Di-methyl-HIST1H3A (K9) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

Di-methyl-HIST1H3A (K9) antibody specifically recognizes the di-methylated form of histone H3.1 at lysine 9 (H3K9me2), a core component of nucleosomes. This modification is critical for:

  • Establishing repressive chromatin states by blocking transcription factor binding .

  • Facilitating chromosome segregation through centromeric chromatin organization .

  • Regulating DNA replication and repair via epigenetic silencing .

Antibody Development and Production

The antibody is manufactured using advanced recombinant technologies:

Key Steps in Production:

  1. Antigen Design: Synthetic peptides corresponding to H3K9me2 are used to immunize rabbits or mice .

  2. Gene Cloning: Antibody genes from immunized hosts are cloned into expression vectors .

  3. Expression: Vectors are transfected into suspension cells (e.g., CHO or HEK293) for large-scale antibody production .

  4. Purification: Affinity chromatography isolates antibodies from cell culture supernatant .

  5. Validation: Specificity is confirmed via ELISA, western blot (WB), and immunohistochemistry (IHC) .

Host Systems and Clonality:

PropertyExamples from Suppliers
Host SpeciesRabbit (polyclonal: Avantor ; monoclonal: HUABIO ), Mouse (monoclonal: Abcam )
ClonalityMonoclonal (Abcam , Cellsignal , HUABIO ) or polyclonal (Avantor )
Purification MethodProtein G affinity chromatography , Antigen-affinity purification

Key Applications:

ApplicationDilution RangeSupported Species
Western Blot (WB)1:500–1:100,000 Human, Mouse, Rat
Immunofluorescence (IF)1:30–1:500 Human, Mouse
Chromatin IP (ChIP)Not specifiedHuman
IHC1:200–1:500 Human tissues

Performance Data:

  • Band Size: 15–17 kDa in WB, consistent with histone H3 .

  • Specificity: No cross-reactivity with H3K9me1, H3K9me3, or methylated residues at K4/K27 .

  • Sensitivity: Detects endogenous H3K9me2 in HeLa, PC-12, and calf thymus lysates .

Supplier Comparison

Commercial availability and pricing:

SupplierCatalog NumberClonalityHostSizePrice (USD)
Abcamab1220 MonoclonalMouse100 µg$410
Cellsignal#9753 MonoclonalRabbit100 µL$39
Avantor77687-920 PolyclonalRabbit100 µLNot listed
HUABIOEM1108 MonoclonalRabbit50–100 µL$148–248

Research Findings and Clinical Relevance

  • Gene Repression: H3K9me2 recruits HP1 proteins to silence euchromatic regions .

  • Disease Link: Dysregulation of H3K9me2 is implicated in cancers and developmental disorders .

  • Centromere Function: Essential for maintaining centromeric integrity during mitosis .

Validation and Quality Control

  • ELISA Specificity: Binds exclusively to H3K9me2 peptides (EC₅₀ = 0.1–1.0 µg/mL) .

  • Negative Controls: No staining observed with unmodified H3 peptides or mismatched secondary antibodies .

  • Batch Consistency: Over 960 publications cite Abcam’s ab1220, demonstrating reproducibility .

