Di-methyl-HIST1H3A (K36) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism

The antibody targets the dimethylated form of lysine 36 on histone H3.1 (HIST1H3A), a post-translational modification (PTM) linked to transcriptional activation and chromatin accessibility . Histone H3K36me2 is recognized as a hallmark of actively transcribed genes, playing a role in recruiting chromatin-modifying enzymes and ensuring transcriptional fidelity .

PropertyDetail
AntigenDimethylated lysine 36 (H3K36me2) on histone H3.1 (HIST1H3A)
Modification TypeEpigenetic methylation (di-methylation)
Biological RoleRegulates transcription elongation, chromatin structure, and DNA repair
Key ApplicationsWB, IHC, IF, ChIP, ChIP-seq, IP

2.1. Polyclonal vs. Monoclonal Antibodies

  • Polyclonal Antibodies (e.g., ab9049, CAB2365):

    • Broad reactivity across species (human, mouse, rat, Drosophila, cow) .

    • Validated for ChIP, WB, and IHC .

    • Example: CAB2365 (Assay Genie) shows no cross-reactivity with mono- or trimethylated H3K36 .

  • Monoclonal Antibodies (e.g., MAB12834, ab318964):

    • Higher specificity for H3K36me2 .

    • Example: ab318964 (Abcam) is suitable for flow cytometry and IHC-P .

2.2. Specificity Validation

  • Negative Controls: Tested against unmodified, mono-, and trimethylated H3K36 peptides to confirm specificity .

  • Cross-Reactivity: No interaction with H3K36me1 or H3K36me3 in most cases .

3.1. Western Blotting (WB)

Protocol StepsDetails
Sample PreparationCalf thymus histone lysate or cell lysates (e.g., HeLa, NIH/3T3) .
Primary Antibody1:500–1:2000 dilution (e.g., CAB2365) ; 1 µg/mL (ab9049) .
DetectionHRP-conjugated secondary antibodies (e.g., goat anti-rabbit IgG) .
Observed Band~17 kDa (matches histone H3 size) .

3.2. Chromatin Immunoprecipitation (ChIP)

  • Purpose: Identifies genomic regions enriched for H3K36me2 .

  • Example: CAB2365 detects H3K36me2 at active promoters (e.g., γ-actin gene) .

3.3. Immunohistochemistry (IHC)

Protocol StepsDetails
SampleParaffin-embedded tissues (e.g., human placenta, mouse spleen) .
Antigen RetrievalHeat-mediated (Tris-EDTA buffer, pH 9.0) .
Primary Antibody1:200–1:2000 dilution (e.g., ab318964) ; 1 µg/mL (ab9049) .
StainingDAB or fluorescent dyes (e.g., AlexaFluor®488) .

4.1. Transcriptional Regulation

  • H3K36me2 marks actively transcribed regions and is linked to elongation efficiency .

  • CAB2365 revealed H3K36me2 enrichment at the γ-actin promoter in 293T cells .

4.2. Disease Implications

  • Cancer: Altered H3K36me2 patterns correlate with tumorigenesis and metastasis .

  • Developmental Biology: Critical for maintaining pluripotency and differentiation .

4.3. Technical Challenges

  • Cross-Reactivity: Avoid using antibodies with non-specific binding to mono- or trimethylated H3K36 .

  • Storage: Store at -20°C; avoid freeze-thaw cycles .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the method of purchase or location. Please consult your local distributor for specific delivery timeframes.
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. Nucleosomes function to wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that utilizes DNA as a template. Histones therefore play a crucial role in the regulation of transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex series of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Data indicate the mechanism for epigenetic regulation in cancer by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course can be helpful for determining whether the tumors are heterochronous PMID: 29482987
  3. Here, we report that JMJD5, a Jumonji C (JmjC) domain-containing protein, is a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that cause a DNA damage response. PMID: 28982940
  4. Data suggest that Ki-67 antigen proliferative index has important limitations and hhosphohistone H3 (PHH3) is an alternative proliferative marker. PMID: 29040195
  5. These results identify 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. This 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 show that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. These 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. We describe, 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 rare. PMID: 28547652
  14. Data 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 whereas 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 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 H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of 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. On the other hand, histone 3 mutations do not seem 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) serves 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 the 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 Di-methyl-HIST1H3A (K36) and its significance in epigenetic research?

