Detects a single band at ~15–18 kDa in human cell lines (HeLa, HEK-293T, THP-1) .
Specificity confirmed via peptide competition assays (Figure 1A in ).
Cross-reactivity observed with H3K36me1 and H3K36me3 in some contexts, necessitating validation with modification-specific controls .
Localizes to euchromatic regions in HeLa cells, consistent with H3K36me2’s role in transcribed genes .
Validated in paraformaldehyde- and methanol-fixed cells with Triton X-100 permeabilization .
Compatible with ChIP protocols in human osteosarcoma (U-2 OS) cells, showing enrichment at active promoters (e.g., GAPDH) over inactive loci (e.g., MYO-D) .
Specificity Concerns: Some H3K36me antibodies exhibit cross-reactivity with adjacent PTMs. For example, H3K14ac antibodies may erroneously bind H3K36ac due to sequence similarity .
Functional Insights:
HIST1H3A is a core histone protein and a member of the histone H3 family. As a core component of nucleosomes, HIST1H3A plays a central role in DNA packaging into chromatin, which directly impacts DNA accessibility to cellular machinery involved in transcription regulation, DNA repair, replication, and chromosomal stability. The importance of HIST1H3A in epigenetic research stems from its post-translational modifications, particularly at lysine 36 (K36), which contribute to the "histone code" that regulates chromatin structure and function .
Histone H3 has multiple variants (including H3.1, H3.2, and H3.3) that are encoded by different genes including HIST1H3A, HIST1H3B, HIST1H3C, and others. These variants can have distinct biological functions despite their high sequence similarity. The Ab-36 antibody specifically targets the lysine 36 region, which undergoes methylation associated with active transcription and other chromatin-associated processes .
When selecting an antibody for H3K36 modifications, researchers must first determine which methylation state (mono-, di-, or tri-methylation) they wish to detect, as each serves distinct biological functions:
H3K36me1: Generally associated with transcriptional regulation
H3K36me2: Distributed within large intragenic regions, regulating H3K27me3 distribution and possibly DNA methylation
H3K36me3: Primarily marks the body regions of actively transcribed genes
For accurate detection, validate antibody specificity using:
Peptide competition assays with modified and unmodified peptides
Knockdown/knockout of relevant methyltransferases
Western blot analysis using recombinant histones with defined modifications
Comparison with published ChIP-seq datasets as reference controls
Importantly, confirm the antibody's cross-reactivity profile against other histone modifications, especially those at nearby residues that might interfere with epitope recognition.
Histone H3K36 antibodies are utilized in multiple experimental contexts with distinct optimization requirements:
Each application requires validation of antibody performance under specific experimental conditions to ensure reliable results.
The relative abundance of different H3K36 methylation states varies across cell types and tissues, directly impacting experimental design and interpretation:
H3K36 methylation exists in three states—mono-, di-, and trimethylation (H3K36me1, H3K36me2, and H3K36me3). Mass spectrometry analyses have shown that regardless of H3 isoform (H3.1, H3.2, or H3.3), unmethylated and dimethylated H3K36 are the most abundant forms, each accounting for approximately 20-45% of total H3 in most examined mouse tissues. H3K36me3 typically occurs in only about 5% of total H3 .
These proportions can vary depending on:
Cell type and differentiation state
Tissue of origin
Disease conditions (particularly important in cancer research)
Experimental manipulations affecting methyltransferase or demethylase activity
Understanding these natural variations is essential when designing controls and interpreting results from antibody-based detection methods.
Different histone H3 variants show distinct functional properties despite high sequence similarity:
Canonical H3 (H3.1/H3.2) is predominantly incorporated into chromatin during DNA replication (replication-dependent), while H3.3 can be deposited independent of replication (replication-independent) . These variants have different roles in gene regulation and chromatin organization.
Studies in Drosophila have shown that mutations affecting lysine 36 in H3.2 (replication-dependent) versus H3.3 (replication-independent) have distinct phenotypic consequences. While K36R mutations in either H3.2 or H3.3 alone generally maintain Polycomb silencing, combined mutations display widespread Hox gene misexpression and developmental failure .
