
  4. Phosphorylation of H1.4S35 by PKA leads to the dissociation of H1.4 from mitotic chromatin, suggesting that this phosphorylation is 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. This raises 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 di-methylation at K16 play in chromatin biology?

HIST1H1E is a member of the H1 histone family that encodes for Histone H1.4, a linker histone responsible for higher-order chromatin structure. This protein plays a critical role in genome stability, DNA replication, and repair mechanisms . The di-methylation of lysine residues in histones, including K16 in HIST1H1E, represents an important post-translational modification that contributes to epigenetic regulation of gene expression. Similar to other histone modifications, di-methylation at K16 can influence chromatin compaction and accessibility, potentially serving as a recruitment site for specific binding partners that affect transcriptional activity.

How does di-methylation of HIST1H1E differ functionally from other histone H1 modifications?

The di-methylation of HIST1H1E at K16 represents one of several possible methylation states at different lysine residues within the histone. While the search results don't specifically detail K16 modification, we can observe from related modifications that different methylation sites serve distinct functions. For instance, monomethylation of H1.4 at K85 by WHSC1 has been associated with transcriptional activation of OCT4 and stemness features in squamous cell carcinoma of the head and neck (SCCHN) . Di-methylation at different lysine positions can recruit varied effector proteins, leading to context-dependent outcomes for gene regulation. Unlike modifications on core histones such as H3K9me2, which is predominantly associated with gene repression and heterochromatin formation , H1 modifications may have more specialized roles in regulating higher-order chromatin structure.

What are the established research applications for Di-methyl-HIST1H1E (K16) Antibody?

Di-methyl-HIST1H1E (K16) Antibody serves as a valuable tool for detecting and analyzing this specific histone modification across various experimental approaches. Based on similar histone antibodies, the primary applications include:

  • Western blotting for protein level detection

  • Chromatin immunoprecipitation (ChIP) for genome-wide or locus-specific analysis

  • Immunofluorescence for visualizing nuclear localization patterns

  • Immunohistochemistry for tissue-specific expression studies

  • ELISA-based quantification methods

These applications enable researchers to investigate the presence and distribution of di-methylated HIST1H1E in different biological contexts, similar to how other histone antibodies are employed .

What are the optimal sample preparation protocols for detecting Di-methyl-HIST1H1E (K16) in different experimental setups?

For optimal detection of di-methyl-HIST1H1E (K16), sample preparation should be tailored to the specific experimental approach:

For Western Blotting:

  • Prepare nuclear extracts using a dedicated nuclear extraction kit to ensure enrichment of nuclear proteins including histones

  • Include protease inhibitor cocktails to prevent degradation

  • Use optimized dilutions (typically 1:500-1:1000 for similar histone antibodies)

  • Transfer to nitrocellulose membrane for better protein retention

For ChIP Assays:

  • Crosslink chromatin with 1% formaldehyde for 10-15 minutes

  • Sonicate to generate DNA fragments of 200-500 bp

  • Use appropriate washing buffers to reduce background

  • Include appropriate positive controls (known targets) and negative controls (IgG)

For Immunofluorescence:

  • Fix cells with 4% paraformaldehyde

  • Permeabilize with 0.2% Triton X-100

  • Block with BSA or serum to reduce non-specific binding

  • Incubate with primary antibody at optimal concentration overnight at 4°C

Each method requires validation and optimization specific to the antibody's characteristics and the biological system under investigation.

How should researchers validate the specificity of Di-methyl-HIST1H1E (K16) Antibody?

