Mono-Methyl-Histone H3(K79) Monoclonal Antibody is a highly specific research tool designed to detect mono-methylation at lysine residue 79 (K79) of histone H3. This modification is catalyzed by the Dot1L methyltransferase and plays critical roles in chromatin structure, transcriptional regulation, and DNA repair . The antibody’s specificity is achieved through immunization with synthetic mono-methylated peptides or recombinant protein fragments, enabling precise recognition of the H3K79me1 epitope without cross-reactivity to di-/tri-methylated states .
Epigenetic Regulation: Used to map H3K79me1 enrichment at gene promoters and enhancers. For example, in liver cancer cells, H3K79me1 levels correlate with transcriptional activation of oncogenic genes .
Viral Pathogenesis: Demonstrated increased H3K79me1 during influenza infection, linked to interferon-mediated antiviral responses .
Detection Sensitivity: Cell Signaling’s #9398 antibody identifies endogenous H3K79me1 in human, mouse, and rat cells .
Subcellular Localization: Confocal microscopy reveals nuclear staining patterns, confirming H3K79me1’s role in chromatin organization .
Specificity Profiling: Abnova’s MAB12838 and Abcam’s ab177183 show high affinity to H3K79me1 peptides, with negligible binding to unmodified or di-/tri-methylated lysine 79 .
Influenza Infection: H3K79me1 levels rise in infected cells, promoting interferon signaling and antiviral gene expression (e.g., IFNβ, ISG56) .
Mechanism: Dot1L inhibition (via EPZ-5676) reduces H3K79me1 and impairs host immunity, increasing viral replication .
HDAC1 Deficiency: Loss of histone deacetylase HDAC1 increases H3K79 methylation (me1/me2/me3), creating a dependency in thymic lymphoma models .
Therapeutic Implications: Combined inhibition of DOT1L (e.g., Pinometostat) and HDACs may synergize to treat MLL-rearranged leukemias .
H3K79 monomethylation serves as a critical epigenetic mark involved in transcriptional regulation. Unlike many histone modifications that occur on N-terminal tails, H3K79 methylation uniquely occurs within the globular domain of histone H3. Experimental evidence shows that H3K79 monomethylation levels vary across histone H3 variants, with H3.3K79 displaying higher levels of monomethylation compared to H3.2 and H3.1 variants . This modification is established by the DOT1L methyltransferase enzyme, which is highly selective for methylating H3K79 . Mechanistically, H3K79 monomethylation alters chromatin structure and influences gene expression patterns through recruitment of specific reader proteins.
H3K79 can exist in unmethylated, mono-, di-, and trimethylated states, each with distinct functional implications. Research has demonstrated that for histone H3.1, the proportions of these methylation states are approximately 88% unmethylated, 11% monomethylated, and 1.5% dimethylated, with trimethylation present at less than 0.1% . Monomethylation of H3K79 represents an initial methylation event that can serve as a substrate for further methylation to di- and trimethylated states. The transition between these states is catalyzed by DOT1L (Disruptor Of Telomeric silencing 1-Like) in mammals. Each methylation state can recruit different protein complexes and may be associated with different transcriptional outcomes.
H3K79 monomethylation can be detected through multiple complementary techniques:
Western blotting: Provides semi-quantitative assessment of global H3K79me1 levels
Chromatin Immunoprecipitation (ChIP): Identifies genomic regions enriched for H3K79me1
Immunofluorescence (IF): Visualizes nuclear distribution patterns of H3K79me1
Immunohistochemistry (IHC): Detects H3K79me1 in tissue sections
Mass spectrometry (MS): Enables precise quantification and distinction between methylation states
For optimal results, antibody specificity for the monomethylated state must be validated against other methylation states of H3K79 . Peptide array testing is recommended to confirm specificity for the monomethylated form versus unmethylated, dimethylated, and trimethylated variants.
Validating the specificity of an H3K79me1 monoclonal antibody requires a multi-step approach:
Peptide array testing: Test antibody against a panel of peptides representing different methylation states of H3K79 (unmethylated, mono-, di-, and trimethylated) at multiple dilutions . The antibody should show strong binding to H3K79me1 peptide with minimal cross-reactivity to other states.
