Di-Methyl-Histone H3(K79) Monoclonal Antibody is a highly specific immunological reagent designed to detect di-methylation at lysine residue 79 (K79) of histone H3, a core nucleosomal protein. This modification is dynamically linked to chromatin structure regulation and transcriptional activation . Histone H3-K79 methylation is catalyzed by the methyltransferase DOT1L and is influenced by trans-tail interactions, such as H2B ubiquitination, which enhances DOT1L activity . Unlike other histone methylations, H3K79 methylation occurs on the globular domain and is associated with actively transcribed genes .
The antibody exhibits high specificity for di-methylation, as demonstrated by peptide array assays where it binds strongly to H3K79me2 but not H3K79me1 or H3K79me3 .
Transcriptional Activation: H3K79me2 is enriched at actively transcribed gene bodies and is associated with elongating RNA polymerase II .
Regulation by Trans-Tail Interactions:
Replication-Dependent Dynamics: Levels fluctuate during the cell cycle, suggesting turnover via histone exchange rather than active demethylation .
While H3K79me2 is not directly linked to disease in current literature, dysregulation of DOT1L or H2B ubiquitination pathways is implicated in cancers (e.g., leukemia) . Antibodies targeting H3K79me2 serve as critical tools for studying these pathways.
H3K79 dimethylation occurs on lysine 79 of histone H3, which is located within the nucleosome core. Nucleosomes wrap and compact DNA into chromatin, thereby limiting DNA accessibility to cellular machineries that require DNA as a template. Histone H3 plays a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability . Specifically, methylation at lysine 79 acts as a marker of inactive chromatin regions that is critical for transcriptional silencing. Silencing proteins such as Sir3 function by blocking Dot1 methylation at this site . This modification is part of the complex "histone code" that regulates gene expression through various post-translational modifications.
H3K79 dimethylation is catalyzed by DOT1L methyltransferase (Disruptor of Telomeric silencing 1-like), which is unique among histone methyltransferases in that it does not contain a SET domain. Prior research has demonstrated that DOT1L exhibits exceptional selectivity for H3K79, with almost no other histone or non-histone substrates being identified . The enzyme can add one, two, or three methyl groups to lysine 79, producing mono-, di-, or trimethylated states. The methylation process involves the transfer of methyl groups from S-adenosyl methionine (SAM) to the target lysine. Unlike many other histone modifications, H3K79 methylation has not been definitively linked to specific demethylase enzymes, although histone demethylases including PADI4, LSD1, JMJD1, JMJD2, and JHDM1 have shown that methylation generally is a reversible epigenetic marker .
Di-methyl-Histone H3(K79) Monoclonal Antibodies are versatile research tools with applications in multiple techniques:
Chromatin Immunoprecipitation (ChIP): For mapping genomic locations of H3K79me2 marks
Western Blotting (WB): For detecting and quantifying H3K79me2 levels
Peptide Arrays (PepArr): For antibody specificity testing and epitope mapping
Immunocytochemistry/Immunofluorescence (ICC/IF): For visualizing nuclear distribution patterns
Immunohistochemistry - Paraffin (IHC-P): For tissue section analysis
Flow Cytometry: For quantitative analysis at the cellular level
Chromatin IP-seq: For genome-wide profiling of H3K79me2 marks
CUT&Tag/CUT&RUN Assays: For high-resolution mapping of protein-DNA interactions
The selected antibody format (monoclonal vs. polyclonal) and host species should be determined based on the specific experimental requirements and anticipated cross-reactivity concerns.
When designing ChIP experiments with Di-methyl-Histone H3(K79) Monoclonal Antibodies, researchers should consider the following methodological approach:
Sample Preparation: Use fresh chromatin preparations with optimal crosslinking (typically 1% formaldehyde for 10 minutes). Over-crosslinking can mask the H3K79me2 epitope.
Sonication Parameters: Aim for chromatin fragments between 200-500bp. Test sonication efficiency with a small aliquot before proceeding.
Antibody Selection: Choose ChIP-grade antibodies specifically validated for this application, such as rabbit recombinant monoclonal antibodies against H3K79me2 .
