Acetyl-HIST1H1E (K45) Antibody is a polyclonal rabbit antibody designed to detect acetylation at lysine residue 45 (K45) of the histone H1.4 protein, encoded by the HIST1H1E gene . This epigenetic modification is critical for chromatin structure regulation, DNA accessibility, and gene expression modulation.
The antibody specifically recognizes the acetylated form of H1.4, distinguishing it from unmodified or other post-translationally modified variants. Its target lies within the protein’s globular domain, which interacts with linker DNA to stabilize nucleosome organization .
The antibody has been validated for:
Western Blot (WB): Detects endogenous H1.4 in HEK293T cells and ectopic overexpression models .
Immunocytochemistry (ICC/IF): Identifies nucleolar localization of acetylated H1.4 in human embryonic stem cells (e.g., H7 cell line) .
Chromatin Dynamics: Acetylation at K45 reduces the positive charge of H1.4’s C-terminal tail, potentially weakening DNA binding and promoting open chromatin states .
Gene Regulation: Altered acetylation patterns may disrupt transcriptional regulation, contributing to neurodevelopmental disorders linked to HIST1H1E mutations (e.g., Rahman syndrome) .
Application | Dilution | Notes |
---|---|---|
ELISA | Optimized by user | - |
ICC/IF | 1:10–1:100 | Strong nuclear signal in fixed cells . |
WB | 1:500–1:2000 | Detects ~21 kDa band for H1.4 . |
Comparison based on cross-referenced data .
Disease Modeling: H1.4 acetylation defects could mimic HIST1H1E mutation phenotypes (e.g., overgrowth, intellectual disability) observed in Rahman syndrome .
Therapeutic Potential: Targeting H1.4 acetylation may offer pathways to modulate chromatin architecture in developmental disorders or cancers.
HIST1H1E (Histone H1.4) is a member of the linker histone family that binds to linker DNA between nucleosomes, facilitating the formation of higher-order chromatin structures. It plays a crucial role in chromatin fiber compaction and acts as a regulator of individual gene transcription through its involvement in chromatin remodeling, nucleosome spacing, and DNA methylation . The protein is 219 amino acids in length and belongs to the Histone H1/H5 family . Functionally, HIST1H1E contributes to efficient compaction of the genome and proper chromosomal segregation during cell division, while also supporting DNA replication, transcription, and repair processes .
K45 (Lysine 45) acetylation of HIST1H1E represents a specific post-translational modification (PTM) that occurs within the region spanning amino acids 43-55 of the protein . This acetylation site is located in a functionally important region of the histone and likely influences the protein's interaction with DNA and other nuclear components. Post-translational modifications of linker histones like HIST1H1E contribute significantly to the functional diversity of these proteins in chromatin arrangement and cellular processes . K45 acetylation specifically may alter the binding affinity of HIST1H1E to chromatin and potentially regulate its role in gene expression and chromatin compaction.
Acetylation is one of several post-translational modifications that affect linker histones. While early studies of linker histone PTMs were limited by technical challenges such as difficulties in separating linker histone subtypes and the lack of sensitive detection methods, advances in mass spectrometry and related techniques have greatly expanded our understanding .
PTMs identified in linker histones include:
Modification Type | Common Sites | Functional Implications |
---|---|---|
Acetylation | Lysine residues (e.g., K45, K51) | Alters DNA binding, affects chromatin accessibility |
Phosphorylation | Serine/Threonine residues | Cell cycle regulation, chromatin condensation |
Methylation | Lysine residues | Gene expression regulation |
Ubiquitination | Lysine residues | Protein turnover, signaling |
Formylation | Various residues | Recently identified, function being studied |
Detection methods have evolved from 32P labeling and Edman degradation to sophisticated techniques including HPLC, HILIC, HPCE, and mass spectrometry approaches like LTQ-FT-ICR and LTQ-Orbitrap, providing high resolution and mass accuracy for reliable identification of specific PTMs .