Validation of antibody specificity is critical for reliable experimental outcomes. Researchers should:

  • Peptide Competition Assay: Pre-incubate the antibody with excess synthetic di-methylated K16 peptide, which should abolish specific signal

  • Knockout/Knockdown Controls: Use HIST1H1E knockout cells or CRISPR-engineered K16 mutants (K16R or K16A) that cannot be methylated

  • Methyltransferase Inhibition: Treat cells with specific histone methyltransferase inhibitors that affect the enzyme responsible for K16 di-methylation

  • Dot Blot Analysis: Test reactivity against a panel of modified and unmodified histone peptides, including:

    • Unmodified K16 peptide

    • Mono-methylated K16 peptide

    • Di-methylated K16 peptide

    • Tri-methylated K16 peptide

    • Peptides with modifications at adjacent lysine residues

  • Western Blot Profile: Compare profiles across different cell types with known differences in histone methylation patterns

This multi-approach validation strategy ensures that signals detected truly represent di-methylation at K16 of HIST1H1E, rather than cross-reactivity with other modifications.

What controls should be included when performing ChIP experiments with Di-methyl-HIST1H1E (K16) Antibody?

For rigorous ChIP experiments, the following controls are essential:

Positive Controls:

  • ChIP with antibodies against abundant histone modifications (e.g., H3K4me3 at active promoters)

  • Known genomic regions where the di-methyl-HIST1H1E (K16) mark is expected to be enriched

Negative Controls:

  • IgG control from the same species as the primary antibody

  • Regions of the genome known to lack this modification

  • Input DNA (pre-immunoprecipitation chromatin)

Technical Controls:

  • Sonication efficiency check by gel electrophoresis

  • Quantitative PCR of immunoprecipitated DNA with primers for positive and negative regions

  • Sequential ChIP (re-ChIP) to confirm co-occurrence with other modifications

Biological Controls:

  • Cells treated with histone methyltransferase inhibitors

  • Cells with genetic manipulation of enzymes responsible for K16 methylation

This comprehensive control strategy helps distinguish true signal from background and validates the specificity of the antibody in the ChIP context.

How should researchers interpret changes in Di-methyl-HIST1H1E (K16) levels in the context of gene expression studies?

Interpreting changes in di-methyl-HIST1H1E (K16) levels requires careful consideration of multiple factors:

  • Correlation Analysis: Compare di-methyl-HIST1H1E (K16) ChIP-seq data with RNA-seq or other transcriptome data to identify correlations between modification patterns and gene expression changes.

  • Context-Dependent Interpretation: Consider that histone H1 modifications may have different effects based on:

    • Genomic context (promoter, enhancer, gene body)

    • Cell type and developmental stage

    • Presence of other histone modifications

  • Temporal Analysis: Track changes in both the modification and gene expression over time to establish causality rather than mere correlation.

  • Integration with Chromatin Structure Data: Combine with techniques like ATAC-seq or DNase-seq to understand how this modification correlates with chromatin accessibility.

  • Functional Validation: Use targeted approaches like site-directed mutagenesis (K16R or K16A) to confirm the functional impact of the modification on specific genes.

For example, similar to how WHSC1-mediated H1.4K85 monomethylation has been linked to transcriptional activation of OCT4 and stemness features in cancer cells , di-methylation at K16 may have distinct gene regulatory functions that need to be carefully characterized in each experimental context.

What are common technical challenges when working with Di-methyl-HIST1H1E (K16) Antibody and how can they be addressed?

Researchers commonly encounter these technical challenges:

Issue 1: High Background Signal

  • Solution: Optimize blocking conditions (5% BSA or milk), increase washing stringency, and titrate antibody concentration

  • Prevention: Pre-clear lysates with protein A/G beads before immunoprecipitation

Issue 2: Low Signal Intensity

  • Solution: Enrich for nuclear fraction, optimize extraction buffers to preserve histone modifications, and adjust antibody incubation time/temperature

  • Prevention: Add histone deacetylase inhibitors (e.g., sodium butyrate) and methylation inhibitors during sample preparation

Issue 3: Inconsistent Results Between Experiments

  • Solution: Standardize sample preparation protocols, use internal loading controls, and normalize to total histone H1 levels

  • Prevention: Prepare larger batches of nuclear extracts, aliquot and store at -80°C

Issue 4: Cross-Reactivity with Other Modifications

  • Solution: Validate specificity using peptide competition assays and dot blots with different modified peptides

  • Prevention: Use antibodies that have been extensively validated for specificity

Issue 5: Epitope Masking by Protein Complexes

  • Solution: Try different extraction conditions or sonication techniques to disrupt protein complexes

  • Prevention: Include detergents or salt washes that disrupt protein-protein interactions without affecting antibody binding

Addressing these challenges ensures more reliable and reproducible results when working with this specialized antibody.

