The Mono-methyl-Histone H3.1 (R2) Recombinant Monoclonal Antibody is a specialized tool for detecting mono-methylation at arginine 2 (R2) of histone H3.1, a post-translational modification critical to chromatin structure and gene regulation. This antibody is engineered to target a specific epigenetic marker, enabling researchers to study histone modifications in diverse biological contexts, including transcriptional control, DNA repair, and cellular differentiation.
The antibody recognizes the mono-methylated form of arginine 2 (R2) on histone H3.1, encoded by the HIST1H3A gene. Histone H3.1 is a core histone variant integral to nucleosome assembly, and its methylation at R2 is associated with active chromatin regions and transcriptional regulation .
Host Species: Rabbit or HEK293F cells (recombinant expression) .
Immunogen: Synthetic peptides corresponding to the R2-methylated region of histone H3.1 .
The antibody demonstrates high specificity for H3.1 R2me1, with no cross-reactivity to unmethylated or other methylated forms (e.g., R2me2 or R2me3) . It can detect all H3.1 variants bearing R2me1, regardless of other histone modifications .
This antibody is validated for multiple techniques:
The mono-methyl-histone H3.1 (R2) recombinant monoclonal antibody is produced through a robust cloning and expression process. Genes encoding for the HIST1H3A antibody are cloned and expressed in mammalian cell expression systems. The heavy and light chains of the HIST1H3A antibody are individually cloned into expression vectors, which are subsequently introduced into host cells via transfection. These host cells then proceed to produce and secrete the antibodies, which are further purified through affinity chromatography. The resulting antibody has been rigorously tested for functionality in ELISA, WB, ICC, and IF applications, demonstrating its efficacy in detecting human HIST1H3A protein mono-methylated at R2.
Mono-methylation of HIST1H3A at arginine 2 (R2) is a significant epigenetic modification that plays a crucial role in influencing gene expression and chromatin structure. Notably, this modification promotes gene activation and accessibility. Its dynamic regulation is essential for maintaining cellular identity and responding to environmental cues, with potential implications in the development of various diseases.
Histone H3.1 is a core component of nucleosomes. Nucleosomes function to wrap and compact DNA into chromatin, effectively limiting DNA accessibility to cellular machinery that requires DNA as a template. As a result, histones play a central role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is regulated through a complex interplay of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Mono-methyl-Histone H3.1 (R2) is a post-translational modification occurring on arginine 2 of the replication-dependent histone variant H3.1. Unlike H3.3 methylation, H3.1 methylation is primarily associated with silent genomic regions and heterochromatin formation. The key distinction between H3.1 and H3.3 variants lies at position 31, with alanine in H3.1 and threonine in H3.3, which affects methylation potential by histone-modifying enzymes . This specific methylation serves distinct epigenetic functions compared to other histone marks, participating in transcriptional regulation, DNA replication timing, and chromatin structure maintenance.
The specificity of these antibodies is determined through rigorous validation protocols that assess cross-reactivity against unmethylated H3.1, di/tri-methylated H3.1(R2), methylated H3.3, and other histone variants. Commercial antibodies undergo validation via multiple approaches:
ELISA using peptide arrays containing various methylated and unmethylated histone variants
Western blot analysis comparing reactivity against purified histones
Immunofluorescence in cells with knockdown/knockout of methyltransferases
ChIP-seq correlation with known genomic distribution patterns
Researchers should select antibodies validated specifically for their intended applications (ELISA, WB, ICC, IF) as validation stringency may vary by application .
