Mono-methylation at lysine 16 of histone H4 (H4K16me1) is a post-translational modification critical for chromatin compaction, transcriptional regulation, DNA repair, and epigenetic memory . The Mono-Methyl-Histone H4 (K16) Recombinant Monoclonal Antibody is a highly specific tool designed to detect this modification, enabling researchers to study its role in cellular processes and disease mechanisms .
The antibody is generated through a multi-step process:
Cloning: Genes encoding the antibody’s heavy and light chains are cloned into expression vectors .
Transfection: Vectors are introduced into host cells (e.g., HEK293F) for antibody production .
Validation:
H4K16me1 is enriched in heterochromatin and associated with transcriptional repression .
Plays a role in DNA damage repair by recruiting repair proteins to sites of double-strand breaks .
Altered H4K16me1 levels correlate with cancer progression, including breast and colorectal cancers .
Serves as a potential diagnostic marker for chromatin-related disorders .
IF Staining: Distinct nuclear localization in HeLa cells confirmed via Alexa Fluor 488 labeling .
ChIP-seq Data: Demonstrates enrichment at transcription start sites in conjunction with other histone marks (e.g., H3K9me3) .
The Mono-methyl-Histone H4 (K16) Recombinant Monoclonal Antibody is generated through a meticulous multi-step process. It begins with cloning the genes responsible for encoding the HIST1H4A antibody, encompassing both the heavy and light chains, and integrating them into expression vectors optimized for high performance. These vectors are then transfected into host cells, enabling the production and secretion of the antibody. The antibody is subsequently purified using affinity chromatography to ensure its purity and efficacy. Finally, it undergoes rigorous ELISA and IF tests to guarantee its precise and reliable detection of human and rat HIST1H4A proteins mono-methylated at K16.
The mono-methylation of HIST1H4A at K16 plays a vital role in various cellular processes. It is a critical epigenetic modification that contributes to chromatin compaction, transcriptional repression, DNA repair, cellular identity, long-range chromatin interactions, and epigenetic memory, thus impacting gene expression and chromatin structure. This modification is implicated in the development and progression of various diseases.
Histone H4 is a core component of the nucleosome, the fundamental unit of chromatin. Nucleosomes wrap and compact DNA, restricting its accessibility to cellular machineries that require DNA as a template. Therefore, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. The accessibility of DNA is meticulously regulated through a complex set of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Mono-methyl-Histone H4 (K16) refers to histone H4 protein that has a single methyl group attached to the lysine residue at position 16. This specific modification is part of the histone code that regulates chromatin accessibility and transcriptional activity. H4K16 methylation is particularly important because it occurs in a region of the histone that directly contacts the DNA, potentially affecting nucleosome stability and higher-order chromatin structure . Unlike the better-studied H4K20 methylation, H4K16 methylation represents a distinct regulatory mechanism that influences both gene silencing and activation depending on cellular context and neighboring modifications.
Recombinant monoclonal antibodies for histone research are produced through recombinant DNA technology rather than traditional hybridoma methods. This approach offers several advantages in histone modification research:
Increased specificity: Recombinant antibodies are engineered to recognize specific epitopes with high precision, crucial for distinguishing between closely related histone modifications (mono-, di-, and tri-methylation) .
Reduced batch-to-batch variability: Unlike conventional antibodies, recombinant antibodies have consistent performance across different production lots.
Defined sequence: The exact amino acid sequence is known, enabling better characterization and quality control.
Renewable source: Once the DNA sequence is established, the antibody can be produced indefinitely without relying on immunized animals.
This consistency is particularly important for longitudinal studies on histone modifications where experimental reproducibility is crucial .