How can researchers differentiate between technical artifacts and biologically significant changes in Di-methyl-HIST1H1E (K16) patterns?

Distinguishing technical artifacts from true biological signals requires systematic approaches:

  • Biological Replicates: Analyze at least three independent biological replicates to establish statistical significance of observed changes.

  • Technical Replicates: Perform technical replicates of critical experiments to assess method reproducibility.

  • Dose-Response Relationships: When using treatments that affect methylation, establish dose-response curves to identify biologically meaningful thresholds.

  • Orthogonal Methods: Confirm key findings using alternative techniques:

    • If ChIP-seq shows enrichment, validate with ChIP-qPCR

    • If Western blot shows changes, confirm with mass spectrometry

    • If immunofluorescence shows localization patterns, validate with cell fractionation

  • Normalization Strategies: Use appropriate normalization methods:

    • Normalize to total histone H1 levels

    • Use spike-in controls in sequencing experiments

    • Apply batch effect correction in large-scale studies

  • Time-Course Analysis: Examine changes over multiple time points to distinguish transient fluctuations from stable biological effects.

  • Genetic Validation: Use genetic approaches (siRNA, CRISPR-Cas9) to manipulate enzymes responsible for the modification and observe whether expected changes occur.

By implementing these strategies, researchers can confidently identify biologically meaningful changes in di-methyl-HIST1H1E (K16) patterns.

How can Di-methyl-HIST1H1E (K16) Antibody be integrated into multi-omics approaches to understand chromatin regulation?

Integration of di-methyl-HIST1H1E (K16) analysis into multi-omics approaches provides comprehensive insights into chromatin regulation:

ChIP-seq Integration:

  • Combine with RNA-seq to correlate K16 di-methylation with transcriptional outcomes

  • Integrate with ATAC-seq or DNase-seq to understand chromatin accessibility

  • Analyze alongside ChIP-seq for other histone modifications to build comprehensive epigenetic maps

Mass Spectrometry Integration:

  • Perform quantitative proteomics to identify proteins that recognize or are affected by K16 di-methylation

  • Use SILAC approaches to quantify changes in modification levels across conditions

  • Employ crosslinking mass spectrometry to identify proteins in proximity to modified histones

Genomic Integration:

  • Correlate K16 di-methylation patterns with genetic variants from GWAS studies

  • Examine modification changes in response to genomic alterations

Imaging Integration:

  • Combine with super-resolution microscopy to visualize nuclear distribution

  • Use live-cell imaging with modification-specific antibodies to track dynamics

Bioinformatic Integration:

  • Develop computational models that predict transcriptional outcomes based on K16 di-methylation and other epigenetic features

  • Apply machine learning approaches to identify patterns across multi-omics datasets

This integrated approach can reveal how di-methylation of HIST1H1E at K16 coordinates with other epigenetic modifications to regulate gene expression, similar to how other histone modifications function within the broader context of the histone code.

What role does Di-methyl-HIST1H1E (K16) play in neurodevelopmental disorders and how can the antibody advance this research?

Research into histone H1 modifications, including di-methylation, may have significant implications for neurodevelopmental disorders:

Current Knowledge:
HIST1H1E mutations have been implicated in neurodevelopmental disorders, with evidence showing that variants in HIST1H1E contribute to conditions characterized by intellectual disability, hypotonia, and distinctive craniofacial features, collectively known as Rahman syndrome . The C-terminal domain of HIST1H1E appears to be a mutation "hot-spot," with variants in this region potentially disrupting the protein's ability to bind linker DNA and regulate chromatin structure .

