The HIST1H1E (Ab-51) Antibody targets the acetylation site at lysine 51 (K51) of Histone H1.4, a replication-dependent linker histone encoded by the HIST1H1E gene on human chromosome 6. This antibody is produced in rabbits using a synthetic peptide immunogen derived from the human Histone H1.4 sequence .
Histone H1.4 facilitates higher-order chromatin compaction by binding linker DNA between nucleosomes. It regulates gene transcription, DNA methylation, and chromatin remodeling . Mutations in HIST1H1E, particularly in its C-terminal domain, are linked to neurodevelopmental disorders (e.g., Rahman syndrome) and premature aging due to disrupted chromatin function .
The antibody detects acetylation at K51, a post-translational modification (PTM) associated with transcriptional activation. It has been used in:
ChIP assays: Mapping histone acetylation patterns at gene promoters .
Cellular imaging: Localizing H1.4 in nuclear compartments during interphase .
Neurodevelopmental Disorders: Frameshift mutations in HIST1H1E (e.g., p.Ala141GlufsTer56) correlate with intellectual disability (ID), craniofacial abnormalities, and autism .
Aging and Senescence: Truncated H1.4 mutants accelerate cellular senescence and chromatin instability, as shown in fibroblast studies .
Cross-Reactivity: Specific to human H1.4; no cross-reactivity with other H1 subtypes confirmed via peptide-blocking assays .
Functional Assays: Validated in HeLa cells for chromatin binding and resistance to high-salt extraction (CSK buffer) .
Mutation Hotspot: All pathogenic HIST1H1E variants (e.g., c.416_419dupAGAA) localize to the C-terminal domain, disrupting DNA binding and chromatin compaction .
Phenotypic Variability: Patients with identical mutations (e.g., Ala144Glyfs*52) exhibit diverse clinical features, suggesting genetic background influences outcomes .
HIST1H1E (also known as H1.4, H1E, or H1F4) is a linker histone protein that binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. This protein plays a crucial role in the condensation of nucleosome chains into higher-order structured fibers. Beyond its structural role, HIST1H1E functions as a regulator of individual gene transcription through multiple mechanisms including chromatin remodeling, nucleosome spacing, and DNA methylation . The protein is part of the H1 histone family and contributes significantly to genome organization and expression regulation.
For maximum stability and performance, HIST1H1E (Ab-51) antibody should be stored at either -20°C or -80°C immediately upon receipt. Repeated freeze-thaw cycles should be strictly avoided as they can compromise antibody integrity and function. The antibody is typically supplied in liquid form with a buffer composition of 0.03% Proclin 300 as a preservative, 50% Glycerol, and 0.01M PBS at pH 7.4 . When planning experiments, it's advisable to prepare working aliquots to minimize freeze-thaw cycles of the stock solution.
The HIST1H1E (Ab-51) antibody has been validated for ELISA (Enzyme-Linked Immunosorbent Assay) and IF (Immunofluorescence) applications. For immunofluorescence specifically, the recommended dilution range is 1:50-1:200 . When establishing a new experimental protocol, it is advisable to perform a titration experiment with multiple dilutions within this range to determine optimal antibody concentration for your specific cell type or tissue. The polyclonal nature of this antibody means it may detect multiple epitopes on the target protein, potentially increasing sensitivity but requiring careful validation for specificity.
For immunofluorescence detection of HIST1H1E using this antibody:
Culture cells on coverslips coated with either 0.01% poly-L-lysine or 0.2% gelatin, depending on your cell type
Fix cells with 3% paraformaldehyde for 10 minutes at room temperature
Permeabilize with 0.5% Triton X-100
Block in a buffer containing 1% BSA, 22.52 mg/mL glycine in PBST (0.1% Tween 20 in PBS) for 30 minutes
Incubate with HIST1H1E (Ab-51) primary antibody (diluted 1:50-1:200) for 1 hour at room temperature
Wash three times with PBST
Incubate with appropriate Alexa Fluor secondary antibody for 1 hour at room temperature
Counterstain with DAPI to visualize nuclei
Mount slides and image using fluorescent or confocal microscopy
This protocol has been successfully employed in studies examining nuclear localization patterns of histone proteins and can be adapted for co-localization studies with other chromatin components.