Based on current research protocols and manufacturer specifications, Mono-methyl-Histone H4 (K16) antibodies have been validated for the following applications:
Application | Validated | Optimal Dilution | Special Considerations |
---|---|---|---|
Western Blot (WB) | Yes | 1:1000-1:2000 | Best results with acid-extracted histones |
Immunofluorescence (IF) | Yes | 1:100-1:500 | Requires proper fixation with paraformaldehyde |
Immunocytochemistry (ICC) | Yes | 1:100-1:500 | Compatible with methanol fixation |
Chromatin Immunoprecipitation (ChIP) | Yes | 2-5 μg per IP | Critical for genome-wide profiling |
ChIP-sequencing | Yes | 5 μg per assay | Requires rigorous optimization |
Flow Cytometry | Limited | 1:50-1:200 | Permeabilization step crucial |
Dot Blot | Yes | 1:5000 | Useful for antibody validation |
Similar antibodies like Acetyl-Histone H4 (K16) have demonstrated excellent results in multiple applications including WB, ICC, IF, and IHC-p, suggesting comparable performance for the mono-methyl variant .
When selecting between different antibodies targeting H4K16 modifications (mono-methylation, acetylation, etc.), researchers should consider:
Specificity validation: Confirm that the antibody can distinguish between mono-methylation and other modifications (di-/tri-methylation or acetylation) at K16 of histone H4.
Cross-reactivity profile: Check for potential cross-reactivity with similar modifications on other histones, particularly H3K16.
Application compatibility: Ensure the antibody has been validated for your specific application (ChIP, IF, WB, etc.).
Clone information: Recombinant monoclonal antibodies from established clones generally show better consistency than polyclonal alternatives.
Publication record: Previously published studies using the antibody provide evidence of reliability.
A peptide competition assay or dot blot array against modified and unmodified histone peptides can verify specificity before proceeding with experiments .
Sample preparation is critical for accurate detection of H4K16 mono-methylation. Here are methodological recommendations:
For Western Blot:
Direct acid extraction: Treat cells with 0.2N HCl for 30 minutes at 4°C to extract histones.
Histone purification kits: Commercial kits that preserve post-translational modifications are preferable.
Buffer considerations: Include histone deacetylase inhibitors (5mM sodium butyrate) and protease inhibitors to prevent modification loss.
Loading amount: 5-15 μg of acid-extracted histones typically yields optimal results.
For ChIP and ChIP-seq:
Crosslinking: 1% formaldehyde for 10 minutes at room temperature.
Sonication: Optimize to achieve chromatin fragments of 200-500 bp.
Pre-clearing: Always pre-clear chromatin with protein A/G beads before immunoprecipitation.
Input control: Reserve 5-10% of chromatin before immunoprecipitation as input control.
For Immunofluorescence:
Fixation: 4% paraformaldehyde for 10 minutes followed by permeabilization with 0.5% Triton X-100.
Blocking: 5% BSA in PBS for 1 hour to reduce background.
Antigen retrieval: Citrate buffer (pH 6.0) treatment can improve epitope accessibility.
The performance of H4K16me1 antibodies is significantly affected by fixation methods, with over-fixation potentially masking the epitope .
A robust experimental design should include the following controls:
Positive Controls:
Cell lines with known H4K16me1 enrichment (e.g., certain embryonic stem cell lines)
Recombinant histones with defined H4K16me1 modification
Synthetic peptides with H4K16me1 modification
Negative Controls:
IgG control from the same species as the primary antibody
Unmodified histone H4 peptides
Samples treated with H4K16 demethylase enzymes
H4K16A mutant cell lines (where lysine is replaced with alanine)
Specificity Controls:
Peptide competition assays using H4K16me1, H4K16me2, H4K16me3, and H4K16ac peptides
Dot blot analysis with modified and unmodified peptides
Western blot of acid-extracted histones from knockdown/knockout cells lacking the enzyme responsible for H4K16 mono-methylation
Including these controls allows proper validation of antibody specificity and experimental results .
Accurate quantification of H4K16 mono-methylation can be achieved through several methodologies:
Western Blot Quantification:
Normalize H4K16me1 signal to total H4 or housekeeping protein
Use standard curves with recombinant H4K16me1 for absolute quantification
Employ digital imaging systems with linear dynamic range for signal detection
ChIP-qPCR Quantification:
Calculate percent input method: (IP signal ÷ input signal) × 100
Relative enrichment: Compare target regions to known negative regions
Spike-in normalization using exogenous chromatin from another species
Mass Spectrometry Methods:
MRM (Multiple Reaction Monitoring) for targeted quantification
SILAC (Stable Isotope Labeling with Amino acids in Cell culture) for relative quantification
Parallel Reaction Monitoring (PRM) for increased selectivity
The most reliable results combine multiple approaches, particularly pairing antibody-based methods with mass spectrometry validation to overcome potential antibody cross-reactivity issues .