Controls: Include:
Positive control: Antibody against total Histone H3
Negative control: IgG from same species as the primary antibody
Input chromatin: To normalize enrichment
Immunoprecipitation: Use 2-5μg of antibody per reaction with chromatin from approximately 1 million cells. Overnight incubation at 4°C on a rotating platform is recommended.
Washing and Elution: Perform stringent washes to minimize background, followed by elution under conditions that do not denature the antibody.
Validation: Confirm enrichment at known H3K79me2-marked regions using qPCR before proceeding to genome-wide analysis.
For downstream applications like ChIP-seq, ensure library preparation preserves the representative nature of immunoprecipitated DNA fragments.
For optimal Western blotting results with Di-methyl-Histone H3(K79) Monoclonal Antibodies, follow these methodological guidelines:
Sample Preparation:
Extract histones using acid extraction (0.2N HCl or 0.4N H₂SO₄) followed by TCA precipitation
Load 5-15μg of histone extract per well
Gel Electrophoresis:
Use 15-18% SDS-PAGE gels to resolve the relatively small histone proteins (~17 kDa)
Include recombinant methylated H3 peptides as positive controls
Transfer Conditions:
Transfer to PVDF membrane (preferred over nitrocellulose for histones)
Use wet transfer at 30V overnight at 4°C for efficient transfer of basic proteins
Blocking:
Block with 5% BSA in TBST rather than milk (milk contains casein which is phosphorylated and can increase background)
Block for 1 hour at room temperature
Antibody Incubation:
Detection:
Use enhanced chemiluminescence detection systems
Expected molecular weight for H3 is approximately 17 kDa
Controls and Normalization:
Run parallel blots with antibodies against total H3 for normalization
Consider dual-color detection systems to simultaneously visualize total H3 and H3K79me2
Validating antibody specificity is critical for reliable experimental outcomes. For Di-methyl-Histone H3(K79) Monoclonal Antibodies, implement these methodological approaches:
Peptide Competition Assays:
Pre-incubate the antibody with excess H3K79me2 peptide
Compare results with antibody-only controls
Signal should be drastically reduced in the presence of competing peptide
Peptide Array Analysis:
DOT1L Knockout/Knockdown:
Analyze extracts from DOT1L-deficient cells
H3K79me2 signal should be significantly reduced or absent
Recombinant Histone Testing:
Use recombinant histones with defined modifications as standards
Compare signals between unmodified H3 and H3K79me2
Mass Spectrometry Validation:
Perform immunoprecipitation followed by mass spectrometry
Confirm enrichment of H3K79me2 peptides in the pulldown
Sequential ChIP:
Perform ChIP with H3K79me2 antibody followed by a second ChIP with another H3-specific antibody
High overlap indicates specificity for H3
This comprehensive validation approach ensures that experimental results truly reflect H3K79me2 biology rather than antibody artifacts.
Advanced research using stable isotope labeling provides a powerful approach to track DOT1L methyltransferase activity with high specificity. The methodology involves:
Synthesis of Heavy-Labeled Co-factors:
Produce 13CD3-BrSAM (bromo-deaza-S-adenosyl-L-methionine with heavy-labeled methyl groups)
Generate 13CD3-SAM as a control co-factor
In Vitro Methylation Reactions:
Incubate nucleosomes with DOT1L enzyme and heavy-labeled co-factors
Allow methylation reactions to proceed under physiological conditions
Mass Spectrometry Analysis:
Digest histones with trypsin or other proteases
Identify peptides containing H3K79
Detect mass shifts corresponding to incorporation of heavy-labeled methyl groups
Specificity Validation:
Research has demonstrated excellent incorporation of heavy labels at H3K79 using both co-factors with DOT1L, while no heavy labeling was observed at other histone sites (H3K4, H3K9, H3K27, H3K36, H4K20), confirming DOT1L's selectivity. Importantly, when other methyltransferases (SETD7, G9a, SETD2, SUB420H2) were used with 13CD3-BrSAM, no incorporation was observed at their respective target sites, showing 13CD3-BrSAM's excellent selectivity for DOT1L .
This methodology provides a powerful approach to track DOT1L activity in various experimental contexts, enabling the study of factors affecting enzyme activity and substrate specificity.