For Western blot applications with Mono-methyl-Histone H3.1 (R2) antibodies, the following protocol has been optimized based on experimental validation:
Parameter | Recommended Conditions | Notes |
---|---|---|
Dilution Range | 1:500 - 1:2000 | Optimize for specific antibody lot |
Blocking Solution | 5% BSA in TBST | Milk can interfere with some epitope recognition |
Sample Preparation | Acid extraction of histones | Improves detection compared to whole-cell lysates |
Loading Amount | 5-15 μg protein | Dependent on cell type and methylation abundance |
Transfer Conditions | Low MW protein settings | Typically 100V for 60-90 minutes |
Membrane | PVDF preferred | Nitrocellulose also acceptable |
Primary Antibody Incubation | Overnight at 4°C | For optimal signal-to-noise ratio |
When interpreting results, it's crucial to include appropriate controls, such as purified recombinant H3.1 with and without methylation, and consider the impact of neighboring histone modifications on epitope recognition .
While standard ChIP protocols can be adapted for Mono-methyl-Histone H3.1 (R2) antibodies, several optimization steps are critical for success:
Chromatin preparation: Use 1% formaldehyde for 10 minutes at room temperature for optimal crosslinking, as overfixation can mask the R2 epitope
Sonication parameters: Generate fragments of 200-500 bp for optimal resolution of H3.1-specific regions
Antibody amount: Use 2-5 μg of antibody per ChIP reaction (adjust based on antibody quality)
Washing stringency: Include high-salt washes (500 mM NaCl) to reduce background
Elution conditions: Standard elution with SDS is effective for most applications
For ChIP-seq applications, it's essential to sequence sufficient depth (>20 million uniquely mapped reads) to capture the predominantly heterochromatic distribution of H3.1 methylation. Bioinformatic analysis should account for repetitive elements where H3.1 is enriched .
For successful immunofluorescence using Mono-methyl-Histone H3.1 (R2) antibodies:
Fixation method: Paraformaldehyde (4%) for 10-15 minutes preserves nuclear architecture while maintaining epitope accessibility
Permeabilization: 0.2% Triton X-100 for 10 minutes is generally effective; avoid methanol which can extract histones
Antibody dilution: Start with 1:50-1:200 dilution range and optimize
Blocking: Use 3-5% BSA with 0.1% Tween-20 to reduce background
Incubation time: Overnight at 4°C typically yields superior signal-to-noise ratios
Epitope retrieval: May be necessary in formaldehyde-fixed tissues (10mM citrate buffer, pH 6.0)
Counterstaining: DAPI for DNA visualization; consider co-staining with markers for specific nuclear compartments
The nuclear distribution pattern should show punctate staining corresponding to heterochromatic regions, which can be confirmed by co-localization with H3K9me3 or HP1 markers .
Genome-wide analyses have revealed distinct patterns of Mono-methyl-Histone H3.1 (R2) distribution that correlate with specific chromatin states:
Enrichment in silent genomic regions: H3.1 is preferentially found in transcriptionally inactive regions, showing inverse correlation with RNA Polymerase II occupancy
Association with repressive marks: Strong co-localization with H3K27me3, H3K9me2/3, and DNA methylation in all sequence contexts (CG, CHG, and CHH)
Replication timing correlation: Enrichment at defined origins of replication, suggesting functional roles in DNA replication regulation
Transposable element association: Significant enrichment over transposable elements, consistent with heterochromatic localization
When examining the relationship between H3.1 methylation and gene expression, RNA-seq data demonstrates an anticorrelation between H3.1 enrichment and transcriptional activity. Approximately 20,097 genomic regions show significant H3.1 enrichment, with these regions displaying elevated DNA methylation levels compared to genome-wide averages .
Several key hypotheses have emerged regarding the functional roles of Mono-methyl-Histone H3.1 (R2):
Inheritance of heterochromatic states: The replication-coupled incorporation of H3.1 may provide a mechanism for propagating silenced chromatin states through cell division
Transcriptional repression: Mono-methylation at R2 potentially interferes with activating modifications or recruits specific repressive complexes
Regulation of DNA replication: Enrichment at origins of replication suggests roles in controlling replication timing or origin firing
Protection against inappropriate recombination: Heterochromatic marks including H3.1 methylation maintain genome stability by preventing recombination between repetitive elements
Cell identity maintenance: Stable patterns of H3.1 methylation may contribute to cell-type-specific gene expression programs
Recent research indicates that specialized histone-modifying enzymes selectively target H3.1 but not H3.3, suggesting evolved mechanisms to distinguish between replication-dependent and independent histone variants .