Researchers frequently encounter these challenges when working with H4K16me1 antibodies:
Issue | Possible Causes | Recommended Solutions |
---|---|---|
High background | Non-specific binding | Increase blocking concentration; optimize antibody dilution; include Tween-20 in wash buffers |
Weak or no signal | Epitope masking; low abundance | Try alternative fixation methods; enrich for histones; increase antibody concentration |
Cross-reactivity | Antibody recognizing similar modifications | Validate with peptide arrays; use more specific clone; perform peptide competition |
Inconsistent results | Batch-to-batch variability | Use recombinant monoclonal antibodies; maintain consistent lot numbers |
Cell type variations | Different H4K16me1 abundance | Include positive control cell lines; adjust exposure times |
Poor ChIP efficiency | Inefficient crosslinking | Optimize crosslinking time; ensure chromatin is properly sonicated |
Pre-adsorption of the antibody against related modified peptides can significantly improve specificity in challenging applications .
Modern epigenetic research often requires simultaneous detection of multiple histone modifications. For multiplex applications with H4K16me1 antibodies:
Co-Immunoprecipitation Strategies:
Sequential ChIP (Re-ChIP): Perform initial IP with H4K16me1 antibody, followed by a second IP with antibody against another modification to identify co-occurrence.
Parallel ChIP: Perform separate IPs from the same chromatin preparation for comparative analysis.
Multicolor Immunofluorescence:
Select primary antibodies from different host species (e.g., rabbit anti-H4K16me1 with mouse anti-H3K9me3).
Use highly cross-adsorbed secondary antibodies conjugated to spectrally distinct fluorophores.
Include appropriate negative controls for each antibody.
Employ spectral unmixing for closely overlapping fluorophores.
Mass Cytometry (CyTOF):
Conjugate H4K16me1 antibody to a unique metal isotope.
Combine with other histone mark antibodies conjugated to different metals.
Analyze single-cell epigenetic profiles with high-dimensional data analysis.
When designing multiplex experiments, consider potential steric hindrance between antibodies binding to neighboring epitopes on the histone tail .
H4K16 mono-methylation functions within a complex network of histone modifications. Current research has identified several key relationships:
Related Modification | Relationship with H4K16me1 | Functional Consequence |
---|---|---|
H4K16ac | Mutually exclusive | Transition from active to poised state |
H3K9me3 | Often co-occurs | Enhanced heterochromatin formation |
H3K4me3 | Context-dependent | Bivalent domains in developmental genes |
H4K20me1 | Sequential | Cell cycle regulation |
H2AK119ub | Co-occurs in repressed regions | Polycomb-mediated silencing |
ChIP-seq studies have shown that H4K16me1 distribution patterns differ significantly from H4K16ac, with mono-methylation more prevalent in facultative heterochromatin regions. The dynamic interplay between methylation and acetylation at H4K16 appears to be a key regulatory mechanism for transitioning between active and repressed chromatin states .
Designing and analyzing ChIP-seq experiments for H4K16me1 requires specific considerations:
Experimental Design:
Input normalization: Always sequence an input control from the same chromatin preparation.
Spike-in controls: Consider adding exogenous chromatin from another species (e.g., Drosophila) for quantitative normalization.
Biological replicates: Minimum of three biological replicates is recommended.
Sequencing depth: Aim for 20-30 million uniquely mapped reads per sample.
Fragment size selection: 200-500bp fragments typically provide optimal results.
Data Analysis Pipeline:
Quality control: Filter low-quality reads and adapter sequences.
Alignment: Map to reference genome with tools like Bowtie2 or BWA.
Peak calling: Use MACS2 with broad peak settings (H4K16me1 typically gives broader peaks than sharp transcription factor binding sites).