Di-methyl-HIST1H3A (K36) refers to histone H3.1 protein (coded by the HIST1H3A gene) that has been dimethylated at the lysine 36 position. This specific histone modification plays a crucial role in chromatin structure and gene transcription regulation. Histone H3K36 dimethylation is associated with active gene expression and is involved in multiple cellular processes including DNA repair, alternative splicing, and transcriptional elongation . The significance of this modification lies in its role as part of the "histone code" that regulates DNA accessibility to cellular machinery, thereby controlling transcription, replication, and chromosomal stability .

How does Di-methyl-HIST1H3A (K36) differ from other histone H3 modifications?

Di-methyl-HIST1H3A (K36) is distinct from other histone H3 modifications in several important ways:

  • Genomic location: H3K36me2 typically occurs within the gene body of actively transcribed genes, distinguishing it from other modifications like H3K4me3 (promoters) or H3K27me3 (repressed regions) .

  • Functional outcomes: Unlike H3K9 methylation which is generally associated with heterochromatin and gene silencing, H3K36 dimethylation is linked to active transcription .

  • Degree of methylation: H3K36 can be mono-, di-, or tri-methylated (H3K36me1, H3K36me2, H3K36me3), with each state contributing to distinct chromatin states and having unique reader proteins and biological consequences .

  • Enzymatic regulation: Different methyltransferases and demethylases specifically target H3K36, such as the NSD family of methyltransferases that catalyze H3K36 dimethylation .

What are the common applications for Di-methyl-HIST1H3A (K36) antibodies?

Di-methyl-HIST1H3A (K36) antibodies are versatile tools with several key applications in epigenetic research:

ApplicationPurposeTypical Dilution
Western Blot (WB)Detection and quantification of H3K36me2 levels1:500 - 1:8000
Immunohistochemistry (IHC)Visualization of H3K36me2 distribution in tissue sections1:50 - 1:200
Immunofluorescence (IF)Cellular localization of H3K36me21:50 - 1:200
Chromatin Immunoprecipitation (ChIP)Genome-wide mapping of H3K36me2 distribution1:20 - 1:100
ChIP-sequencing (ChIP-seq)High-resolution mapping of H3K36me2 across the genome1:20 - 1:100
ELISAQuantitative detection of H3K36me21:32000 - 1:128000

These applications enable researchers to study the presence, distribution, and dynamics of H3K36 dimethylation in various biological contexts .

How can I verify the specificity of Di-methyl-HIST1H3A (K36) antibodies in my experimental system?

Verifying antibody specificity is crucial for reliable results in histone modification research. For Di-methyl-HIST1H3A (K36) antibodies, consider these methodological approaches:

  • Peptide competition assays: Pre-incubate the antibody with excess immunizing peptide (containing K36me2) before application to your samples. Specific signals should be dramatically reduced or eliminated .

  • Methyltransferase knockdown/knockout validation: Deplete known H3K36 methyltransferases (such as NSD family proteins) and confirm reduced signal with the antibody .

  • Cross-reactivity testing: Compare signals from your K36me2 antibody with antibodies specific for K36me1 and K36me3, as well as other methyl-lysine modifications (e.g., K9me2) to ensure specificity .

  • Positive and negative control samples: Use cell lines with known H3K36me2 status. For instance, HeLa, NIH/3T3, HEK-293T, and C2C12 cells have been validated as positive controls for many H3K36me2 antibodies .

  • Multiple antibody validation: Compare results from different vendors' H3K36me2 antibodies to rule out antibody-specific artifacts .