Several technical challenges can compromise experiments using Mono-methyl-Histone H3.1 (R2) antibodies:
Cross-reactivity with other methylated arginines: Validate that antibodies distinguish between methylation at R2, R8, R17, and R26 positions on histone H3
Failure to discriminate methylation states: Ensure antibodies specifically recognize mono-methylation rather than di- or tri-methylation
H3 variant specificity concerns: Confirm specificity for H3.1 versus H3.3, particularly if the antibody epitope spans regions containing variant-specific residues
Lot-to-lot variability: Perform validation for each new antibody lot, especially for quantitative applications
Application-specific optimization: An antibody validated for Western blot may not perform optimally in ChIP applications
To avoid these pitfalls, researchers should:
Request validation data specific to their application of interest
Perform peptide competition assays with methylated and unmethylated peptides
Include appropriate controls (knockdown/knockout of relevant methyltransferases)
Consider using orthogonal techniques to validate key findings
When encountering signal problems with Mono-methyl-Histone H3.1 (R2) antibodies, consider this structured troubleshooting approach:
Problem | Potential Causes | Solutions |
---|---|---|
Weak signal | Insufficient antigen | Increase protein loading; use histone extraction protocols |
Epitope masking | Try alternative fixation methods; consider epitope retrieval | |
Low antibody concentration | Optimize antibody dilution; increase incubation time | |
Degraded antibody | Ensure proper storage; avoid freeze-thaw cycles | |
High background | Insufficient blocking | Increase blocking time/concentration; try alternative blocking agents |
Cross-reactivity | Use more stringent washing; pre-absorb antibody | |
Secondary antibody issues | Include secondary-only controls; try different secondary | |
Multiple bands in WB | Cross-reactivity | Confirm with peptide competition; try more specific antibody |
Histone modifications | Consider the impact of neighboring modifications on epitope | |
Degradation products | Use fresh samples; add protease inhibitors |
For particularly challenging samples, consider enriching for histone fraction using acid extraction (0.2N HCl) before immunoblotting, which can dramatically improve signal-to-noise ratio .
Emerging single-cell technologies are being adapted for Mono-methyl-Histone H3.1 (R2) detection:
Single-cell CUT&Tag/CUT&RUN: These antibody-directed genomic mapping techniques can be applied at single-cell resolution to profile H3.1 methylation patterns across heterogeneous populations
Mass cytometry (CyTOF): Using metal-conjugated antibodies allows simultaneous detection of multiple histone modifications including H3.1-R2me1 in thousands of individual cells
Single-cell imaging techniques: Advanced microscopy approaches like structured illumination microscopy (SIM) and stochastic optical reconstruction microscopy (STORM) enable visualization of H3.1 methylation distribution within nuclear subcompartments
Implementation requires:
Rigorous antibody validation for specificity in single-cell contexts
Appropriate normalization strategies to account for technical variation
Integration with other single-cell modalities (scRNA-seq, scATAC-seq) for multi-omic analyses
Computational approaches to distinguish biologically meaningful patterns from technical noise
Researchers pioneering these approaches should include comprehensive controls and validate findings with orthogonal techniques when possible .