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 purchasing method or location. Please consult your local distributor for specific delivery timelines.
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 the nucleosome, which is responsible for packaging and compacting DNA into chromatin. This process limits the accessibility of DNA to cellular machinery requiring DNA as a template, including those involved in transcription regulation, DNA repair, DNA replication, and chromosomal stability. Histones play a crucial role in these processes. DNA accessibility is regulated through a complex system of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer involves the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Studies have shown that increased expression of H3K27me3 during a patient's clinical course can aid in determining whether tumors are heterochronous. PMID: 29482987
  3. Recent findings demonstrate that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Research indicates that the Ki-67 antigen proliferative index has limitations, and phosphohistone H3 (PHH3) is an alternative marker for proliferation. 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. Data suggests that in the early developing human brain, HIST1H3B comprises the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, the histone H3-K27M mutation was found to be mutually exclusive with the IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with the BRAF-V600E mutation and was frequently associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Evidence suggests that the histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, and deposits histone H3.3 onto these genomes. PMID: 28981850
  9. Experiments have shown 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 study provides the first description of the MR imaging features of pediatric diffuse midline gliomas with a 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 associated with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. Research indicates that the H3F3A K27M mutation is not uncommon in adult cerebellar HGG. PMID: 28547652
  14. Data suggests 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. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Research suggests that the 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. This interaction 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 the formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications have been observed in leukocytes exposed to traffic-derived airborne particulate matter. PMID: 27918982
  20. Persistent histone H3 serine 10 or serine 28 phosphorylation plays a key role in chemical carcinogenesis through the regulation of gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, a predisposition to recurrence, and tumor location in the right cerebellar hemisphere. Conversely, histone 3 mutations appear to be absent in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1, effectively controlling the DHRS4 gene cluster. PMID: 26864944
  23. Data suggests that nuclear antigen Sp100C is a multifaceted sensor for histone H3 methylation and phosphorylation. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters chromatin structure during specific phases of mitosis to facilitate timely condensin I and cohesin disassociation, which is essential for proper chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA promotes an open conformation of UHRF1, facilitating its recognition of H3 histones. PMID: 27045799
  26. H3K9me3 plays a significant role in hypoxia, apoptosis, and the repression of APAK. PMID: 25961932
  27. These findings confirmed that histone H3 is a real substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. Circulating H3 levels were found to correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Studies demonstrate that double mutations on residues in the interface (L325A/D328A) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Research suggests that minichromosome maintenance protein 2 (MCM2) binding is not required for the incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a crucial mechanistic role in the maintenance of leukemia stem cells (LSC). 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 indicates that lower-resolution mass spectrometry instruments can be used for the analysis of histone post-translational modifications (PTMs). PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the 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 are regulated by a balance between histone H3K9 acetylation and methylation. 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 (K9) and why is it significant in epigenetic research?

Di-methyl-HIST1H3A (K9), commonly known as H3K9me2, refers to histone H3 that has been di-methylated at the lysine 9 residue. This post-translational modification represents one of the numerous modifications that histones can undergo, including acetylation, methylation, phosphorylation, and ubiquitylation. H3K9me2 is particularly significant as it serves as a hallmark of repressive chromatin and is often found at silent gene loci, playing a crucial role in the formation of facultative heterochromatin . This modification creates a repressive chromatin environment by preventing the binding of transcription factors and other regulatory proteins to DNA . The significance of H3K9me2 extends beyond gene silencing to essential cellular processes such as chromosome segregation, as this modification is prominently found at centromeres . Understanding H3K9me2 patterns provides researchers with valuable insights into epigenetic regulation mechanisms underlying development, differentiation, and disease progression.

How does H3K9me2 differ functionally from H3K9me3?

While both H3K9me2 and H3K9me3 are repressive marks, they exhibit distinct genomic distribution patterns and functional roles. H3K9me2 is predominantly associated with facultative heterochromatin and is dynamically regulated during cellular processes. It typically spans broader genomic regions including gene bodies and intergenic regions. In contrast, H3K9me3 is more strongly associated with constitutive heterochromatin formation, particularly at repetitive elements and pericentromeric regions . H3K9me3 typically creates a more stable and condensed chromatin state than H3K9me2. The enzymes responsible for establishing these marks are also different—various methyltransferases including G9a and GLP primarily catalyze H3K9me2, while SUV39H1/2 are the main enzymes responsible for H3K9me3. Functionally, H3K9me2 often participates in lineage-specific gene silencing and stress responses, whereas H3K9me3 maintains long-term silencing of repetitive elements and contributes to genome stability .

What is the role of H3K9me2 in chromatin organization?

H3K9me2 plays a fundamental role in organizing chromatin structure by contributing to the formation of compacted, transcriptionally repressive chromatin domains. This modification works in concert with other repressive marks, such as H3K27me3 and H2AK119u1, to establish and maintain heterochromatin . At centromeres, H3K9me2 contributes to proper chromosome segregation during cell division by helping to maintain centromeric structure and function . The presence of H3K9me2 serves as a docking site for heterochromatin protein 1 (HP1), which oligomerizes and recruits other factors involved in chromatin compaction. This modification participates in the establishment of nuclear lamina-associated domains (LADs) and other repressive compartments within the nucleus. The genomic distribution of H3K9me2 is not random but forms specific patterns that help organize the genome into functional domains, contributing to the three-dimensional organization of chromatin within the nucleus and ultimately influencing cellular identity and function.