These methods collectively ensure that your observed signals truly represent H3K36 dimethylation rather than cross-reactivity or non-specific binding .

What are the best practices for optimizing ChIP protocols with Di-methyl-HIST1H3A (K36) antibodies?

Optimizing ChIP protocols for H3K36me2 requires attention to several critical factors:

  • Crosslinking conditions: For H3K36me2, which occurs in gene bodies, standard formaldehyde crosslinking (1% for 10 minutes) is generally effective, but optimization may be needed based on cell type .

  • Sonication parameters: Aim for chromatin fragments of 200-500bp for optimal resolution. Over-sonication can destroy epitopes while under-sonication reduces efficiency .

  • Antibody amount: Start with the recommended ratio (1:20 - 1:100 dilution) and titrate as needed . The optimal antibody amount depends on the abundance of H3K36me2 in your samples.

  • Washing stringency: For H3K36me2 ChIP, include high-salt washes to reduce background, but avoid overly stringent conditions that may disrupt specific interactions .

  • Input controls and normalization: Always process an input sample (pre-immunoprecipitation) alongside your ChIP to normalize for technical biases and starting material differences .

  • Positive control loci: Include primers for genes known to be enriched for H3K36me2 (active gene bodies) in qPCR validation, such as housekeeping genes .

  • Negative control regions: Include primers for regions expected to lack H3K36me2 (intergenic regions, silent genes) to confirm specificity .

Implementing these practices will help achieve high signal-to-noise ratios in H3K36me2 ChIP experiments .

How does genomic context influence H3K36 methylation patterns and antibody detection efficiency?

Genomic context significantly impacts both the distribution of H3K36 methylation and the efficiency of antibody detection:

  • Chromatin state variability: H3K36 methylation status varies with the local chromatin environment. Open, accessible chromatin regions may show different antibody binding efficiency compared to more compact chromatin regions .

  • Co-occurring modifications: The presence of other histone modifications can influence antibody accessibility to H3K36me2. For example, regions with high levels of H3K27 acetylation may affect the binding efficiency of H3K36me2 antibodies through steric hindrance or conformational changes .

  • Locus-specific effects: Research indicates that depletion of K36me3 has variable, locus-specific effects on the interactions of epigenetic readers, suggesting context-dependent functions of this modification .

  • Nucleosome positioning: The positioning of nucleosomes affects the exposure of H3K36me2 epitopes, potentially influencing antibody binding efficiency across different genomic regions .

  • Transcriptional status: Actively transcribed regions generally have different patterns of H3K36 methylation compared to silent regions, which can affect antibody detection threshold requirements .

Understanding these contextual influences is crucial for correctly interpreting ChIP-seq or immunofluorescence data that maps H3K36me2 distribution across the genome .

What are the key technical considerations when using Di-methyl-HIST1H3A (K36) antibodies in Western blotting?

Western blotting with Di-methyl-HIST1H3A (K36) antibodies requires attention to several technical aspects for optimal results:

  • Sample preparation: Extract histones using specialized acid extraction protocols to efficiently isolate histones from nuclei. Standard protein extraction methods may not effectively capture histone proteins .

  • Gel selection: Use high-percentage (15-18%) gels or specialized Triton-Acid-Urea gels to achieve good separation of histone proteins, which have low molecular weights (H3 appears at 15-18 kDa) .

  • Transfer conditions: Implement special transfer conditions (lower voltage, longer time) optimized for small proteins to ensure efficient transfer of histones to membranes .

  • Blocking optimization: Use BSA-based blocking solutions rather than milk, as milk contains casein which has high phosphate content that may interfere with phospho-specific antibodies that might be used alongside methylation-specific antibodies .

  • Dilution optimization: Start with the recommended dilution range (1:1000-1:8000) and adjust based on signal strength and background levels .

  • Loading controls: Use total H3 antibodies or housekeeping proteins as loading controls to normalize for variations in histone loading .

