What is the biological significance of histone H4 arginine 35 methylation in chromatin regulation?
Histone H4 arginine 35 (R35) methylation represents a critical modification that influences nucleosome structure and DNA accessibility. This residue is located at the lateral surface of the nucleosome where it interacts with DNA. According to studies of histone lateral surface modifications, R35 is positioned near the N-terminus of the H4 α1 helix with its side chain extended into the major groove of DNA . The methylation of R35 affects chromatin compaction through altered histone-DNA interactions.
Unlike tail modifications, R35 methylation directly influences the structural properties of the nucleosome core by altering the water-mediated hydrogen bonds that normally exist between the unmodified arginine and the DNA phosphate backbone . This modification contributes to the "regulated nucleosome mobility" model, working alongside other modifications to determine chromatin states that impact transcriptional regulation, DNA repair, and replication .
How does histone H4 arginine methylation differ functionally from lysine methylation?
Arginine and lysine methylation on histone H4 serve distinct regulatory functions:
While both modifications can regulate chromatin states, arginine methylation generally functions in different molecular contexts than lysine methylation, with distinct reader proteins and downstream effects .
What cellular processes are regulated by histone H4 arginine methylation?
Histone H4 arginine methylation regulates multiple cellular processes:
Transcriptional activation: H4R3 methylation by PRMT1 is particularly associated with active transcription and facilitates subsequent histone acetylation .
Cell division regulation: Studies have shown that H4R35 methylation is notably enriched on mitotic chromosomes, suggesting a role in cell division processes .
Chromatin architecture: H4 arginine methylation contributes to higher-order chromatin structure by modulating nucleosome-nucleosome interactions .
DNA damage response: Arginine methylation can influence the recruitment of DNA repair factors to damaged chromatin sites.
Development: Changes in H4 arginine methylation patterns occur during cellular differentiation and development, as seen in parasite life cycles studied in T. gondii and P. falciparum .
What are the optimal conditions for using Mono-methyl-HIST1H4A (R35) Antibody in immunofluorescence experiments?
For optimal immunofluorescence results with Mono-methyl-HIST1H4A (R35) Antibody:
Dilution ratio: 1:1-1:10 (as recommended for CSB-PA010429OA35me1HU)
Fixation protocol: 4% formaldehyde fixation is recommended (as demonstrated in successful experiments)
Permeabilization: 0.2% Triton X-100 ensures proper antibody access to nuclear epitopes
Blocking solution: 10% normal goat serum efficiently reduces background
Incubation conditions: Overnight incubation at 4°C provides optimal binding
Secondary antibody: Alexa Fluor 488-conjugated AffiniPure Goat Anti-Rabbit IgG(H+L) works effectively with this primary antibody
Counterstaining: DAPI for nuclear visualization
For reproducible results, it is critical to maintain consistent fixation times, antibody concentrations, and washing steps across experimental replicates.
What controls should be included when using Mono-methyl-HIST1H4A (R35) Antibody?
Rigorous controls are essential for reliable histone modification analysis:
Peptide competition assay: Pre-incubate antibody with synthetic H4R35me1 peptide to confirm specificity (similar to approaches used for other histone methylation antibodies)
Dot-blot specificity test: Test antibody against a panel of modified and unmodified histone peptides to ensure it doesn't cross-react with similar modifications (H4K31me1, H4K20me1, etc.)
Negative controls:
Secondary antibody-only control
Isotype control (rabbit IgG at equivalent concentration)
Unmodified histone H4 peptide control
Positive controls:
Histone demethylase treatment: As a functional control, treating samples with appropriate demethylases should reduce antibody signal
How can ChIP-seq be optimized for Mono-methyl-HIST1H4A (R35) Antibody?
For successful ChIP-seq with Mono-methyl-HIST1H4A (R35) Antibody:
Cross-linking optimization: 1% formaldehyde for 10 minutes at room temperature is typically effective for histone modifications
Chromatin fragmentation: Aim for fragments of 150-300 bp using either sonication or enzymatic digestion
Antibody amount: Start with 2-5 μg antibody per ChIP reaction and optimize based on preliminary results
Immunoprecipitation conditions:
Library preparation: Use specialized library preparation kits designed for limited ChIP material
Sequencing depth: Minimum 20 million uniquely mapped reads for sufficient coverage
Bioinformatic analysis: Use specialized tools like MACS2 for peak calling, with appropriate input controls
For genome-wide mapping, compare H4R35me1 distribution patterns with other histone marks (H3K4me3, H3K27ac) to understand its relationship to chromatin states and transcriptional activity.
How can quantitative analysis be performed with Mono-methyl-HIST1H4A (R35) Antibody staining?
Quantitative analysis of H4R35me1 immunostaining involves several methodological approaches:
Fluorescence intensity measurement:
Capture images using consistent exposure settings
Define nuclear regions (ROIs) using DAPI counterstain
Measure mean fluorescence intensity within nuclear ROIs
Subtract background from non-nuclear regions
Nuclear pattern analysis:
Classify staining patterns (uniform, speckled, peripheral)
Quantify percentage of cells showing each pattern
Compare pattern distribution between experimental conditions
Co-localization analysis:
Calculate Pearson's or Manders' coefficients for co-localization with other chromatin marks
Use specialized software (ImageJ with JACoP plugin, CellProfiler)
Western blot quantification (for bulk analysis):
Flow cytometry:
Fix and permeabilize cells appropriately
Use consistent antibody dilutions (1:100-1:200)
Gate populations based on cell cycle stages
Compare median fluorescence intensity between samples
How do H4R35 methylation patterns correlate with gene expression data?