While not explicitly listed among the validated applications, researchers experienced with HIST1H1E antibodies have adapted protocols for ChIP analysis. For optimal results:
Crosslink chromatin using 1% formaldehyde for 10 minutes at room temperature
Quench with 125 mM glycine for 5 minutes
Lyse cells and sonicate chromatin to fragments of approximately 200-500 bp
Pre-clear chromatin with protein A/G beads
Immunoprecipitate with HIST1H1E (Ab-51) antibody (5-10 μg per reaction) overnight at 4°C
Capture antibody-chromatin complexes using protein A/G beads
Wash stringently to remove non-specific binding
Reverse crosslinks and purify DNA for downstream analysis
For quantitative assessment, compare enrichment to input and IgG control. Expect enrichment at heterochromatic regions and repressed genes due to HIST1H1E's role in chromatin compaction. Since HIST1H1E belongs to a family of highly similar proteins, validation of ChIP specificity through knockout controls is strongly recommended.
Rahman syndrome is associated with de novo frameshift mutations within the region coding for the C-terminal domain (CTD) of HIST1H1E. These mutations typically cluster within a 94-bp region in the CTD of the gene . To distinguish between normal and mutant variants:
| Feature | Wild-type HIST1H1E | Rahman Syndrome HIST1H1E |
|---|---|---|
| C-terminal structure | Intact CTD with normal positive charge | Truncated with altered C-terminal sequence |
| Terminal amino acids | Normal sequence | Contains a stretch of 38 altered amino acids |
| Net charge | Higher positive charge | Reduced positive charge |
| Chromatin binding | Normal binding pattern | Altered binding dynamics |
| Effects on H3 methylation | Normal H3K4me2, H3K9me3, H3K27me3 | Reduced methylation at these sites |
| HP1β binding | Normal binding | Decreased binding |
| Chromatin compaction | Normal heterochromatin formation | Enhanced chromatin relaxation |
For experimental detection, standard HIST1H1E antibodies may not distinguish between wild-type and mutant variants. Consider employing:
Genotyping with primers flanking the mutation hotspot
Western blotting to detect size differences (mutants typically show altered migration)
Engineered tagged constructs (e.g., PA-GFP-H1.4) as implemented in cellular models
To investigate the real-time dynamics of HIST1H1E in living cells, researchers have developed several sophisticated approaches:
Photoactivatable GFP (PA-GFP) tagging: As demonstrated in the mouse embryonic stem cell model for Rahman syndrome, HIST1H1E can be tagged with PA-GFP . This approach allows:
Precise activation of fluorescence in specific nuclear regions
Measurement of protein mobility and exchange rates between chromatin-bound and free states
Comparison between wild-type and mutant protein dynamics
Fluorescence Recovery After Photobleaching (FRAP):
Bleach a small region of interest containing fluorescently-tagged HIST1H1E
Record recovery of fluorescence over time
Calculate binding/unbinding kinetics from recovery curves
Compare residence times across different chromatin states
Single-molecule tracking:
Label HIST1H1E with photoconvertible fluorophores
Track individual molecules over time
Analyze diffusion coefficients and binding events
Determine differences in mobility between euchromatin and heterochromatin regions
When implementing these approaches, careful controls must be performed to ensure that tagging does not disrupt native protein function or localization.
HIST1H1E mutations have been implicated in Rahman syndrome, a neurodevelopmental disorder characterized by intellectual disability and overgrowth . To apply HIST1H1E (Ab-51) antibody in studying chromatin dysregulation:
Comparative immunofluorescence:
Compare nuclear distribution patterns between patient-derived and control cells
Quantify signal intensity across different nuclear compartments
Co-stain with markers of heterochromatin (H3K9me3, HP1β) to assess chromatin organization
Chromatin accessibility studies:
Combine antibody staining with ATAC-seq or DNase hypersensitivity assays
Correlate HIST1H1E localization with regions of altered chromatin accessibility
Investigate relationship between antibody binding sites and differential gene expression
Protein interaction networks:
Use HIST1H1E (Ab-51) for co-immunoprecipitation followed by mass spectrometry
Identify differential protein interactions between wild-type and disease states
Map interaction changes to pathways involved in neurodevelopment
These approaches can provide insights into how HIST1H1E variants contribute to pathogenesis beyond simple descriptive observations.