Research Applications:
The Di-methyl-HIST1H1E (K16) Antibody can be utilized to:

  • Comparative Profiling: Compare K16 di-methylation patterns between:

    • Patient-derived cells versus controls

    • Brain organoids developed from patient iPSCs versus controls

    • Mouse models of neurodevelopmental disorders

  • Developmental Trajectory Analysis: Track changes in K16 di-methylation during:

    • Neural differentiation

    • Brain development stages

    • Critical periods of synaptic plasticity

  • Therapeutic Screening: Evaluate how potential therapeutic compounds affect K16 di-methylation in:

    • High-throughput drug screening platforms

    • Patient-derived cellular models

    • Animal models of HIST1H1E-related disorders

  • Mechanistic Studies: Investigate how K16 di-methylation affects:

    • Expression of neurodevelopmental genes

    • Interaction with neuron-specific transcription factors

    • Regulation of chromatin accessibility in neural cells

Understanding these epigenetic mechanisms may provide insights into the molecular pathways disrupted in HIST1H1E-related neurodevelopmental disorders, potentially leading to targeted therapeutic approaches.

How can researchers investigate the enzymes responsible for Di-methyl-HIST1H1E (K16) and their regulatory mechanisms?

Investigating the enzymes responsible for di-methylation of HIST1H1E at K16 requires a multi-faceted approach:

Enzyme Identification:

  • Candidate Approach: Test known histone methyltransferases (HMTs) for activity on H1.4K16:

    • Perform in vitro methyltransferase assays with recombinant enzymes

    • Use synthetic peptides containing the K16 residue as substrates

    • Quantify methylation using mass spectrometry or radioactive assays

  • Unbiased Screening: Conduct genome-wide screens to identify responsible enzymes:

    • CRISPR-Cas9 screens targeting known methyltransferases

    • Affinity purification using unmodified H1 peptides followed by mass spectrometry

    • Yeast two-hybrid screening with H1.4 as bait

Regulatory Mechanism Analysis:

  • Expression Regulation: Analyze transcriptional and post-transcriptional regulation of identified enzymes:

    • Promoter analysis and transcription factor binding studies

    • miRNA regulation studies

    • mRNA stability assays

  • Enzyme Activity Regulation: Investigate factors controlling enzyme activity:

    • Post-translational modifications of the enzyme itself

    • Protein-protein interactions affecting enzyme activity

    • Metabolic factors (e.g., SAM availability)

    • Cellular localization and compartmentalization

  • Context-Dependent Regulation: Examine how activity changes across:

    • Cell cycle phases

    • Developmental stages

    • Cellular stress conditions

    • Disease states

This systematic approach can reveal the enzymatic machinery responsible for di-methylation at K16 and how it is regulated in different biological contexts, similar to investigations of WHSC1, which has been identified as an enzyme that mono-methylates H1.4 at K85 .

How does Di-methyl-HIST1H1E (K16) compare to other histone methylation marks in terms of stability, inheritance, and biological significance?

Comparative analysis of di-methyl-HIST1H1E (K16) with other histone methylation marks reveals important distinctions:

Stability and Dynamics:

Histone ModificationTypical StabilityTurnover RateKey Features
H3K9me2HighSlowMajor heterochromatin mark
H3K4me3ModerateMediumActive promoter mark
H3K27me3HighSlowRepressive mark
H1.4K85me1Under investigationUnknownAssociated with stemness in SCCHN
H1.4K16me2Under investigationUnknownRequires further characterization

Inheritance Patterns:

  • Core histone methylation marks like H3K9me2 are generally more stable through cell division

  • Linker histone modifications may be more dynamic due to higher turnover rates of H1 histones

  • The inheritance mechanisms for H1 modifications remain less well-characterized than those for core histones

Biological Significance:

  • Core histone methylations directly affect nucleosome stability and transcription factor accessibility

  • H1 modifications like di-methyl-HIST1H1E (K16) likely influence higher-order chromatin structure

  • While H3K9me2 is strongly associated with gene silencing , the specific functional outcomes of H1 modifications are still being elucidated

  • Recent evidence suggests H1.4K85 monomethylation contributes to stemness features , indicating H1 modifications have critical biological functions

Understanding these comparative aspects helps position di-methyl-HIST1H1E (K16) within the broader histone code and informs experimental approaches for studying its specific functions.