Understanding the complex interrelationships between H3K79 dimethylation and other histone modifications requires sophisticated methodological approaches:
Sequential ChIP (Re-ChIP):
Perform initial ChIP with H3K79me2 antibody
Use the enriched material for a second ChIP with antibodies against other modifications
Quantify regions carrying both modifications
Compare to single ChIP datasets to determine co-occurrence patterns
Mass Spectrometry-Based Combinatorial Analysis:
Isolate intact histone proteins or nucleosomes
Employ middle-down or top-down MS approaches that preserve modification combinations
Quantify co-occurrence frequencies of H3K79me2 with other modifications on the same histone tail
Multicolor ChIP-Seq:
Use antibody cocktails with different epitope tags
Develop multiplexed sequencing libraries
Bioinformatically resolve modification patterns
Targeted Perturbation Experiments:
Manipulate specific histone-modifying enzymes (e.g., DOT1L inhibition)
Measure consequent changes in other modifications
Establish causal relationships and sequence of modification events
Proximity Ligation Assays (PLA):
Detect closely positioned histone modifications in intact nuclei
Quantify spatial relationships between H3K79me2 and other modifications
Current research indicates that H3K79 methylation coordinates with other modifications in complex ways. Lysine methylation occurs primarily on histones H3 (Lys4, 9, 27, 36, 79) and H4 (Lys20) and has been implicated in both transcriptional activation and silencing . These modifications work together to coordinate the recruitment of chromatin modifying enzymes containing methyl-lysine binding modules such as chromodomains (HP1, PRC1), PHD fingers (BPTF, ING2), tudor domains (53BP1), and WD-40 domains (WDR5) .
Investigating the functional impacts of altered H3K79 dimethylation patterns requires a multifaceted experimental approach:
DOT1L Inhibition or Genetic Manipulation:
Employ small molecule DOT1L inhibitors at various concentrations
Use genetic approaches (CRISPR/Cas9, RNAi) to modulate DOT1L expression
Create point mutations in H3K79 (K79A, K79R) to prevent methylation
Transcriptome Analysis:
Perform RNA-seq following DOT1L perturbation
Compare expression changes with H3K79me2 ChIP-seq data
Identify direct transcriptional targets versus secondary effects
Use time-course experiments to track dynamic responses
Chromatin Accessibility Assays:
Implement ATAC-seq or DNase-seq to measure chromatin accessibility changes
Correlate accessibility with H3K79me2 distribution
Map nucleosome positioning relative to H3K79me2 marks
Functional Genomics Screens:
Conduct CRISPR screens in DOT1L-inhibited cells
Identify synthetic lethal or rescue interactions
Map genetic networks associated with H3K79me2 function
Proteomic Approaches:
Identify readers of H3K79me2 using SILAC-based pull-downs
Compare protein interactomes in normal versus DOT1L-inhibited conditions
Map signaling pathways affected by H3K79me2 levels
Single-Cell Analysis:
Measure cell-to-cell variation in H3K79me2 levels
Correlate with transcriptional heterogeneity
Track cellular differentiation trajectories
By integrating these approaches, researchers can establish causal relationships between H3K79 dimethylation patterns and specific biological processes, moving beyond correlative observations to mechanistic understanding.
ChIP experiments with Di-methyl-Histone H3(K79) Monoclonal Antibodies can present several technical challenges. Here are common issues and methodological solutions:
Low Signal-to-Noise Ratio:
Problem: High background or weak specific signal.
Solutions:
Epitope Masking:
Problem: Overfixation can mask the H3K79me2 epitope.
Solutions:
Optimize formaldehyde concentration (typically 0.75-1%)
Limit crosslinking time (8-10 minutes is often optimal)
Implement epitope retrieval steps if necessary
Consider native ChIP for some applications
Cross-Reactivity:
Inefficient Chromatin Fragmentation:
Problem: Suboptimal sonication leading to inconsistent results.
Solutions:
Optimize sonication parameters (time, amplitude, pulse duration)
Verify fragment size distribution by gel electrophoresis
Consider enzymatic fragmentation alternatives
Ensure consistent cell numbers across experiments
PCR Bias in ChIP-seq:
Problem: Amplification artifacts during library preparation.