Comprehensive validation of HIST1H1E (Ab-51) antibody specificity requires multiple controls:
Genetic controls:
HIST1H1E knockout or knockdown cells (negative control)
Cells overexpressing HIST1H1E (positive control)
Cells expressing tagged HIST1H1E for co-localization studies
Peptide competition:
Pre-incubate antibody with excess immunizing peptide
Observe elimination of specific signal in immunoassays
Cross-reactivity assessment:
Test reactivity against related H1 variants (H1.1-H1.5)
Evaluate species cross-reactivity if working with non-human models
Antibody dilution series:
Perform titration experiments to determine optimal concentration
Verify signal-to-noise ratio across dilution range
Application-specific controls:
For IF: secondary antibody only control
For Western blotting: molecular weight verification and loading controls
For ChIP: IgG and input controls
Thorough validation is particularly important given the high sequence similarity between histone H1 family members.
| Challenge | Possible Causes | Solutions |
|---|---|---|
| High background in IF | Inadequate blocking, excessive antibody concentration | Increase blocking time, optimize antibody dilution, include 0.1-0.3% Triton X-100 in antibody diluent |
| Poor signal in fixed tissues | Epitope masking due to fixation | Try alternative fixation methods (e.g., methanol, acetone), perform antigen retrieval |
| Non-specific bands in Western blot | Cross-reactivity with other H1 variants | Use gradient gels for better separation, more stringent washing, competitive blocking with peptides |
| Variable staining intensity | Cell cycle-dependent expression of HIST1H1E | Synchronize cells, co-stain with cell cycle markers |
| Loss of signal over time | Antibody degradation | Store in small aliquots, avoid freeze-thaw cycles, add carrier protein (BSA) |
| Inconsistent ChIP results | Variable chromatin preparation | Standardize fixation time, optimize sonication, use internal controls for normalization |
For particularly challenging applications, consider using alternative detection methods, such as genetically encoded tags (HA, FLAG) for which highly specific antibodies are available .
Optimization of chromatin extraction for efficient HIST1H1E detection requires consideration of this protein's dynamic association with chromatin:
Fixation optimization:
For chemical crosslinking: Use 1-2% formaldehyde for 10-15 minutes (avoid over-fixation)
For native chromatin: Extract in low-salt buffers (≤150 mM NaCl) to maintain H1 association
Extraction buffer composition:
Include divalent cations (2-5 mM MgCl₂) to stabilize chromatin structure
Add protease inhibitors to prevent HIST1H1E degradation
Consider phosphatase inhibitors to maintain native phosphorylation state
Fractionation approach:
For comprehensive HIST1H1E analysis, collect multiple chromatin fractions using sequential extraction:
a. Low salt buffer (e.g., 10 mM Tris-HCl, pH 7.4, 2.5 mM MgCl₂, 0.5% NP-40) for soluble fraction
b. Nuclease digestion (MNase) for loosely bound chromatin fraction
c. High salt extraction (300-600 mM NaCl) for tightly bound fraction
Handling considerations:
Perform extractions at 4°C to minimize protein degradation
Avoid excessive mechanical shearing before intended fragmentation
For ChIP applications, optimize sonication to achieve 200-500 bp fragments
These optimizations enhance HIST1H1E retention and detection across multiple experimental platforms.
Based on recent research, a sophisticated cellular model for Rahman syndrome can be established using CRISPR/Cas9 genome engineering in murine embryonic stem cells (mESCs) . The key methodological steps include:
Design and cloning of CRISPR components:
Design sgRNAs targeting the mouse H1.4 locus using tools like Chopchop
Clone sgRNAs into a Cas9-GFP expression vector (e.g., pX458)
Create a targeting vector containing:
5' and 3' homology arms from mouse H1.4 locus
Human H1.4 gene with HA tag
Promoterless neomycin selection marker flanked by loxP sites
Photoactivatable GFP (PA-GFP) tag
Cell transfection and selection:
Transfect mESCs with the CRISPR/Cas9 and targeting vectors
Select transfected cells with Geneticin (200 μg/mL) for 2 weeks
Pick and expand drug-resistant colonies with undifferentiated morphology
Genotyping and validation:
Extract DNA from selected clones
Perform PCR to confirm successful recombination at both 5' and 3' homology arms
Use restriction enzyme digestion (e.g., HindIII) to determine zygosity
Validate expression through RT-PCR and western blotting
Cre recombinase-mediated induction:
Treat validated clones with Cre Recombinase Adenovirus
Allow 36 hours for transgene expression
Confirm induction through RT-PCR and western blotting
This model enables detailed investigations of HIST1H1E dynamics, including protein interactions, nuclear localization, and functional consequences of Rahman syndrome mutations .