When selecting an antibody, consider:
Whether your research question requires distinguishing between H3 variants
If the antibody epitope includes regions that differ between variants
Whether post-translational modifications might differ in prevalence between variants
Distinguishing between methylation states requires careful antibody selection and experimental design:
Antibody specificity validation: Test against peptide arrays containing all three methylation states of H3K36 as well as other common histone modifications. Commercial antibodies should provide cross-reactivity data, but independent validation is recommended.
Sequential ChIP approach: For studying co-occurrence patterns, perform ChIP with one methylation state-specific antibody followed by a second immunoprecipitation with another state-specific antibody.
Genome-wide distribution analysis: Each methylation state shows characteristic genomic distribution patterns:
Correlation with functional outcomes: Analyze correlation with transcription rates, splicing patterns, and DNA repair efficiency to confirm the expected biological functions of each methylation state.
H3K36 methylation exists within a complex network of histone modifications with important regulatory relationships:
H3K36me and H3K27me3 antagonism: H3K36me2/3 inhibits Polycomb Repressive Complex 2 (PRC2), which catalyzes H3K27 methylation. This creates mutually exclusive chromatin domains and helps maintain proper gene expression patterns. Chromatin profiling revealed that K36R mutations in H3.2 disrupt H3K27me3 levels broadly throughout silenced domains .
Coordination with H3K4 methylation: H3K4me3 (marking active promoters) often works in concert with H3K36me3 (marking active gene bodies) to maintain active transcription states.
Interplay with histone acetylation: H3K36 methylation can recruit histone deacetylases to prevent cryptic transcription within gene bodies.
This crosstalk has important implications for experimental design and data interpretation. For example, when analyzing mutations in H3K36, researchers should also assess effects on H3K27me3 distribution, as demonstrated in the Drosophila studies where H3.2 K36R mutation disrupted H3K27me3 levels throughout silenced domains .
Non-specific binding is a common challenge when working with histone modification antibodies. To troubleshoot:
Increase stringency in washing buffers: Gradually increase salt concentration (from 150mM to 300mM NaCl) or add low concentrations of detergents like Tween-20 or Triton X-100.
Perform peptide competition assays: Pre-incubate antibody with specific and non-specific histone peptides to determine which signals are specific.
Test multiple antibody concentrations: Optimize antibody dilutions to find the concentration that maximizes specific signal while minimizing background.
Include specific controls:
Use samples with known K36 methylation status (e.g., cell lines with SETD2 knockout for H3K36me3)
Include IgG controls from the same species as the primary antibody
Include histone H3 knockout/knockdown controls where possible
Consider fixation conditions: Excessive cross-linking can mask epitopes and increase non-specific binding. Test different fixation protocols for immunofluorescence or ChIP applications.
H3K36 methylation shows context-dependent distribution patterns that researchers must consider:
In Drosophila development, studies have shown that H3.2K36 and H3.3K36 can functionally compensate for one another to repress Hox genes, but their mechanisms differ. H3.2K36 appears more important for maintaining global H3K27me3 levels even at late developmental time points .
The abundance of H3K36 methylation states also varies across mammalian tissues. While unmethylated and dimethylated H3K36 are typically the most abundant forms (20-45% of total H3), H3K36me3 accounts for only about 5% of total H3 in most tissues .
Additionally, changes in H3K36 methylation are associated with:
Cellular differentiation processes
Tissue-specific gene expression programs
Response to environmental stimuli
Disease progression, particularly in cancer contexts
When designing experiments, include appropriate tissue-matched or developmental stage-matched controls and consider the biological context when interpreting results.
H3K36 methylation serves critical functions in maintaining genome integrity through DNA damage repair mechanisms:
Double-strand break (DSB) repair pathway choice: H3K36me3 promotes homologous recombination (HR) repair by recruiting LEDGF, which subsequently helps localize CtIP to sites of DNA damage.
Nucleotide excision repair (NER): H3K36me3 facilitates recruitment of XPC and other NER factors to damaged DNA.
Mismatch repair (MMR): H3K36me3 recruits MSH2-MSH6 complex to chromatin during DNA replication, enhancing MMR efficiency in newly synthesized DNA.