Acetyl-HIST1H1E (K45) antibodies have been validated for several experimental applications in epigenetic and chromatin research. Based on the available data, these antibodies can be reliably used for:
ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative detection of acetylated HIST1H1E
ICC (Immunocytochemistry): For cellular localization studies, typically at dilutions of 1:10-1:100
IF (Immunofluorescence): For visualizing the distribution of acetylated HIST1H1E in cells
ChIP (Chromatin Immunoprecipitation): For investigating chromatin-associated functions and identifying genomic binding sites
These applications enable researchers to study the distribution, abundance, and functional associations of acetylated HIST1H1E at K45 in various experimental contexts.
When validating the specificity of Acetyl-HIST1H1E (K45) antibodies, researchers should include the following controls:
Peptide Competition Assay: Pre-incubate the antibody with excess synthetic peptide containing acetylated K45 to demonstrate specific blocking of antibody binding.
Non-acetylated Peptide Control: Test antibody reactivity against the same peptide sequence without acetylation to confirm modification specificity.
Alternative Acetylation Site Control: Use peptides with acetylation at other lysine residues (e.g., K51) to verify site-specificity .
Knockout/Knockdown Controls: If available, test samples from HIST1H1E knockout/knockdown models to confirm absence of signal.
Histone Deacetylase (HDAC) Inhibitor Treatment: Compare samples from cells treated with and without HDAC inhibitors to show increased signal with increased acetylation.
Cross-reactivity Assessment: Test against other histone H1 family members to ensure specificity for HIST1H1E.
Comparative Analysis: Use multiple antibodies targeting the same modification from different suppliers to verify consistent results.
Optimizing ChIP protocols for Acetyl-HIST1H1E (K45) antibodies requires attention to several critical parameters:
Crosslinking Optimization: Linker histones like HIST1H1E bind chromatin more dynamically than core histones. Test different formaldehyde concentrations (0.75-2%) and crosslinking times (5-15 minutes) to achieve optimal results.
Sonication Conditions: Carefully optimize sonication to generate chromatin fragments of 200-500 bp, performing pilot experiments with different sonication cycles and amplitudes.
Antibody Amount: Titrate antibody concentrations, testing 2-10 μg per ChIP reaction to determine the optimal amount for specific enrichment.
Washing Stringency: Balance between removing non-specific binding (higher stringency) and preserving specific interactions (lower stringency) by testing different salt concentrations in wash buffers.
Pre-clearing and Blocking: Implement thorough pre-clearing of chromatin and blocking with BSA and non-specific IgG to reduce background.
Sequential ChIP: Consider sequential ChIP (Re-ChIP) approaches if studying co-occurrence with other histone modifications.
Quantification and Controls: Include input controls, IgG controls, and positive/negative control regions for qPCR validation.
The low abundance of specific acetylation marks may require protocol adaptations like increased starting material or reduced washing stringency while maintaining specificity.
Recent studies have connected HIST1H1E dysfunction with cellular senescence and accelerated aging phenotypes . While the specific contribution of K45 acetylation hasn't been fully characterized, research suggests the following potential mechanisms:
Chromatin Compaction Regulation: Acetylation of K45 likely reduces the positive charge of HIST1H1E, potentially decreasing its binding affinity to DNA. This could result in altered chromatin compaction states that affect senescence-associated heterochromatin foci (SAHF) formation.
Transcriptional Reprogramming: Changes in K45 acetylation states may influence the expression of senescence-associated genes through altered accessibility of transcription factors to their target sites.
DNA Damage Response: Acetylation status at K45 may affect how HIST1H1E participates in DNA damage responses, with implications for senescence triggered by persistent DNA damage.
Cell Cycle Regulation: HIST1H1E has been connected to cell cycle progression, with frameshift mutations resulting in cells that "hardly enter into the S phase, and undergo accelerated senescence" . K45 acetylation might modulate these functions.