Several cell-based assays can provide insights into the functional impact of HIST1H1E mutations:
Chromatin relaxation assessment:
MNase digestion assay to measure chromatin accessibility
ATAC-seq to map open chromatin regions genome-wide
DNase-seq to identify hypersensitive sites
Histone modification analysis:
Immunofluorescence or ChIP for key histone marks (H3K4me2, H3K9me3, H3K27me3)
Western blotting to quantify global levels of histone modifications
Mass spectrometry for comprehensive PTM profiling
Cell proliferation and cycle analysis:
Growth curve generation by daily cell counting
Flow cytometry with propidium iodide staining for cell cycle distribution
EdU incorporation assays for DNA synthesis assessment
Gene expression profiling:
RNA-seq to identify differentially expressed genes
RT-qPCR validation of key target genes
Analysis of pluripotency markers (Sox2, Oct4) in stem cell models
Protein interaction studies:
Co-immunoprecipitation with HA-tagged HIST1H1E
Proximity ligation assay for in situ detection of protein interactions
ChIP-seq to map genome-wide binding patterns
These assays have revealed that Rahman syndrome mutations lead to enhanced chromatin relaxation, reduced methylation of specific histone H3 lysine residues, and decreased binding of heterochromatin protein HP1β .
Post-translational modifications (PTMs) of HIST1H1E play critical roles in regulating its function and can impact antibody recognition:
| Modification | Sites | Functional Impact | Effect on Antibody Recognition |
|---|---|---|---|
| Phosphorylation | Ser/Thr residues in CTD | Reduces chromatin binding affinity, promotes chromatin decompaction, cell cycle-dependent regulation | May mask epitopes, especially for phospho-sensitive antibodies |
| Acetylation | Multiple Lys residues | Weakens DNA binding, associated with transcriptionally active regions | Can reduce recognition by antibodies targeting modified lysines |
| Methylation | Lys residues | Context-dependent effects on chromatin structure | May interfere with antibody binding depending on epitope location |
| ADP-ribosylation | Multiple sites | Reduces DNA binding affinity, involved in DNA damage response | Can sterically hinder antibody access to epitopes |
| Ubiquitination | Lys residues | Signals for protein turnover | May prevent antibody binding due to steric hindrance |
When using HIST1H1E (Ab-51) antibody, which targets the region around Lys-51, consider that modifications at or near this residue could affect antibody binding efficiency. If studying specific PTMs, specialized phospho-specific or other modification-specific antibodies may be required. For comprehensive PTM analysis, mass spectrometry approaches are recommended to complement antibody-based detection methods.
Recent structural studies have provided significant insights into HIST1H1E's role in chromatin architecture:
Nucleosome binding asymmetry:
Linker DNA organization:
Chromatin fiber formation:
By bringing linker DNA segments closer, HIST1H1E promotes formation of zigzag-type chromatin fibers
HIST1H1E stabilizes nucleosome-nucleosome contacts in higher-order structures
The CTD's high positive charge neutralizes negative charges on linker DNA
These structural insights explain why Rahman syndrome mutations in the CTD significantly impact chromatin organization, leading to enhanced chromatin relaxation and altered gene expression patterns observed in patient cells .
Several promising research directions could benefit from application of HIST1H1E (Ab-51) antibody:
Single-cell epigenomics:
Combining antibody-based detection with single-cell technologies
Mapping HIST1H1E distribution in heterogeneous cell populations
Correlating HIST1H1E binding patterns with cell-specific gene expression programs
Brain development and neurological disorders:
Investigating HIST1H1E's role in neural differentiation
Comparing chromatin organization in control versus Rahman syndrome models
Exploring potential therapeutic approaches targeting chromatin structure
Epigenetic inheritance:
Examining HIST1H1E dynamics during gametogenesis and early embryonic development
Assessing potential transgenerational effects of altered HIST1H1E function
Investigating interactions with DNA methylation machinery
Chromatin phase separation:
Exploring HIST1H1E's contribution to biomolecular condensate formation
Analyzing how CTD mutations affect phase separation properties
Developing assays to visualize and quantify chromatin compartmentalization
Therapeutic targeting:
Screening for compounds that normalize chromatin structure in disease models
Developing peptide-based approaches to mimic normal HIST1H1E function
Exploring gene therapy approaches for Rahman syndrome
These research directions could significantly advance our understanding of chromatin biology and potentially lead to therapeutic interventions for chromatin-related disorders.