What are the optimal conditions for using Di-methyl-HIST1H3A (K9) antibodies in ChIP experiments?

For optimal chromatin immunoprecipitation (ChIP) using Di-methyl-HIST1H3A (K9) antibodies, researchers should follow several key procedures. Begin with proper crosslinking—typically 1% formaldehyde for 10 minutes at room temperature is effective for capturing H3K9me2 interactions. Sonication should be optimized to generate chromatin fragments between 200-500bp, as larger fragments may reduce resolution while smaller fragments might decrease efficiency. When selecting an antibody, highly specific monoclonal antibodies such as mAbcam 1220 have been validated extensively for ChIP applications . For immunoprecipitation, use 2-5μg of antibody per 25μg of chromatin, and include overnight incubation at 4°C with gentle rotation. Washing stringency is critical—typically four washes with increasing salt concentration help reduce background while maintaining specific interactions. For elution and reversal of crosslinks, incubate samples at 65°C for 4-6 hours. Include appropriate controls: an IgG control to assess non-specific binding, input chromatin (5-10%) to normalize enrichment, and positive/negative genomic regions where H3K9me2 is known to be present or absent. Many commercial antibodies for H3K9me2 have been validated in ChIP applications with specific recommended dilutions—for instance, the mAbcam 1220 has been cited in over 960 publications, demonstrating its reliability for this application .

How can I ensure specificity when detecting H3K9me2 in Western Blotting?

Ensuring specificity when detecting H3K9me2 in Western Blotting requires careful attention to several experimental parameters. First, extract histones using an acid extraction protocol (0.2N HCl or 0.4N H2SO4) to isolate histones efficiently from other nuclear proteins. Run SDS-PAGE using 15-18% gels, which provide better resolution for low molecular weight histone proteins. Transfer proteins to PVDF membranes rather than nitrocellulose, as PVDF has better retention properties for small proteins like histones. Block with 5% BSA rather than milk, as milk contains casein which has been reported to cross-react with some histone antibodies. For the primary antibody incubation, use validated antibodies at manufacturer-recommended dilutions—typically 1:500-1:5000 for Western blot applications . Include a peptide competition assay where the antibody is pre-incubated with the specific peptide containing H3K9me2 which should abolish the signal if the antibody is specific. Always include appropriate controls: unmodified H3, other methylation states of H3K9 (me1, me3), and if possible, samples from cells treated with specific methyltransferase inhibitors. Strip and reprobe blots with a total H3 antibody to normalize loading. Additionally, the specificity of the signal can be verified using samples where the modification is known to be enriched or depleted, such as cells treated with G9a/GLP inhibitors which should show reduced H3K9me2 levels.

What protocols provide the best results for immunofluorescence using Di-methyl-HIST1H3A (K9) antibodies?

For optimal immunofluorescence (IF) results with Di-methyl-HIST1H3A (K9) antibodies, a detailed protocol should include the following steps. Begin with effective fixation using 4% paraformaldehyde for 10 minutes at room temperature, followed by permeabilization with 0.2% Triton X-100 for 10 minutes. A critical step often overlooked is antigen retrieval—incubate slides in 10mM sodium citrate buffer (pH 6.0) at 95°C for 20 minutes to expose epitopes that may be masked during fixation. Block thoroughly using 3% BSA with 10% normal serum from the species of the secondary antibody for 1 hour at room temperature. For primary antibody incubation, use validated antibodies at appropriate dilutions (typically 1:30-1:200 for IF applications) . Incubate overnight at 4°C in a humidified chamber to ensure consistent staining. For washing, use TBS-T (0.1% Tween-20) with at least three 5-minute washes between steps. Use highly cross-adsorbed secondary antibodies to prevent non-specific binding. Counterstain with DAPI (1μg/ml) to visualize nuclei. Mount using an anti-fade mounting medium to preserve fluorescence. Include proper controls: a no-primary antibody control to assess secondary antibody specificity, and if possible, samples with known patterns of H3K9me2 enrichment. When imaging, capture multiple fields and z-stacks to ensure representative analysis. Quantitative analysis of H3K9me2 staining patterns can be performed using software like ImageJ with appropriate background subtraction and normalization to DAPI signal.