Epigenetic Landscape: Alterations in K45 acetylation could contribute to broader epigenetic changes associated with cellular aging, including specific methylation patterns observed in cells expressing mutant HIST1H1E proteins .
Research examining the relationship between K45 acetylation states and these senescence pathways would provide valuable insights into epigenetic regulation of cellular aging.
The functional interaction between HIST1H1E K45 acetylation and other histone modifications represents a complex regulatory network in chromatin biology:
Coordination with Core Histone Acetylation: K45 acetylation of HIST1H1E may work cooperatively with acetylation marks on core histones (e.g., H3K27ac, H3K9ac) to create permissive chromatin environments for transcription.
Relationship with Histone Methylation: Evidence suggests potential cross-talk between linker histone modifications and methylation marks on core histones. For example, HIST1H1E acetylation might influence recruitment of proteins that recognize or establish H3K4me3 (active) or H3K9me3 (repressive) marks.
Sequential Modification Patterns: K45 acetylation may participate in sequential modification patterns, where one modification enables or prevents others, creating a temporal code for chromatin regulation.
Reader Protein Interactions: Acetylated K45 likely creates binding sites for specific "reader" proteins containing bromodomains, which could themselves recruit additional chromatin modifiers.
Modification Territories: Different histone modifications often exist in defined genomic territories. Mapping the co-occurrence of K45 acetylation with other modifications using ChIP-seq approaches can reveal functional domains and regulatory units.
Enzyme Crosstalk: The enzymes responsible for K45 acetylation may physically or functionally interact with enzymes that modify other histone residues, suggesting coordinated regulation.
Understanding these interactions requires advanced multi-omics approaches combining ChIP-seq, proteomics, and functional genomics to map modification landscapes and their effects on chromatin structure and function.
Germline frameshift mutations in the C-terminal tail of HIST1H1E have been linked to intellectual disability and premature aging . The relationship between these mutations and K45 acetylation presents an important area for investigation:
Structural Consequences: C-terminal mutations in HIST1H1E may alter the protein's conformation, potentially exposing or masking K45 to acetyltransferases and deacetylases, thereby affecting acetylation levels.
Enzyme Recruitment: Mutant HIST1H1E proteins might have altered interactions with histone acetyltransferases (HATs) and histone deacetylases (HDACs) that target K45, changing the equilibrium of acetylation/deacetylation.
Genomic Distribution: The genomic distribution of K45-acetylated HIST1H1E may be altered in cells with C-terminal mutations, potentially affecting different gene sets than in normal cells.
Stability and Dynamics: While research has shown that C-terminal frameshift mutations result in stable proteins that reside in the nucleus and bind to chromatin , these mutations might alter the dynamics of HIST1H1E binding and exchange, with consequences for K45 acetylation.
Developmental Timing: The temporal pattern of K45 acetylation during development might be disrupted in mutation carriers, contributing to neurodevelopmental phenotypes.
Compensation Mechanisms: Other histone H1 variants might show altered expression or modification patterns to compensate for mutant HIST1H1E function, including changes to acetylation sites equivalent to K45.
Investigating these aspects could provide mechanistic insights into how HIST1H1E mutations contribute to developmental disorders and potentially identify therapeutic targets for intervention.