When studying DNA damage repair processes, researchers should consider:
Cell cycle phase (as repair pathway choice and H3K36me3 levels vary through the cell cycle)
DNA damage induction method (as different types of damage may interact differently with H3K36-methylated chromatin)
The relationship between transcription and repair in H3K36me3-enriched regions
Rigorous controls are essential for interpreting ChIP experiments with H3K36 antibodies:
Input control: Chromatin sample before immunoprecipitation (typically 5-10% of IP material)
Negative controls:
IgG from same species as primary antibody
Non-transcribed regions (for H3K36me3)
Regions with known absence of specific modification
Positive controls:
Housekeeping gene bodies (for H3K36me3)
Regions with well-characterized H3K36 methylation patterns based on published datasets
Antibody validation controls:
Peptide competition with modified and unmodified peptides
Samples from cells with genetic manipulation of H3K36 methyltransferases
Western blot confirmation of antibody specificity
Spike-in controls: Consider using spike-in chromatin from a different species (e.g., Drosophila chromatin in human ChIP) to enable normalization across samples with potentially different global levels of H3K36 methylation.
Multi-layered validation approaches ensure antibody specificity:
Peptide array analysis:
Test against H3K36 peptides with different modification states
Test against peptides containing nearby modifications (e.g., K27, K37)
Test against peptides from different H3 variants
Western blot validation:
Compare signal in wildtype cells vs. cells with KO/KD of relevant methyltransferases
Test reactivity with recombinant histones carrying defined modifications
Peptide competition assays to confirm specificity
ChIP-seq correlation analysis:
Compare your results with published datasets using validated antibodies
Assess expected genomic distribution patterns (H3K36me3 enriched in gene bodies)
Correlation with RNA-seq data (H3K36me3 should correlate with transcription levels)
Immunofluorescence validation:
Compare nuclear localization patterns with published results
Evaluate co-localization with other nuclear markers
Test signal reduction in methyltransferase-depleted cells
The antibody should be validated specifically for each experimental application (WB, ChIP, IF) as performance can vary across techniques .
Optimize sample preparation for each application:
For Western blotting:
Use acid extraction methods (e.g., 0.2N HCl) to efficiently isolate histones
Include deacetylase and phosphatase inhibitors in extraction buffers
Optimize gel percentage (15-18% recommended for histones)
Use PVDF membranes rather than nitrocellulose for better retention
Consider using 5% BSA instead of milk for blocking (milk contains histones)
For ChIP/ChIP-seq:
Optimize fixation time (typically 10-15 minutes with 1% formaldehyde)
Fine-tune sonication to achieve 200-500bp fragments
Pre-clear chromatin with protein A/G beads
Include competing protein (e.g., BSA) in IP buffers
For Immunofluorescence:
Test different fixation methods (formaldehyde vs. methanol)
Include permeabilization step (0.1-0.5% Triton X-100)
Optimize epitope retrieval methods if necessary
Use sufficient blocking (3-5% BSA) to reduce background
Histone extraction methods can significantly impact antibody detection efficiency:
The choice of extraction method should be based on:
The specific modification being studied (some are more labile)
The downstream application requirements
The sensitivity of the antibody to different sample preparations
Always validate antibody performance with your chosen extraction method using appropriate controls.
For quantitative comparisons of H3K36 methylation:
Western blot quantification:
ChIP-qPCR quantification:
Express results as percent of input or as enrichment over IgG control
Use multiple primer pairs targeting regions with expected enrichment and depletion
Apply normalization to account for differences in chromatin preparation efficiency
ChIP-seq quantification:
Use spike-in controls for between-sample normalization
Apply appropriate normalization methods (RPKM, TMM, etc.)
Consider both peak intensity and breadth when comparing H3K36me3 patterns
Analyze correlation with transcription data
Mass spectrometry approach:
Provides absolute quantification of modification abundance
Can detect combinations of modifications on the same histone tail
Requires specialized equipment and expertise
Research has shown significant differences in H3K36 methylation levels between tissues and in disease states, with H3K36me3 accounting for only about 5% of total H3 in most tissues compared to 20-45% for unmethylated and dimethylated H3K36 .