Differential binding analysis: Compare between conditions using DiffBind or similar tools.
Integration with other data: Correlate with RNA-seq, other histone marks, and chromatin accessibility data.
Visualization and Interpretation:
Generate heatmaps centered on transcription start sites, enhancers, or other features.
Use genome browsers like IGV or UCSC to visualize specific loci.
Perform Gene Ontology enrichment analysis on genes associated with H4K16me1 peaks.
Consider chromatin state analysis using tools like ChromHMM to identify H4K16me1-associated states.
The broad nature of H4K16me1 peaks sometimes requires specialized analysis approaches compared to sharp transcription factor peaks .
Recent research has revealed several important functions of H4K16 mono-methylation:
Developmental Regulation:
Studies have shown that H4K16me1 levels change dynamically during embryonic development, suggesting a role in cell fate decisions and developmental transitions. The modification appears particularly important during gastrulation and neuronal differentiation.
Disease Associations:
Altered H4K16me1 patterns have been observed in several pathological conditions:
Disease | H4K16me1 Pattern | Potential Mechanism |
---|---|---|
Various cancers | Generally decreased | Dysregulation of heterochromatin |
Neurodegenerative disorders | Region-specific changes | Altered gene expression in neurons |
Inflammatory conditions | Increased at specific loci | Enhanced expression of inflammatory genes |
Aging | Global decrease | Chromatin destabilization |
Cell Cycle Regulation:
H4K16me1 shows dynamic changes throughout the cell cycle, with levels peaking during G1 phase and decreasing during S phase. This pattern suggests coordination with DNA replication timing and potential roles in maintaining genome integrity.
Chromatin Structure:
Recent biophysical studies indicate that H4K16me1 affects internucleosomal interactions differently than H4K16ac, potentially promoting a more condensed chromatin structure while still allowing for regulated accessibility of specific factors .
Single-cell epigenomic technologies are revolutionizing our understanding of histone modifications including H4K16me1:
Single-Cell Technologies:
Single-cell CUT&Tag: Allows profiling of H4K16me1 in individual cells, revealing cell-to-cell heterogeneity.
Single-cell ChIP-seq: Though technically challenging, protocols have been optimized for histone modifications.
scNOMe-seq: Combines accessibility and methylation profiling at single-cell resolution.
Mass cytometry (CyTOF): Enables quantification of H4K16me1 alongside other protein markers in thousands of individual cells.
Analytical Approaches:
Trajectory analysis: Maps H4K16me1 changes during cellular transitions and differentiation.
Clustering algorithms: Identifies cell subpopulations with distinct H4K16me1 patterns.
Integration with scRNA-seq: Correlates H4K16me1 patterns with transcriptional states in the same cells.
Research Applications:
Developmental biology: Tracking epigenetic changes during lineage specification.
Cancer heterogeneity: Identifying epigenetically distinct subclones within tumors.
Aging research: Examining cell-specific epigenetic drift in H4K16me1 patterns.
These approaches reveal that H4K16me1 distribution shows greater cell-to-cell variability than previously appreciated, potentially contributing to cellular plasticity and response to environmental signals .
The accurate detection and distinction of H4K16 mono-methylation presents several technical challenges:
Antibody Specificity Limitations:
Structural similarity: The chemical structures of mono-, di-, and tri-methylated lysines are very similar, making absolute specificity difficult.
Context dependence: Neighboring modifications can affect antibody recognition efficiency.
Validation requirements: Each new antibody lot requires rigorous validation against modified peptide arrays.
Mass Spectrometry Challenges:
Peptide fragmentation: H4 tryptic peptides containing K16 are often suboptimal for MS/MS analysis.
Co-occurrence of modifications: Multiple modifications on the same peptide complicate analysis.
Quantification accuracy: Ion suppression can affect quantitative comparisons.
Emerging Solutions:
Recombinant antibody engineering: Using phage display to generate highly specific antibodies.
Middle-down proteomics: Analyzing larger histone fragments to preserve modification patterns.