Research into the relationship between Mono-methyl-Histone H3.1 (R2) and disease states is still emerging, but several important connections have been established:
Cancer epigenetics: Altered patterns of H3.1 methylation have been observed in various cancer types, potentially contributing to aberrant gene silencing of tumor suppressors
Epigenetic inheritance: Dysregulation of H3.1 variant incorporation and its associated modifications may affect transgenerational epigenetic inheritance
Developmental disorders: Mutations in enzymes regulating H3.1 methylation have been implicated in developmental abnormalities and intellectual disability syndromes
Cellular senescence: Changes in H3.1 distribution and modification contribute to senescence-associated heterochromatin formation
For researchers investigating these connections, it's crucial to:
Establish baseline H3.1-R2me1 patterns in normal tissues corresponding to disease models
Use multiple antibody-based approaches to confirm findings (ChIP-seq, immunohistochemistry)
Incorporate genetic or pharmacological manipulation of relevant methyltransferases
Consider the interplay between H3.1 methylation and other epigenetic mechanisms in disease progression
The enzymatic regulation of Mono-methyl-Histone H3.1 (R2) involves specialized molecular machinery:
Methyltransferase specificity: Crystal structure studies of histone methyltransferases that target H3.1 reveal a bipartite catalytic domain that specifically "reads" alanine 31 of H3.1, distinguishing it from H3.3 which contains threonine at this position
Structural basis for variant discrimination: Key residues in the methyltransferase (such as E212 and R334 identified in plant ATXR5/6) form a specificity pocket that accommodates the H3.1-specific alanine 31 but generates steric clashes with threonine 31 in H3.3
"Safety belt" conformational mechanism: A loop structure in some methyltransferases folds back on top of H3.1, locking the peptide in a conformation that forces alanine 31 into the specificity pocket
Methyl-reader domains: Specific protein domains have evolved to recognize mono-methylated R2 in the context of H3.1, potentially recruiting additional factors to regulate chromatin structure
Experimental approaches to study these mechanisms include:
Site-directed mutagenesis of key residues in methyltransferases and reader proteins
In vitro histone lysine methyltransferase (HKM) assays comparing activity on H3.1 vs. H3.3 substrates
Structural biology approaches including X-ray crystallography and cryo-EM
Genetic studies manipulating the expression of methyltransferases and evaluating genomic consequences
Integrative approaches yield more comprehensive understanding:
ChIP-seq with other epigenetic marks: Sequential or parallel ChIP-seq for H3.1-R2me1 and other modifications (H3K27me3, H3K9me3, DNA methylation) enables correlation analyses and identification of bivalent domains
CUT&RUN/CUT&Tag alternatives: These techniques offer higher signal-to-noise ratio and require fewer cells than traditional ChIP, particularly valuable for rare cell populations
Hi-C and chromatin conformation: Combining H3.1 methylation data with chromosome conformation capture techniques reveals relationships between this modification and 3D genome organization
ATAC-seq integration: Correlating H3.1 methylation with chromatin accessibility provides insights into functional consequences of this modification
Multi-omics approaches: Integrated analyses with transcriptomics and proteomics datasets help establish causal relationships between H3.1 methylation and gene regulation
Implementation requires careful experimental design, including:
Performing techniques on the same or matched biological samples
Appropriate computational pipelines for multi-omic data integration
Statistical approaches to distinguish correlation from causation
Computational analysis of H3.1-R2me1 ChIP-seq requires specialized approaches:
Peak calling considerations: Standard peak callers may not optimally capture broad domains of H3.1 enrichment; algorithms designed for broad histone marks (SICER, MACS2 with broad peak settings) are recommended
Dealing with repetitive elements: H3.1 is enriched in repetitive regions, requiring careful handling of multi-mapping reads (consider using tools like RepEnrich2)
Differential binding analysis: DESeq2 or diffBind packages can identify regions with altered H3.1 methylation between conditions
Integration with chromatin states: Tools like ChromHMM can incorporate H3.1 methylation data into broader chromatin state models
Visualization approaches: Specialized browsers like WashU Epigenome Browser or UCSC Genome Browser with custom tracks enable visualization of H3.1 patterns in genomic context
A typical analysis pipeline includes:
Quality control (FastQC, multiqc)
Alignment (Bowtie2 with parameters optimized for histone marks)
Peak calling (MACS2 with broad peak settings)
Annotation (HOMER, ChIPseeker)
Motif analysis (MEME-ChIP)
Integration with other datasets (deepTools, bedtools)
For researchers with limited computational experience, several integrated workflows and Galaxy-based tools can simplify this process .