How can I validate the specificity of a Di-methyl-HIST1H3A (K9) antibody?

Validating the specificity of a Di-methyl-HIST1H3A (K9) antibody requires a comprehensive approach using multiple methods. Begin with peptide array analysis, where the antibody is tested against a panel of synthetic histone peptides containing various modifications (including H3K9me1, H3K9me2, H3K9me3, and other methylated lysines on H3). Results should show binding primarily to H3K9me2 peptides with minimal cross-reactivity. Conduct dot blot analysis with decreasing concentrations of modified peptides to determine the detection limit and confirm specificity. Western blot validation should include histone extracts from wild-type cells alongside extracts from cells treated with G9a/GLP inhibitors (such as UNC0638) which should show decreased H3K9me2 signal. Ideally, include CRISPR-generated knockout/knockdown models of H3K9 methyltransferases as additional controls. For ChIP validation, perform ChIP-qPCR on regions known to be enriched or depleted for H3K9me2, such as certain heterochromatic regions versus active promoters. ChIP-seq analysis should show enrichment patterns consistent with established H3K9me2 distributions. Immunofluorescence patterns should show expected nuclear distributions—typically peripheral and perinucleolar heterochromatin staining for H3K9me2. The mAbcam 1220 antibody (ab1220) serves as an example of a well-validated antibody, having been cited in over 960 publications and trusted by researchers since 2005 . Proper validation ensures experimental results are reliable and interpretable in the context of epigenetic research.

What controls should be included in H3K9me2 ChIP-seq experiments?

A robust H3K9me2 ChIP-seq experiment requires several essential controls to ensure data reliability and interpretability. First, include an input control (non-immunoprecipitated chromatin) from the same sample, processed identically except for the immunoprecipitation step, to normalize for biases in chromatin preparation and sequencing. An IgG control immunoprecipitation using the same antibody isotype and concentration helps establish background enrichment levels. For antibody specificity control, perform parallel ChIP-seq with antibodies against different methylation states (H3K9me1, H3K9me3) to confirm distinct enrichment patterns. Include a spike-in normalization control using a small amount of chromatin from a different species (e.g., Drosophila) and a species-specific antibody to allow for quantitative comparisons between samples. Technical replicates (at least 2-3) are crucial to assess experimental variability. For biological validation, include samples where H3K9me2 levels have been experimentally altered—such as cells treated with G9a/GLP inhibitors or cells with knockdown/knockout of relevant methyltransferases. Positive and negative control regions should be validated by ChIP-qPCR prior to sequencing; regions like satellite repeats typically show high H3K9me2 enrichment, while active promoters show depletion. A total H3 ChIP-seq in parallel helps normalize for nucleosome occupancy, which is particularly important when comparing different cell types or treatment conditions where chromatin accessibility may vary.

How can I resolve cross-reactivity issues with other histone modifications?

Resolving cross-reactivity issues with Di-methyl-HIST1H3A (K9) antibodies requires a systematic approach addressing multiple aspects of the experimental workflow. First, perform careful antibody selection—monoclonal antibodies like mAbcam 1220 or recombinant antibodies typically offer higher specificity than polyclonal antibodies. Review validation data including peptide arrays and dot blots that test against multiple histone modifications. Implement peptide competition assays by pre-incubating the antibody with excess H3K9me2 peptide and other modified peptides (H3K9me1, H3K9me3, etc.) to identify potential cross-reactivity. Optimize antibody concentration—excessive antibody can increase non-specific binding; follow manufacturer-recommended dilutions (e.g., 1:500-1:5000 for WB, 1:30-1:200 for IF) . Increase washing stringency by using higher salt concentrations or adding detergents like Tween-20 or Triton X-100 to reduce non-specific interactions. Consider using blocking peptides containing potentially cross-reactive modifications to selectively block unwanted interactions. For Western blotting, run multiple histone modification standards in parallel and compare band patterns. In ChIP experiments, perform sequential ChIP (re-ChIP) by first pulling down with an H3K9me2 antibody followed by immunoprecipitation with antibodies against potentially cross-reactive modifications; lack of enrichment confirms specificity. If cross-reactivity persists, consider antibody affinity purification against the specific H3K9me2 peptide to remove cross-reactive antibodies from the preparation. Data analysis approaches, such as computational removal of signals that correlate with known cross-reactive modifications, can help mitigate remaining issues post-experiment.

