HIST1H3A (Ab-36) Antibody

Shipped with Ice Packs
In Stock

Description

Western Blot Performance

  • 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 .

Immunofluorescence/Immunocytochemistry (ICC/IF)

  • 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 .

Chromatin Immunoprecipitation (ChIP)

  • 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) .

Comparative Analysis of H3K36-Targeting Antibodies

AntibodySpecificityHostApplicationsKey Distinction
ab9050 (Abcam) H3K36me3RabbitWB, ICC/IF, ChIPValidated in >975 publications
ab9048 (Abcam) H3K36me1RabbitWB, ICC/IFLower cross-reactivity with H3K36me2/3
29202-1-AP (Proteintech) H3K36me2RabbitWB, ELISAHigher affinity for di-methylated states
AF7836 (Affinity) Pan-H3 (unmodified)RabbitWBBroad reactivity across species

Research Findings and Challenges

  • 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:

    • H3K36me2 is deposited by SETD2 and linked to mRNA splicing fidelity .

    • Loss of H3K36me2 correlates with transcriptional readthrough and genomic instability .

Recommended Workflow for Validation

  1. Positive Controls: Use histone extracts from HeLa or HEK-293T cells .

  2. Blocking Conditions: 1% BSA with 10% normal goat serum .

  3. Secondary Antibodies: Alexa Fluor® 488/594 conjugates for multiplex imaging .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
Histone H3 is a core component of nucleosomes, which wrap and compact DNA into chromatin. This compaction limits DNA accessibility to cellular machinery that requires DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is controlled by a complex set of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer is influenced by the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course may be helpful in determining whether tumors are heterochronous. PMID: 29482987
  3. Recent studies show that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Findings suggest that the Ki-67 antigen proliferative index has significant limitations and that phosphohistone H3 (PHH3) offers an alternative proliferative marker. PMID: 29040195
  5. These results pinpoint cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Data demonstrate that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. Experiments showed that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl) where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This research provides, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3), which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Studies show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest, while the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. Results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. Conversely, histone 3 mutations do not appear to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. Taken together, the authors verified that histone H3 is a real substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) plays a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Data indicate that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays is regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is HIST1H3A and why is it important in epigenetic research?

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 .

How do I determine the appropriate antibody specificity for studying H3K36 modifications?

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.

What are the common applications for histone H3K36 antibodies in research?

Histone H3K36 antibodies are utilized in multiple experimental contexts with distinct optimization requirements:

ApplicationCommon UsageKey Considerations
Western Blotting (WB)Quantifying global H3K36 methylation levelsRequires denaturing conditions; validated for detecting specific methylation states
ChIP/ChIP-seqGenome-wide profiling of H3K36 methylationRequires optimization of fixation, sonication, and antibody concentration
Immunocytochemistry/ Immunofluorescence (ICC/IF)Visualizing nuclear distribution of H3K36 methylationFixation method critical; validated for cellular localization studies
CUT&RUN/CUT&TagHigh-resolution mapping with lower cell inputsIncreasingly popular alternatives to traditional ChIP

Each application requires validation of antibody performance under specific experimental conditions to ensure reliable results.

What affects the abundance of H3K36 methylation states in experimental samples?

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.

How do histone H3 variants differ and how might this affect antibody selection?

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

How can I differentiate between H3K36me1, H3K36me2, and H3K36me3 marks in my experiments?

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:

    • H3K36me3: Enriched in gene bodies of actively transcribed genes, particularly in exons

    • H3K36me2: More broadly distributed within large intragenic regions

    • H3K36me1: Often transitional and less studied

  • 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.

What is the relationship between H3K36 methylation and other histone modifications?

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 .

How can I troubleshoot non-specific binding when using H3K36 antibodies?

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.

How do H3K36 methylation patterns differ between cell types and developmental stages?

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.

What role does H3K36 methylation play in DNA damage repair?

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

What controls should I include when performing ChIP experiments with H3K36 antibodies?

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.

How can I effectively validate the specificity of an H3K36 antibody?

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 .

What sample preparation techniques maximize signal-to-noise ratio with H3 antibodies?

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

  • Optimize antibody concentration and incubation time

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

  • Include DAPI counterstain to confirm nuclear localization

What are the implications of different histone extraction methods for antibody-based detection?

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.

How can I quantitatively compare H3K36 methylation levels between experimental conditions?

For quantitative comparisons of H3K36 methylation:

  • Western blot quantification:

    • Always normalize H3K36me signals to total H3 levels

    • Use a standard curve of recombinant histones for absolute quantification

    • Apply appropriate statistical tests for replicate experiments

    • Consider that certain H3 antibodies may not bind equally to all H3 variants

  • 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 .

How do I interpret contradictory results between ChIP-seq and immunofluorescence data?

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 .

What biological processes are most strongly associated with alterations in H3K36 methylation?

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:

    • H3K36 mutations in Drosophila cause developmental defects and Hox gene misexpression

    • Proper H3K36 methylation is required for lineage-specific gene expression

    • Dysregulation is associated with developmental disorders

When interpreting H3K36 methylation changes, consider which of these processes might be affected in your experimental context.

How do H3K36 mutations affect antibody binding and experimental interpretation?

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:

    • K36M acts as a dominant negative, inhibiting methylation on wild-type H3

    • Different effects depending on H3 variant (H3.2K36R vs. H3.3K36R have distinct phenotypes)

    • Global redistribution of other modifications (especially H3K27me3)

  • 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 .

What emerging technologies are enhancing H3K36 methylation research?

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.

How do I integrate H3K36 methylation data with other genomic and epigenomic datasets?

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 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.