  • Positive controls: Include lysates from cells known to express H3K36me2, such as HeLa, NIH/3T3, C2C12, or HEK-293T cells .

Addressing these considerations will help achieve clear, specific detection of H3K36me2 in Western blotting applications .

How can researchers effectively troubleshoot non-specific binding or weak signals with Di-methyl-HIST1H3A (K36) antibodies?

Troubleshooting non-specific binding or weak signals requires systematic investigation of multiple parameters:

  • Weak signal troubleshooting:

    • Increase antibody concentration (decrease dilution)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Enhance detection system (switch to more sensitive chemiluminescence substrates)

    • Optimize extraction methods to improve histone yield

    • Check for protein degradation during sample preparation

  • Non-specific binding troubleshooting:

    • Increase antibody dilution to reduce background

    • Optimize blocking conditions (try different blocking agents or concentrations)

    • Increase washing stringency (more washes, higher salt concentration)

    • Pre-adsorb antibody with non-specific proteins

    • Filter secondary antibody to remove aggregates

    • Confirm antibody lot-to-lot consistency

  • Cross-reactivity analysis:

    • Perform peptide competition assays with both specific (K36me2) and non-specific (other methylated lysines) peptides

    • Compare binding patterns with alternative antibodies from different sources

    • Validate signals using genetic approaches (e.g., methyltransferase knockdown)

  • Sample-specific issues:

    • Ensure proper sample preparation to expose the epitope

    • Check if target modification is present in your biological system

    • Consider fixation effects on epitope accessibility (for IF/IHC)

Systematic investigation of these factors will help resolve most issues with antibody performance .

What are the recommended storage and handling conditions to maintain Di-methyl-HIST1H3A (K36) antibody efficacy?

Proper storage and handling are essential for maintaining antibody efficacy over time:

ParameterRecommendationRationale
Storage temperatureStore at -20°CPrevents antibody degradation
Freeze/thaw cyclesMinimize; aliquot upon receiptRepeated freeze/thaw cycles can reduce activity
Buffer compositionPBS with 0.02% sodium azide and 50% glycerol, pH 7.3Maintains stability and prevents microbial growth
Working dilution storageStore diluted antibody at 4°C for short term use onlyDiluted antibodies are less stable
Shipping conditionsShipped with ice packsMaintains cold chain during transport
Long-term stabilityStable for one year after shipment when stored properlyManufacturer testing data
Aliquoting necessityRecommended for larger volumesUnnecessary for -20°C storage of small volumes (≤20μl)
BSA additionSome preparations contain 0.1% BSAEnhances stability

Proper adherence to these storage and handling recommendations ensures maximum antibody performance and extends shelf life .

How can Di-methyl-HIST1H3A (K36) antibodies be used to investigate the relationship between histone modifications and disease states?

Di-methyl-HIST1H3A (K36) antibodies serve as valuable tools for investigating connections between this histone modification and various disease states:

  • Cancer research applications:

    • Mapping altered H3K36me2 patterns across cancer genomes to identify epigenetic signatures

    • Correlating H3K36me2 levels with oncogene expression or tumor suppressor silencing

    • Investigating NSD methyltransferase dysregulation, which is implicated in multiple cancer types through aberrant H3K36 methylation

  • Neurodegenerative disease studies:

    • Examining H3K36me2 changes in models of neurodegeneration

    • Correlating H3K36me2 patterns with altered gene expression in affected tissues

    • Investigating potential epigenetic therapeutic targets

  • Developmental disorders research:

    • Analyzing H3K36me2 distribution during normal and abnormal development

    • Studying consequences of mutations in H3K36 methyltransferases or demethylases

    • Correlating developmental gene expression programs with H3K36me2 patterns

  • Methodological approaches:

    • ChIP-seq to map genome-wide H3K36me2 changes in disease vs. normal states

    • Immunohistochemistry to visualize altered H3K36me2 in patient samples

    • Western blotting to quantify global H3K36me2 levels in different disease stages

    • Integration with transcriptome data to correlate H3K36me2 changes with gene expression

These applications help elucidate the mechanistic role of H3K36 dimethylation in disease pathogenesis and identify potential epigenetic biomarkers or therapeutic targets .