How can Di-methyl-HIST1H3A (K9) antibodies be used to study heterochromatin dynamics?

Di-methyl-HIST1H3A (K9) antibodies provide powerful tools for investigating heterochromatin dynamics across various biological contexts. For studying cell cycle-dependent heterochromatin changes, combine H3K9me2 immunofluorescence with cell cycle markers and perform synchronized cell experiments, collecting samples at different time points for ChIP-seq or immunofluorescence analysis. To examine heterochromatin establishment during development, perform H3K9me2 ChIP-seq at different developmental stages, correlating changes with transcriptional reprogramming events. For investigating heterochromatin boundary formation and maintenance, combine H3K9me2 ChIP-seq with ChIP-seq for boundary elements like CTCF or active histone marks (H3K4me3, H3K27ac). Live-cell imaging of heterochromatin dynamics can be achieved using tagged reader proteins that specifically recognize H3K9me2, such as modified HP1 domains, combined with photo-bleaching techniques (FRAP/FLIP) to measure turnover rates. Super-resolution microscopy (STORM, PALM, or SIM) with H3K9me2 antibodies allows visualization of heterochromatin domains at nanoscale resolution, revealing structural details not visible with conventional microscopy. For dissecting the role of specific heterochromatin regions, combine H3K9me2 ChIP with DNA FISH to visualize the spatial organization of specific genomic regions enriched for this mark. Genome editing of key heterochromatin regions followed by H3K9me2 ChIP-qPCR can determine how sequence elements contribute to heterochromatin establishment. Since H3K9me2 is associated with centromeres and plays a role in chromosome segregation , these antibodies can also be used to study centromere function during mitosis through co-localization studies with centromeric proteins.

What approaches can be used to study the interplay between H3K9me2 and other histone modifications?

Studying the interplay between H3K9me2 and other histone modifications requires integrative approaches spanning multiple techniques. Sequential ChIP (re-ChIP) represents a powerful method where chromatin is first immunoprecipitated with H3K9me2 antibodies, then subjected to a second immunoprecipitation with antibodies against other modifications, revealing regions where both marks co-exist. Mass spectrometry analysis of immunoprecipitated histones can identify combinations of modifications present on the same histone tail or within the same nucleosome. Proximity ligation assays (PLA) using antibodies against H3K9me2 and other modifications can detect their co-occurrence within 40nm in situ. Genome-wide correlation analysis of ChIP-seq data for H3K9me2 and other modifications helps identify patterns of co-enrichment or mutual exclusivity. Perturbation experiments targeting specific modifying enzymes (e.g., G9a/GLP inhibitors) followed by ChIP-seq for multiple marks can reveal hierarchical relationships and dependencies between modifications. Co-immunoprecipitation of chromatin-modifying complexes followed by activity assays can determine how the presence of one modification affects the deposition of others. Developmental time course experiments tracking multiple modifications simultaneously can reveal sequential establishment patterns. Research has demonstrated important crosstalk relationships, such as those between H3K9me2 and H3K4me3, where repressive H3K9me2 marks generally show mutual exclusivity with the active H3K4me3 mark at promoters . Similarly, studies have shown relationships between H3K9me2 and histone acetylation, where acetylation is typically depleted in regions enriched for H3K9me2 . Understanding these relationships provides insight into the complex regulatory mechanisms governing chromatin states and gene expression.

How does H3K9me2 distribution change during cellular differentiation?