When faced with discrepancies between techniques:
Consider method-specific biases:
ChIP-seq measures population averages across millions of cells
Immunofluorescence captures cell-to-cell variation but with lower resolution
Western blot provides bulk measurements without spatial information
Evaluate potential technical issues:
Antibody may perform differently under different experimental conditions
Fixation conditions affect epitope accessibility differently between methods
ChIP-seq signal is influenced by chromatin accessibility
Biological explanations:
Cell cycle differences (H3K36 methylation varies through the cell cycle)
Heterogeneity in cell populations
Context-dependent regulation of H3K36 methylation
Validation approaches:
Use alternative antibodies targeting the same modification
Apply orthogonal techniques (e.g., CUT&RUN, Mass spectrometry)
Genetic manipulation of writers/erasers to confirm specificity
Studies with H3 variant-specific mutations (H3.2K36R vs. H3.3K36R) demonstrate that seemingly subtle differences can have profound biological effects, with different impacts on H3K27me3 distribution and gene expression .
H3K36 methylation regulates multiple essential cellular processes:
Transcriptional regulation:
H3K36me3 marks actively transcribed gene bodies
Prevents cryptic transcription initiation within gene bodies
Influences RNA polymerase II elongation rate
RNA processing:
Regulates alternative splicing through recruitment of splicing factors
Influences mRNA export and stability
DNA repair:
Promotes homologous recombination at double-strand breaks
Facilitates mismatch repair and nucleotide excision repair
Maintains genome stability
Chromatin domain regulation:
Antagonizes Polycomb-mediated silencing (H3K27me3)
Influences higher-order chromatin structure
May regulate DNA methylation patterns
Development and differentiation:
When interpreting H3K36 methylation changes, consider which of these processes might be affected in your experimental context.
H3K36 mutations have significant implications for antibody-based studies:
Direct effects on antibody binding:
K36M mutations (lysine to methionine) prevent methylation and abolish antibody recognition
K36R mutations (lysine to arginine) similarly prevent methylation
Nearby mutations may alter epitope structure and affect antibody affinity
Biological effects complicating interpretation:
Experimental considerations:
Include sequencing of histone genes when working with cancer samples (H3K36 mutations are recurrent in certain cancers)
Use antibodies recognizing total H3 independently of K36 status
Consider the specific H3 variant being targeted in your experiment
Research in Drosophila has shown that K36R mutations in H3.2 significantly disrupt H3K27me3 levels throughout silenced domains, while these regions are mostly unaffected in H3.3K36R animals, highlighting the variant-specific effects of these mutations .
Several cutting-edge approaches are advancing H3K36 methylation studies:
CUT&RUN and CUT&Tag:
Higher signal-to-noise ratio than traditional ChIP
Require fewer cells (as few as 1,000 compared to millions for ChIP)
Better resolution of H3K36 methylation patterns
Single-cell epigenomics:
Reveals cell-to-cell variation in H3K36 methylation
Allows correlation with single-cell transcriptomics
Uncovers rare cell populations with distinct modification patterns
Long-read sequencing:
Enables detection of combinatorial histone modifications
Improves mapping to repetitive regions
Resolves allele-specific modification patterns
CRISPR-based epigenome editing:
Targeted modulation of H3K36 methylation at specific genomic loci
Allows causality testing between methylation and gene expression
Enables site-specific studies without global disruption
Mass spectrometry innovations:
Enhanced sensitivity for detecting low-abundance modifications
Improved quantification of modification stoichiometry
Better detection of co-occurring modifications
These technologies are providing unprecedented insights into H3K36 methylation dynamics and function, enabling more precise understanding of its roles in chromatin regulation.
Integrative analysis approaches enhance interpretation of H3K36 methylation data:
Correlation with transcriptomic data:
RNA-seq to correlate H3K36me3 with expression levels
NET-seq or PRO-seq to examine relationship with transcription elongation
RNA-splicing analyses to investigate connections with alternative splicing
Integration with other histone modifications:
Compare with H3K27me3 to identify antagonistic relationships
Analyze co-occurrence with H3K4me3 at active genes
Examine relationships with histone acetylation patterns
Chromatin accessibility correlation:
ATAC-seq or DNase-seq to relate H3K36 methylation to chromatin openness
Nucleosome positioning data to understand methylation in context of nucleosome organization
DNA methylation integration:
Whole-genome bisulfite sequencing to explore H3K36me-DNA methylation relationships
Targeted methylation analysis at H3K36me-enriched regions
Computational approaches:
Machine learning to identify complex patterns
Segmentation algorithms to define chromatin states
Network analysis to understand regulatory relationships
Studies have shown that H3K36 methylation influences distribution of H3K27me3 and potentially DNA methylation, highlighting the importance of integrative analysis approaches .