What methodological differences should researchers consider when working with linker histone modifications compared to core histone modifications?

Working with linker histone modifications presents distinct methodological considerations compared to core histone modifications:

Extraction and Enrichment:

  • Linker histones (H1) dissociate more easily from chromatin than core histones

  • Higher salt concentrations in extraction buffers may be needed for consistent H1 extraction

  • Nuclear extraction protocols need optimization to retain H1 modifications

Antibody Selection and Validation:

  • Antibodies against linker histone modifications typically require more extensive validation

  • Cross-reactivity testing against various H1 variants is essential (H1.1-H1.5)

  • Peptide competition assays should include related H1 modification sites

ChIP Protocol Adjustments:

  • Crosslinking conditions may need optimization for H1 (longer fixation times)

  • Sonication conditions should be adjusted to preserve H1-DNA interactions

  • Washing conditions might require optimization to reduce background

Data Analysis Considerations:

  • Genome-wide distribution patterns differ between H1 and core histone modifications

  • Bioinformatic algorithms may need adjustment for linker histone binding patterns

  • Reference datasets for normalization may be less abundant for H1 modifications

Functional Assays:

  • Different reporter systems may be needed to assess the functional impact

  • CRISPR-based approaches for H1 modifications should consider potential redundancy among H1 variants

  • In vitro reconstitution assays should incorporate higher-order chromatin structures

By accounting for these methodological differences, researchers can more effectively study di-methyl-HIST1H1E (K16) and other linker histone modifications with appropriate technical approaches.

How can researchers design experiments to determine if Di-methyl-HIST1H1E (K16) is a cause or consequence of transcriptional changes?

Distinguishing whether di-methyl-HIST1H1E (K16) causes transcriptional changes or results from them requires carefully designed experimental approaches:

Temporal Resolution Studies:

  • Time-Course Analysis:

    • Induce transcriptional changes with well-characterized stimuli

    • Collect samples at multiple time points (minutes to hours)

    • Simultaneously analyze di-methyl-HIST1H1E (K16) levels and transcriptional output

    • Determine which change occurs first

  • Synchronized Cell Systems:

    • Synchronize cells at specific cell cycle stages

    • Track both modification and transcription through cell cycle progression

    • Identify temporal relationships between modification appearance and transcriptional changes

Causal Intervention Studies:

  • Enzyme Manipulation:

    • Overexpress or inhibit the methyltransferase responsible for K16 di-methylation

    • Assess direct transcriptional consequences using RNA-seq

    • Perform rescue experiments with wild-type or enzymatically dead versions

  • Site-Specific Mutations:

    • Generate K16R or K16A mutants that cannot be methylated

    • Create designer histones that mimic constitutive methylation

    • Assess transcriptional outcomes of these modifications

Mechanistic Studies:

  • Protein Interaction Studies:

    • Identify proteins that specifically recognize di-methyl-HIST1H1E (K16)

    • Determine if these readers recruit transcriptional machinery

    • Perform domain swapping or mutational analysis to disrupt specific interactions

  • Chromatin Accessibility Analysis:

    • Measure how di-methylation affects nucleosome positioning and stability

    • Assess chromatin accessibility changes using ATAC-seq or DNase-seq

    • Correlate structural changes with transcriptional outcomes

Mathematical Modeling:

  • Predictive Modeling:

    • Develop mathematical models that predict transcriptional outcomes based on modification levels

    • Test model predictions with experimental validation

    • Refine models based on experimental feedback

By integrating these approaches, researchers can establish whether di-methyl-HIST1H1E (K16) plays a causative role in transcriptional regulation or represents a downstream consequence of other regulatory events.

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.