Targeted MS approaches: Multiple Reaction Monitoring (MRM) for specific detection of H4K16me1.
Specialized enrichment: Using combinatorial antibody approaches to increase specificity.
The table below summarizes methods for distinguishing between different H4K16 modifications:
Method | Advantages | Limitations | Best For |
---|---|---|---|
Specific antibodies | In situ detection possible | Cross-reactivity | ChIP, IF, IHC |
Middle-down MS | Multiple marks on same peptide | Lower throughput | Detailed PTM mapping |
Chemical derivatization | Distinguishes methylation states | Complex workflow | Mass spec analysis |
SNAP-ChIP | Quantitative assessment | Requires specialized reagents | Antibody validation |
Researchers are increasingly using orthogonal approaches to confirm H4K16me1 identification, combining antibody-based detection with mass spectrometry validation .
The establishment and removal of H4K16 mono-methylation involve specific enzymes with regulated activities:
Writer Enzymes (Methyltransferases):
Several methyltransferases have been implicated in H4K16 mono-methylation, including members of the SET domain family. These enzymes show remarkable specificity:
Substrate recognition: Specific amino acid sequences flanking K16 are recognized by the enzyme.
Product specificity: Some enzymes can add only one methyl group (mono-methylation), while others can catalyze successive methylation reactions.
Regulatory mechanisms: Writer activity is often regulated by:
Post-translational modifications of the enzyme itself
Interaction with scaffold proteins
Chromatin context and neighboring modifications
Cell cycle-dependent expression
Eraser Enzymes (Demethylases):
Lysine-specific demethylases that target H4K16me1 include members of the KDM family:
Reaction mechanism: Most use FAD-dependent amine oxidation or Fe(II) and α-ketoglutarate-dependent hydroxylation.
Specificity determinants: Structural features that distinguish mono-methylation from di- or tri-methylation.
Context-dependent activity: Often function within multi-protein complexes that target them to specific genomic regions.
Regulation of Writer/Eraser Balance:
The dynamic equilibrium between methylation and demethylation at H4K16 is regulated by:
Metabolic state: SAM/SAH ratio affects methyltransferase activity
Oxygen levels: Many demethylases are oxygen-dependent
Signaling pathways: Growth factor and stress signaling modulate enzyme activity
Development: Expression levels of writers and erasers change during differentiation
Understanding these enzymes provides potential therapeutic targets for diseases with dysregulated H4K16 methylation patterns .
Advanced computational methods have been developed to analyze the complex distribution and functional implications of H4K16me1:
Peak Calling and Analysis:
Specialized algorithms: Modified versions of MACS2 optimized for broad histone marks
Signal processing: Wavelet-based methods for identifying diffuse enrichment patterns
Differential binding: DESeq2 or edgeR-based approaches for comparing conditions
Integration with Other Data Types:
Multi-omics integration: Correlating H4K16me1 with:
Transcriptome (RNA-seq)
Chromatin accessibility (ATAC-seq, DNase-seq)
DNA methylation
Other histone modifications
Genomic feature association: Enhancer-gene linking algorithms to connect H4K16me1-marked enhancers with target genes
Chromatin state modeling: Using ChromHMM or similar tools to define H4K16me1-containing chromatin states
Pattern Recognition and Machine Learning:
Supervised learning: Training classifiers to predict H4K16me1 locations from DNA sequence and other features
Deep learning: Convolutional neural networks to identify complex patterns associated with H4K16me1 enrichment
Motif analysis: Identifying transcription factor binding motifs enriched in H4K16me1 regions
Visualization Strategies:
Genome browsers with multiple track support
Metaplot analysis around features of interest (TSS, enhancers, etc.)
Heatmaps clustered by pattern similarity
3D chromatin interaction visualization to link H4K16me1 with higher-order structure
These computational approaches have revealed that H4K16me1 distribution follows distinct patterns from other histone marks, often forming broader domains and showing unique associations with chromatin compartments and gene expression states .
Despite significant advances, several limitations remain in H4K16me1 research:
Technical Limitations:
Antibody specificity: Even the best antibodies show some cross-reactivity with other methylation states.