Western blot analysis: Run histone extracts alongside recombinant H3 standards with defined methylation states. Compare band patterns and intensities.
Competition assays: Pre-incubate antibody with excess H3K79me1 peptide before application in your experimental assay. This should abolish specific signal if the antibody is truly specific.
Knockout/knockdown controls: Test antibody in cells where DOT1L has been depleted or inhibited, which should reduce H3K79 methylation signals.
Mass spectrometry correlation: Compare antibody-based detection with MS-based quantification of methylation states.
A high-quality H3K79me1 antibody should demonstrate at least 10-fold higher affinity for the monomethylated form compared to other methylation states of K79 and minimal cross-reactivity with other methylated lysine residues on histones .
| Feature | Monoclonal Anti-H3K79me1 | Polyclonal Anti-H3K79me1 |
|---|---|---|
| Epitope recognition | Single epitope | Multiple epitopes |
| Batch-to-batch consistency | High | Variable |
| Background signal | Lower | Typically higher |
| Specificity | Highly specific, but potential for missing conformational variants | Broader recognition, potential for cross-reactivity |
| Applications | Excellent for quantitative applications | Better for detection in diverse species |
| Production | Hybridoma cell lines | Immunized animals |
| Cost | Higher | Lower |
| Long-term supply | Consistent | May require revalidation between lots |
Chromatin Immunoprecipitation (ChIP) with H3K79me1 antibodies requires careful optimization:
ChIP Protocol Optimization Table for H3K79me1 Antibodies:
| Parameter | Recommended Condition | Notes |
|---|---|---|
| Crosslinking | 1% formaldehyde, 10 min at RT | Shorter times may be preferred as H3K79 is within globular domain |
| Sonication | Aim for 200-500bp fragments | Over-sonication can damage epitopes |
| Antibody amount | 2-5 μg per ChIP reaction | Monoclonal antibodies typically require less |
| Antibody incubation | Overnight at 4°C with rotation | Minimum 4 hours |
| Blocking agent | 1-2% BSA in PBS | Reduces background |
| Washing stringency | Medium-high (350-500 mM NaCl) | Balance between specificity and yield |
| Elution conditions | 1% SDS, 0.1M NaHCO₃, 65°C | Standard elution buffer |
| Controls | IgG control and input (1-5%) | Essential for normalization |
| Validation loci | Known H3K79me1-positive regions | Include inactive gene regions as negative controls |
For optimal results, include spike-in controls for normalization across samples and validate your ChIP-seq peaks with alternative methods such as ChIP-qPCR for selected regions . The antibody's ChIP efficiency can be verified by enrichment at known DOT1L target genes and depleted signals in DOT1L inhibitor-treated cells.
Accurate quantification of H3K79 monomethylation requires complementary approaches:
Mass Spectrometry-Based Quantification: The gold standard for accurate measurement uses stable isotope labeling approaches. This method can distinguish between different methylation states (mono-, di-, tri-) and even differentiate between old and new methylation events on pre-existing versus newly synthesized histones .
Implementation:
Extract histones using acid extraction
Perform propionylation of lysines to block trypsin cleavage at non-methylated lysines
Digest with trypsin or Arg-C protease
Analyze by LC-MS/MS
Quantify using extracted ion chromatograms of specific peptides containing H3K79
Western Blot Quantification:
ChIP-seq for Genome-Wide Distribution:
Perform spike-in normalization with exogenous chromatin
Use consistent sequencing depth
Apply appropriate normalization algorithms
When tracking dynamic changes in H3K79me1 levels, the stable isotope labeling approach provides the most precise measurements, allowing differentiation between methylation on new versus old histones with rates of approximately 1.1-1.2% per hour for new methylation events .