Solutions:
Minimize PCR cycles
Use unique molecular identifiers (UMIs)
Implement multiple library replicates
Apply computational corrections for GC content
Addressing these challenges through methodical optimization will enhance the reliability and reproducibility of ChIP experiments targeting H3K79 dimethylation.
Antibody batch variation can significantly impact experimental reproducibility. Here's a methodological approach to address this challenge:
Proactive Quality Control:
Validation Protocol: Establish a standardized testing procedure for each new antibody batch:
Peptide array analysis against modified and unmodified peptides
Western blot using positive control samples (e.g., cell lines with known H3K79me2 levels)
ChIP-qPCR for known target regions with established enrichment values
Internal Reference Standards:
Create and maintain reference samples:
Aliquot and freeze histone extracts from standard cell lines
Generate recombinant or synthetic H3K79me2 standards
Use these standards when testing new antibody batches
Collaborative Batch Testing:
Quantitative Benchmarking:
Develop quantitative metrics for antibody performance:
Signal-to-noise ratio in Western blots
Enrichment values for control regions in ChIP
Cross-reactivity percentages from peptide arrays
Compare these metrics across batches
Mitigation Strategies:
By implementing these methodological approaches, researchers can minimize the impact of batch variation and enhance the reproducibility of their epigenetic studies.
Detecting low-abundance H3K79 dimethylation presents methodological challenges, particularly in limiting or difficult sample types. Consider these approaches:
Sample Enrichment Techniques:
FACS Sorting: Isolate specific cell populations of interest
Laser Capture Microdissection: For tissue-specific analysis
Nuclear Isolation: Remove cytoplasmic contaminants
Sequential Extraction: Employ salt fractionation to enrich chromatin-bound histones
Signal Amplification Methods:
Tyramide Signal Amplification (TSA): For immunofluorescence detection
Proximity Ligation Assay (PLA): Detect protein-protein interactions or modifications
3D-SIM Super-Resolution Microscopy: For spatial resolution beyond diffraction limit
ChIP-qPCR with Pre-amplification: For very low input samples
Enhanced Detection Chemistry:
Highly-Sensitive ECL Substrates: For Western blotting detection
Fluorescent Antibody Conjugates: Direct labeling to reduce background
Quantum Dots: For multiplexed detection with broader dynamic range
Mass Cytometry: For single-cell analysis of multiple modifications
Optimized Experimental Protocols:
Carrier ChIP: Add carrier chromatin/DNA to very low input samples
Micro-ChIP: Modified protocols for small cell numbers (<10,000)
Modified Histone Extraction: Gentler methods to preserve modifications
Low-Input Library Preparation: For ChIP-seq from limited material
Computational Enhancement:
Deconvolution Algorithms: To resolve mixed cell populations
Pattern Recognition: To identify characteristic H3K79me2 distributions
Integrative Analysis: Combine multiple data types to enhance confidence
Imputation Methods: For sparsely covered genomic regions
By combining these methodological approaches, researchers can enhance the detection of low-abundance H3K79 dimethylation in challenging biological samples, expanding the range of experimental contexts in which this important epigenetic mark can be studied.
H3K79 dimethylation has emerged as a critical epigenetic mark with significant implications for disease mechanisms. Current research directions include:
Cancer Biology:
DOT1L overexpression and aberrant H3K79 dimethylation patterns have been implicated in several cancers
MLL-fusion leukemias show particular dependence on DOT1L activity
Research is ongoing to understand how H3K79me2 distribution contributes to oncogenic gene expression programs
DOT1L inhibitors are being developed as potential therapeutic agents for specific leukemia subtypes
Neurodegenerative Disorders:
Altered H3K79 methylation has been observed in models of neurodegenerative diseases
Research is exploring how these changes affect neuronal gene expression and function
Studies are investigating potential connections between H3K79me2 and DNA repair deficiencies in neurodegeneration
Cardiovascular Disease:
Emerging evidence suggests roles for H3K79 methylation in cardiac development and disease
Research is examining how H3K79me2 patterns influence cardiac gene expression programs
Studies are investigating potential therapeutic modulation of this mark in heart failure models
Methodological Approaches:
Patient-derived samples are being analyzed for H3K79me2 patterns as potential biomarkers
Single-cell technologies are being applied to understand heterogeneity in H3K79me2 distribution
CRISPR screens are identifying synthetic lethal interactions with DOT1L inhibition
Chemical biology approaches are developing selective DOT1L inhibitors with improved pharmacological properties
Therapeutic Development:
Small molecule inhibitors targeting DOT1L are in preclinical and early clinical development
Precision approaches aim to selectively target cells with aberrant H3K79me2 patterns
Combination therapies with other epigenetic modulators are being investigated for synergistic effects
These research directions highlight the growing recognition of H3K79 dimethylation as both a disease mechanism and therapeutic target across multiple pathological contexts.