Low abundance: H4K16me1 can be present at relatively low levels in some cell types, making detection challenging.
Dynamic modification: The transient nature of the modification during cellular processes complicates analysis.
Knowledge Gaps:
Cell type specificity: Comprehensive maps across diverse cell types are lacking.
Writer/eraser enzymes: The complete set of enzymes specifically targeting H4K16me1 remains incompletely characterized.
Functional consequences: Direct causality between H4K16me1 and specific gene expression outcomes is often unclear.
Future Approaches to Address Limitations:
Development of highly specific recombinant antibodies through directed evolution
Implementation of more sensitive detection methods, including proximity ligation assays
CRISPR-based approaches to modulate H4K16me1 at specific loci
Single-molecule imaging to track H4K16me1 dynamics in living cells
Computational integration of multi-omics data to infer functional relationships
The field is increasingly moving toward causal experimental designs rather than correlative studies, which will significantly advance our understanding of H4K16me1 function .
Emerging research suggests several potential clinical applications related to H4K16 mono-methylation:
Diagnostic Biomarkers:
H4K16me1 patterns show alterations in several diseases, particularly cancer, suggesting potential as diagnostic or prognostic biomarkers. For example:
Disease | H4K16me1 Pattern | Potential Clinical Application |
---|---|---|
Breast cancer | Decreased at tumor suppressors | Early detection biomarker |
Glioblastoma | Regional increases | Tumor classification |
Neurodegenerative disease | Altered neuronal patterns | Disease progression marker |
Inflammatory disorders | Increased at specific loci | Treatment response prediction |
Therapeutic Targeting:
Small molecule inhibitors: Development of specific inhibitors targeting writer or eraser enzymes for H4K16me1
Degrader approaches: Proteolysis-targeting chimeras (PROTACs) to selectively degrade H4K16me1 regulatory machinery
Epigenetic editing: CRISPR-based approaches to modify H4K16me1 at specific genomic loci
Monitoring Treatment Response:
Changes in global or locus-specific H4K16me1 patterns could serve as pharmacodynamic biomarkers for epigenetic therapies, helping to determine optimal dosing and treatment schedules.
Precision Medicine Applications:
Integrating H4K16me1 profiles with other molecular data could help stratify patients for targeted therapies, particularly in cancers where epigenetic dysregulation plays a key role.
While these applications show promise in preclinical research, significant validation and standardization will be required before clinical implementation .
Several emerging technologies are revolutionizing our ability to study H4K16 mono-methylation:
Cutting-Edge Genomic Technologies:
CUT&Tag and CUT&RUN: More efficient alternatives to traditional ChIP with lower input requirements and improved signal-to-noise ratio
Cleavage Under Targets and Release Using Nuclease (CUT&RUN): Provides higher resolution mapping of H4K16me1
TAF-ChIP: Targets alternative features of transcription to improve specificity
Spatial Technologies:
Imaging mass cytometry: Allows visualization of H4K16me1 in tissue sections with cellular resolution
Spatial-CUT&Tag: Combines epigenomic profiling with spatial information
Super-resolution microscopy: Visualizes H4K16me1 distribution at nanoscale resolution
Single-Cell Approaches:
scCUT&Tag: Profiles H4K16me1 in individual cells
scMultiome: Simultaneously profiles chromatin modifications and gene expression
Live-cell histone modification sensors: Enables real-time monitoring of H4K16me1 dynamics
Proteomics Innovations:
SNAP-ChIP: Quantitatively assesses antibody specificity using modified designer nucleosomes
Targeted proteomics: Improves sensitivity for detecting low-abundance histone modifications
Crosslinking mass spectrometry: Maps protein interactions with H4K16me1-containing chromatin
Computational Advances:
Deep learning approaches for integrating multi-omics data
Network analysis tools to understand H4K16me1 in broader regulatory contexts
Spatial modeling of chromatin to link H4K16me1 with 3D genome organization
These technologies are expected to provide unprecedented insights into the dynamic regulation and functional consequences of H4K16 mono-methylation across diverse biological contexts .