For immunofluorescence experiments with H3K79me1 antibodies, include these essential controls:
Primary antibody controls:
Omission of primary antibody (secondary antibody only)
Isotype control (matched IgG at same concentration)
Blocking peptide competition (pre-incubation with H3K79me1 peptide)
Dilution series to establish optimal antibody concentration
Biological controls:
DOT1L inhibitor-treated cells (should show reduced signal)
DOT1L knockdown/knockout cells
Cell cycle synchronized populations (to account for replication-dependent dynamics)
Technical controls:
Multi-channel controls to check for bleed-through
Counterstaining with DAPI for nuclear localization
Co-staining with general histone H3 antibody for reference
Visualization parameters:
Consistent exposure times between samples
Z-stack imaging to capture the full nuclear volume
Quantification using automated, unbiased image analysis
Document all imaging parameters, including microscope settings, exposure times, and post-acquisition processing steps to ensure reproducibility . For quantitative analysis, use high-content analysis systems (such as Operetta CLS) with maximum intensity projection of confocal sections to accurately assess nuclear distribution patterns.
Tracking H3K79 monomethylation dynamics through the cell cycle requires sophisticated approaches:
Cell Synchronization and Release Protocol:
Synchronize cells at G1/S boundary using double thymidine block or serum starvation
Release into normal medium and collect timepoints (0h, 2h, 4h, 8h, 12h, 24h)
Confirm synchronization by flow cytometry with propidium iodide staining
Stable Isotope Labeling Approach:
Grow cells in media containing heavy arginine (+6 Da) and heavy methyl donors (+4 Da)
Synchronize cells and release into media with normal isotopes
Extract histones at defined timepoints
Analyze by mass spectrometry to distinguish:
Old histones with old methylation
Old histones with new methylation
New histones with new methylation
RITE (Recombination-Induced Tag Exchange) Assay:
Research has shown that H3K79 monomethylation occurs at similar rates on both newly synthesized and pre-existing histone H3, with methylation rates of approximately 1.1-1.2% per hour . This suggests a "scrambling" of methylation positions through the cell cycle rather than strict maintenance of position-specific methylation patterns.
When adapting protocols across species, consider:
Extraction methods: Organism-specific cell lysis conditions (e.g., different buffers for yeast vs. mammalian cells)
Antibody cross-reactivity: Validate antibody recognition across species due to sequence variations
Chromatin structure differences: Adjust sonication/digestion parameters
Available genetic tools: CRISPR in mammals vs. homologous recombination in yeast
Expression systems: Endogenous vs. ectopic expression considerations
Research has demonstrated different distributive mechanisms for Dot1 enzymes across species, despite conservation of the catalytic domain . This necessitates organism-specific optimization of experimental protocols.
Distinguishing between the functional effects of H3K79 mono-, di-, and trimethylation requires sophisticated experimental approaches:
Enzyme Kinetics Modeling:
Specific Reader Protein Identification:
Perform pull-down assays with peptides bearing specific methylation states
Identify differential binding partners using mass spectrometry
Validate interactions with co-immunoprecipitation and ChIP-reChIP
Methylation State-Specific Mutants:
Create DOT1L catalytic mutants that preferentially generate specific methylation states
Express DOT1L mutants in DOT1L-knockout backgrounds
Analyze resulting phenotypes and gene expression patterns
Chemical Biology Approaches:
Research indicates distributive mechanisms for DOT1L, where each methylation step requires a separate enzyme-substrate interaction . This allows for differential regulation of each methylation state and potentially distinct functional outcomes.
Key considerations for successful ChIP with H3K79me1 antibodies:
Epitope accessibility: Since H3K79 is located within the globular domain of histone H3, optimization of crosslinking and sonication is critical. Over-crosslinking can reduce accessibility.
Control for cell cycle effects: H3K79 methylation patterns change throughout the cell cycle, with methylation of new histones occurring at a rate of approximately 1.1-1.2% per hour . Synchronize cells or account for cell cycle distribution in your population.
Antibody validation: Always validate antibody specificity using peptide competition assays and histone peptide arrays to confirm specificity for monomethylated K79 versus other methylation states .
When facing discrepancies between different methods for detecting H3K79 monomethylation, consider this systematic approach:
Understand method-specific biases:
Western blot: Affected by antibody specificity, extraction efficiency
ChIP: Influenced by chromatin accessibility, crosslinking efficiency
Mass spectrometry: May be affected by ionization efficiency, extraction bias
Reconciliation strategy:
Method-specific optimization:
For Western blot: Test multiple antibody clones; optimize extraction protocol
For ChIP: Adjust crosslinking conditions; test different sonication parameters
For MS: Ensure complete digestion; optimize chromatography conditions
Interpretation framework:
Research has demonstrated that pre-existing "old" histones continue to be K79-monomethylated at a rate equal to newly synthesized histones . This dynamic nature may contribute to discrepancies between methods if temporal factors are not controlled.