What are the recent advances in understanding the functional roles of different degrees of H3K36 methylation (mono-, di-, tri-)?

Recent research has revealed distinct functional roles for different degrees of H3K36 methylation:

  • H3K36 monomethylation (H3K36me1):

    • Recently identified as a distinct functional state rather than just an intermediate

    • Associated with enhancer regions in some cell types

    • May serve as a precursor mark that primes chromatin for subsequent modifications

    • Regulated by different enzymes than di- and tri-methylation in some contexts

  • H3K36 dimethylation (H3K36me2):

    • Enriched across gene bodies of actively transcribed genes

    • Plays a role in regulating alternative splicing by recruiting splicing factors

    • Involved in DNA damage response pathways

    • Mediated primarily by NSD family methyltransferases (NSD1, NSD2, NSD3)

    • Auto-inhibitory state of NSD enzymes is relieved by nucleosome engagement

  • H3K36 trimethylation (H3K36me3):

    • Highly enriched at the 3' ends of gene bodies

    • Marks active chromatin and is interpreted by specific epigenetic readers

    • In Drosophila, bound by MSL3 for X-chromosome dosage compensation

    • Interacts with PWWP-domain protein JASPer to recruit JIL1 kinase to active chromatin

    • Shows locus-specific effects when depleted, challenging previous uniform models

  • Context-dependent interactions:

    • Recent studies show that the three methylation states contribute to distinct chromatin states

    • The transition between different methylation states is regulated by specific writer and eraser enzymes

    • Reader proteins show different affinities for mono-, di-, and tri-methylated H3K36

    • Genomic context influences the biological outcomes of different methylation states

These discoveries challenge prevailing models and highlight the nuanced roles of different H3K36 methylation states in chromatin regulation .

How can Di-methyl-HIST1H3A (K36) antibodies be integrated into multi-omics approaches to study epigenetic regulation?

Integration of Di-methyl-HIST1H3A (K36) antibodies into multi-omics approaches enables comprehensive analysis of epigenetic regulation:

  • ChIP-seq and RNA-seq integration:

    • Map H3K36me2 distribution using ChIP-seq with the antibody

    • Correlate with gene expression data from RNA-seq

    • Analyze how H3K36me2 patterns relate to transcriptional activity

    • Identify genes whose expression correlates strongly with H3K36me2 enrichment

  • Epigenome and proteome connections:

    • Use H3K36me2 antibodies in ChIP followed by mass spectrometry (ChIP-MS)

    • Identify proteins that interact with H3K36me2-enriched chromatin regions

    • Characterize reader proteins that specifically recognize H3K36me2

  • Multi-mark epigenomic profiling:

    • Perform sequential ChIP (Re-ChIP) with H3K36me2 antibodies and antibodies against other histone marks

    • Identify regions with co-occurrence of multiple modifications

    • Implement CUT&RUN or CUT&Tag methods with H3K36me2 antibodies for higher resolution mapping

    • Combine with ATAC-seq to correlate H3K36me2 with chromatin accessibility

  • Single-cell approaches:

    • Adapt H3K36me2 antibodies for single-cell ChIP-seq or CUT&Tag

    • Integrate with single-cell RNA-seq to correlate H3K36me2 and gene expression at single-cell resolution

    • Study cell-to-cell variability in H3K36me2 distribution

  • 4D Nucleome integration:

    • Combine H3K36me2 ChIP-seq with Hi-C or other chromosome conformation capture techniques

    • Analyze how H3K36me2 correlates with three-dimensional genome organization

    • Study the role of H3K36me2 in forming topologically associating domains (TADs)

These integrated approaches provide a comprehensive understanding of H3K36me2's role in coordinating various aspects of genome function and regulation .

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.