The correlation between H4R35me1 and gene expression follows specific patterns:
Genomic distribution analysis: H4R35me1 shows distinct genomic localization compared to other histone marks like H4K31ac and H4K31me1. While H4K31ac is typically enriched at promoters of active genes, methylation marks like H4R35me1 may have different distribution patterns .
Correlation methodology:
Integrate ChIP-seq data for H4R35me1 with RNA-seq expression data
Calculate correlation coefficients between H4R35me1 enrichment at promoters/gene bodies and corresponding transcript levels
Perform gene set enrichment analysis (GSEA) to identify functional pathways associated with H4R35me1-marked genes
Expression correlation patterns: Based on studies of similar modifications:
H4 methylation marks may have context-dependent correlations with gene expression
H4R35me1 might show inverse correlation with gene expression in some contexts, similar to H4K31me1 which is inversely correlated with gene expression in T. gondii
The correlation may differ between promoters and gene bodies
Cell type specificity: The relationship between H4R35me1 and gene expression may vary between cell types, as observed with other histone modifications in studies comparing adult versus neonatal oligodendrocyte progenitors .
What is the interplay between H4R35 methylation and other post-translational modifications on the nucleosome surface?
The nucleosome lateral surface contains several modifiable residues that functionally interact:
Modification crosstalk:
Structural implications:
Reader protein coordination:
Modification hierarchy:
Condensability effects:
How does H4R35 methylation contribute to chromatin organization during cell division?
H4R35 methylation has distinct roles during cell division:
Mitotic enrichment pattern:
Cell cycle dynamics:
Chromosome condensation role:
The enrichment on mitotic chromosomes suggests a role in chromosome condensation
The positively charged methylated arginine may strengthen DNA binding, contributing to chromatin compaction
Mitotic bookmarking potential:
H4R35me1 might serve as an epigenetic bookmark that preserves certain chromatin states through cell division
This could contribute to epigenetic memory mechanisms that maintain cell identity
Cell type specificity:
What are the enzymes responsible for H4R35 methylation and demethylation?
The enzymatic regulation of H4R35 methylation involves specific writers and erasers:
Methyltransferases (writers):
While the specific methyltransferase for H4R35 has not been definitively identified in the search results, protein arginine methyltransferases (PRMTs) are the likely candidates
PRMT1 is known to methylate H4R3 and may potentially target other arginine residues
The enzyme specificity may be influenced by the structural context of R35 within the nucleosome
Demethylases (erasers):
Enzymatic assays:
Regulation of enzymatic activity:
How can H4R35 methylation patterns be manipulated experimentally to study its function?
Experimental manipulation of H4R35 methylation can be achieved through several approaches:
Enzymatic inhibition:
Genetic approaches:
CRISPR/Cas9-mediated mutation of H4R35 to a non-methylatable residue (R35K or R35A)
Knockdown/knockout of the responsible methyltransferase
Overexpression of candidate demethylases
Peptide competition:
Cell-permeable H4R35me1 peptides can be used to compete with readers of this modification
This approach disrupts the function without altering the modification level
In vitro nucleosome reconstitution:
Experimental readouts:
Effects on gene expression can be measured by RNA-seq
Chromatin accessibility changes can be assessed by ATAC-seq
Changes in cell cycle progression can be monitored by flow cytometry
Effects on nuclear architecture can be visualized by super-resolution microscopy
How can specificity issues with Mono-methyl-HIST1H4A (R35) Antibody be resolved?
Ensuring antibody specificity is critical for reliable results:
Common cross-reactivity issues:
Cross-reactivity with other methylated arginines on histones (H3R2, H3R17, H4R3)
Non-specific binding to other histone modifications with similar chemical properties
Specificity validation methods:
Optimization strategies:
Titrate antibody concentration to minimize background (starting with 1:50-1:200 dilution)
Increase blocking stringency (5% BSA or 10% normal serum)
Add competitive inhibitors for common cross-reactive epitopes
Optimize washing conditions (increase wash number, duration, or detergent concentration)
Batch variability management:
Test each new antibody lot against a standard sample
Maintain reference images/data for comparison
Consider pooling multiple antibody lots for long-term studies
Alternative confirmation approaches:
Use multiple antibodies targeting the same modification from different suppliers
Validate with orthogonal techniques (mass spectrometry)
What are the challenges in detecting H4R35 methylation in different sample types?
Different biological samples present unique challenges for H4R35me1 detection:
Tissue sections:
Challenge: Epitope masking due to formaldehyde fixation and paraffin embedding
Solution: Optimize antigen retrieval (citrate buffer pH 6.0, 95-100°C for 20 minutes)
Cell cultures:
Challenge: Variable expression levels across cell cycle
Solution: Synchronize cells or co-stain with cell cycle markers
Mitotic chromosomes:
Challenge: Preserving chromosome structure while ensuring antibody accessibility
Solution: Use chromosome spreading techniques with gentle fixation
Heterogeneous tissues:
Challenge: Cell type-specific variations in H4R35me1
Solution: Co-stain with cell type markers; consider single-cell approaches
ChIP-seq samples:
Challenge: Low abundance of H4R35me1 and potential epitope masking during crosslinking
Solution: Increase starting material; optimize crosslinking conditions
Western blot detection:
How do storage and handling conditions affect Mono-methyl-HIST1H4A (R35) Antibody performance?
Proper handling is essential for maintaining antibody activity:
Storage recommendations:
Stability considerations:
Working dilutions should be prepared fresh
Most antibodies remain stable at 4°C for 1-2 weeks
Monitor for signs of deterioration (precipitation, declining signal)
Buffer composition effects:
Transportation considerations:
Shipping on ice or ice packs for short transits
Dry ice recommended for longer shipping times
Performance monitoring:
Validate each aliquot before critical experiments
Maintain reference samples with known H4R35me1 levels for comparison
Record lot numbers and performance characteristics