Distinguishing between new methylation and methylation turnover on H3K79 requires sophisticated experimental designs:
Dual Isotopic Labeling Strategy:
Label both histones (using heavy arginine, +6 Da) and methyl donors (using heavy SAM, +4 Da)
Synchronize cells and release into media with normal isotopes
Extract histones at defined timepoints
Analyze by high-resolution mass spectrometry to distinguish:
Old histones with old methylation (heavy histone, heavy methyl)
Old histones with new methylation (heavy histone, light methyl)
New histones with new methylation (light histone, light methyl)
Decay Rate Analysis:
Compare decay rates of unmodified and modified peptides
If turnover (demethylation + remethylation) is significant, the old histones with old methylation should decay faster than unmodified control peptides
Research has shown similar decay rates (approximately 1.1% per hour) for both modified and unmodified peptides, suggesting limited turnover
DOT1L Inhibitor Pulse-Chase:
Treat cells with reversible DOT1L inhibitors
Wash out inhibitor and monitor re-establishment of methylation
Compare kinetics on old versus new histones
RITE Assay Combined with MS:
Recent methodological advances have significantly enhanced our ability to study H3K79 methylation dynamics:
Advanced Mass Spectrometry Approaches:
Improved Genetic Tools:
Mathematical Modeling Approaches:
Single-Cell Technologies:
CUT&Tag for single-cell profiling of H3K79 methylation
Integration with single-cell transcriptomics
Live-cell imaging of methylation dynamics using engineered readers
Multiplexed detection systems offer powerful advantages for comprehensive H3K79 methylation analysis:
Co-Detection of Multiple Methylation States:
Simultaneous detection of H3K79me0/me1/me2/me3 using antibody panels
Sequential immunoprecipitation (Re-ChIP) to identify bivalent domains
Mass cytometry (CyTOF) for single-cell quantification of multiple histone marks
Integration with Other Histone Modifications:
Co-ChIP for H3K79me1 with H3K4me3, H3K27ac, or other marks
Correlation analysis between different modification states
Sequential ChIP to identify co-occurrence patterns
Multi-Omics Approaches:
Integration of ChIP-seq with RNA-seq to correlate H3K79me1 with gene expression
ATAC-seq combination to assess chromatin accessibility
Proteomics identification of readers specific to each methylation state
Technical Implementation:
Barcode-based multiplexing for high-throughput ChIP-seq
Multi-channel immunofluorescence imaging
Sequential antibody staining and elution protocols
These multiplexed approaches have revealed that H3K79 methylation patterns are not strictly maintained at the same genomic positions after DNA replication, suggesting "scrambling" of methylation positions . This finding challenges previous models of epigenetic inheritance and highlights the dynamic nature of this modification.
When investigating H3K79 methylation in disease contexts, consider these critical methodological aspects:
Sample Preparation Considerations:
Fresh vs. frozen vs. fixed tissues (FFPE requires modified extraction protocols)
Matched normal-disease sample pairs for comparative analysis
Cell type heterogeneity in tissue samples (consider single-cell approaches)
Patient treatment history that may affect methylation (e.g., prior chemotherapy)
Disease-Specific Controls:
Include appropriate disease and normal controls
Consider disease progression stages
Account for confounding factors (age, sex, treatment status)
Quantification Approaches:
Absolute quantification with isotope-labeled internal standards
Relative quantification normalized to total H3
Locus-specific vs. global methylation changes
Validation Strategies:
Cross-validation with multiple antibody clones
Orthogonal methods (ChIP-seq, MS, IF)
Functional validation with DOT1L inhibitors
Data Analysis Frameworks:
Correlation with clinical parameters
Integration with other epigenetic marks
Machine learning approaches for biomarker identification
These methodological considerations are particularly important as DOT1L-mediated H3K79 methylation has been implicated in various diseases, including leukemias with MLL rearrangements, making it a therapeutically interesting target for pharmaceutical intervention .