Genome-wide mapping of H3K79 dimethylation has benefited from significant methodological advances, enhancing resolution, sensitivity, and biological insight:
Single-Cell ChIP-Seq Technologies:
New protocols allow H3K79me2 profiling at single-cell resolution
Droplet-based and microfluidic approaches enable high-throughput processing
Computational methods address sparse data challenges inherent to single-cell approaches
These advances reveal cell-to-cell variation in H3K79me2 patterns within tissues
CUT&RUN and CUT&Tag Adaptations:
Long-Read Sequencing Applications:
Oxford Nanopore and PacBio technologies are being adapted for epigenetic analysis
These approaches allow simultaneous detection of multiple modifications on the same DNA molecule
Computational methods are improving for base modification detection on long reads
Integration with chromatin conformation data provides 3D context for H3K79me2 distribution
Direct Detection Methods:
SMRT sequencing and nanopore approaches are being developed for direct detection of modifications
These methods eliminate potential biases introduced by antibody-based enrichment
Emerging technologies may enable real-time tracking of dynamic changes in H3K79me2
Multi-Modal Data Integration:
Simultaneous profiling of H3K79me2, transcription, and chromatin accessibility
Spatial transcriptomics integration to map modification patterns in tissue context
Machine learning approaches to predict functional consequences of H3K79me2 distribution
Network analysis to identify regulatory hubs associated with H3K79me2 marks
These methodological advances are transforming our understanding of H3K79 dimethylation from static snapshots to dynamic, three-dimensional, and functionally integrated views across diverse biological contexts.
Advanced computational methods are increasingly essential for extracting maximum biological insight from H3K79me2 datasets. Current approaches include:
Peak Calling Optimizations:
Specialized algorithms for H3K79me2's distinctive distribution patterns
Machine learning approaches that incorporate chromatin state information
Methods that account for regional biases and sequence composition effects
Bayesian frameworks that integrate prior knowledge about H3K79me2 distribution
Integrative Analysis Frameworks:
Multi-omics data integration platforms combining H3K79me2 with:
Transcriptomic data (RNA-seq, nascent RNA)
Chromatin accessibility (ATAC-seq, DNase-seq)
Other histone modifications
Transcription factor binding
Network-based approaches to identify regulatory circuits influenced by H3K79me2
Causal inference methods to distinguish drivers from passengers in gene regulation
Evolutionary and Comparative Analyses:
Cross-species comparison of H3K79me2 conservation patterns
Identification of species-specific versus conserved regulatory roles
Evolutionary constraint analysis at H3K79me2-marked regions
Phylogenetic approaches to trace the evolution of DOT1L-regulated pathways
Deep Learning Applications:
Convolutional neural networks for pattern recognition in H3K79me2 data
Attention-based models to capture long-range dependencies
Transfer learning approaches using pre-trained models from related epigenetic marks
Generative models to predict the effects of perturbations on H3K79me2 patterns
Single-Cell Computational Methods:
Deconvolution algorithms for bulk H3K79me2 data
Imputation strategies for sparse single-cell data
Trajectory inference to map dynamic changes in H3K79me2 during cellular processes
Integration with single-cell transcriptomics and other modalities
These computational approaches transform raw H3K79me2 data into mechanistic insights about gene regulation, development, and disease, creating opportunities for hypothesis generation and experimental design that would not be possible with conventional analysis methods.