Researchers frequently encounter several technical challenges when working with Acetyl-HIST1H1E (K45) antibodies:
Low Signal Intensity:
Cause: Relatively low abundance of K45 acetylation in most cell types
Solution: Increase starting material, optimize antibody concentration, use signal amplification methods, or enrich target cells with treatments that increase acetylation (e.g., HDAC inhibitors for controlled experiments)
Background Signal:
Cause: Cross-reactivity with other acetylated histones or proteins
Solution: Implement more stringent washing conditions, increase blocking, pre-absorb antibody with unrelated acetylated peptides, or use more selective secondary antibodies
Inconsistent Results:
Cause: Acetylation levels varying with cell cycle, culture conditions, or processing time
Solution: Standardize cell harvesting procedures, synchronize cells when appropriate, and minimize processing time to prevent enzymatic deacetylation
Epitope Masking:
Cause: K45 may be obscured by chromatin compaction or protein interactions
Solution: Optimize extraction conditions, test different fixation methods, or use epitope retrieval techniques for immunostaining applications
Antibody Lot Variation:
Cause: Different manufacturing batches may have varying specificities
Solution: Validate each new lot against previous lots, maintain reference samples, and consider pooling antibodies from different lots for consistency in long-term projects
Detection in Specific Cell Types:
Cause: Cell-type specific differences in chromatin accessibility
Solution: Adjust permeabilization and extraction protocols for specific cell types, potentially using cell-type specific optimization
When faced with contradictory ChIP-seq data for Acetyl-HIST1H1E (K45) binding patterns, researchers should consider several factors for proper interpretation:
Antibody Specificity Assessment:
Evaluate if different antibodies were used across studies
Re-validate antibody specificity in the specific experimental context
Consider orthogonal approaches like CUT&RUN with alternative antibodies
Biological Variables:
Cell type differences: HIST1H1E distribution varies substantially between cell types
Cell cycle status: Binding patterns may change dramatically through the cell cycle
Differentiation state: Consider if cells were at different developmental stages
Technical Variables:
Crosslinking conditions: Different protocols may capture different subpopulations of HIST1H1E
Chromatin preparation: Sonication vs. enzymatic digestion can affect epitope availability
Sequencing depth: Shallow sequencing might miss low-abundance binding sites
Peak calling algorithms: Different computational approaches can yield different results
Contextual Interpretation:
Consider genomic context of binding sites (promoters, enhancers, etc.)
Analyze correlation with other histone marks and chromatin features
Examine DNA sequence motifs associated with binding sites
Functional Validation:
Perform targeted ChIP-qPCR at discrepant regions
Use genetic approaches (mutation/deletion) to validate functional importance
Consider 3D chromatin organization that might explain apparent contradictions
Integrated Analysis:
Correlate with gene expression data
Perform multivariate analysis incorporating multiple datasets
Consider broader chromatin state maps to resolve contradictions
Determining whether HIST1H1E K45 acetylation is a cause or consequence of observed cellular phenotypes requires sophisticated experimental designs:
Temporal Resolution Studies:
Use time-course experiments with high temporal resolution
Apply rapid induction systems (e.g., auxin-inducible degron) to manipulate factors of interest
Employ real-time imaging of acetylation using engineered reader domains fused to fluorescent proteins
Site-Specific Modification Tools:
Use histone mimetics (K45Q to mimic acetylation, K45R to prevent acetylation)
Apply CRISPR-based epigenome editing to specifically modify K45 acetylation at target loci
Develop and deploy acetylation-specific degron systems
Enzyme Manipulation:
Identify and modulate the specific HATs and HDACs that regulate K45 acetylation
Use rapid chemical inhibition with specific inhibitors
Apply genetic approaches with inducible systems for temporal control
Single-Cell Approaches:
Perform single-cell ChIP-seq or CUT&TAG to capture cellular heterogeneity
Correlate K45 acetylation with cell state markers at single-cell resolution
Track cells through division or differentiation to establish precursor-product relationships
Computational Causal Inference:
Apply causal inference algorithms to multi-omics data
Use dynamic Bayesian networks to model temporal dependencies
Implement structural equation modeling to test causal hypotheses
Orthogonal Validation:
Correlate with multiple independent readouts (transcription, chromatin accessibility)
Use alternative methods to manipulate the system and confirm directionality
Perform rescue experiments to establish necessity and sufficiency
These approaches, particularly when used in combination, can help establish whether K45 acetylation drives specific cellular processes or occurs as a consequence of those processes.