H3K9me2 distribution undergoes significant remodeling during cellular differentiation, reflecting its role in cell fate decisions and lineage commitment. In embryonic stem cells (ESCs), H3K9me2 is initially present at relatively low levels and in small domains, primarily restricting the expression of lineage-inappropriate genes. As differentiation progresses, H3K9me2 domains expand dramatically, forming large organized chromatin K9-modification (LOCK) domains that can span hundreds of kilobases. These expanded domains correlate with the silencing of pluripotency genes and alternative lineage genes inappropriate for the chosen differentiation path. ChIP-seq studies comparing H3K9me2 profiles between ESCs and differentiated cells reveal a progressive increase in genome coverage, from approximately 10-15% in ESCs to 40-50% in fully differentiated cells. The recruitment of H3K9me2 occurs in a lineage-specific manner, with different genomic regions targeted in different cell types. For instance, neural differentiation involves H3K9me2 accumulation at genes associated with mesodermal and endodermal lineages. Time-course ChIP-seq during differentiation shows that H3K9me2 establishment often precedes transcriptional silencing, suggesting a causative role in gene repression rather than simply reflecting already silent genes. The H3K9 methyltransferases G9a and GLP show increased binding to target loci during differentiation, correlating with increased H3K9me2 levels. Single-cell approaches have revealed significant heterogeneity in H3K9me2 distribution during early differentiation stages, which resolves into more homogeneous patterns as cells commit to specific lineages. Integration of H3K9me2 ChIP-seq with Hi-C data shows that these modifications contribute to the establishment of closed chromatin compartments and TAD boundaries during differentiation.

How should ChIP-seq data for H3K9me2 be normalized and analyzed?

H3K9me2 ChIP-seq data analysis requires specialized approaches due to the broad distribution patterns of this modification. For normalization, always use input control to correct for biases in chromatin preparation, mapability, and copy number variations. Spike-in normalization with exogenous chromatin (e.g., Drosophila chromatin) is particularly valuable for quantitative comparisons between samples, as H3K9me2 can cover large portions of the genome making global normalization methods inappropriate. For peak calling, standard narrow peak callers (e.g., MACS2) are insufficient for H3K9me2; instead, use domain-based approaches like SICER, RSEG, or diffReps that identify broad enriched regions. H3K9me2 typically forms large domains rather than sharp peaks, so analysis should focus on identifying domains and their boundaries. Signal smoothing using a window approach (e.g., 5-10kb bins) helps identify meaningful H3K9me2 domains while reducing noise. Compare H3K9me2 ChIP-seq data with other histone modifications, particularly active marks like H3K4me3 and H3K27ac, to identify regions of mutual exclusivity or unexpected co-occurrence. For differential analysis between conditions, tools like diffBind or DiffReps that account for the broad nature of H3K9me2 enrichment are preferable. When visualizing H3K9me2 data, use extended genomic views (50kb-1Mb) to appreciate domain structures, rather than focusing on promoter-proximal regions. Integrative analysis with gene expression data often reveals inverse correlations, with high H3K9me2 corresponding to low expression. Aggregate analyses examining average H3K9me2 profiles across genomic features (TSS, gene bodies, enhancers) can reveal global patterns of enrichment and depletion.

What bioinformatic approaches are recommended for interpreting H3K9me2 distribution patterns?

Interpreting H3K9me2 distribution patterns requires specialized bioinformatic approaches that account for its unique genomic distribution. Domain identification algorithms that employ hidden Markov models (ChromHMM, EpiCSeg) or dynamic programming approaches (RSEG) can effectively capture the broad, domain-like nature of H3K9me2 enrichment. Genome segmentation approaches integrating multiple histone modifications, including H3K9me2, provide a comprehensive view of chromatin states across the genome. Correlation analysis with DNA sequence features can reveal underlying determinants of H3K9me2 distribution—for instance, GC content, repetitive elements, and CpG density often correlate with H3K9me2 patterns. Comparative analysis across cell types or conditions should incorporate statistical frameworks that account for the broad distribution, such as diffHic or diffTF. Meta-region analysis examining H3K9me2 distribution across annotated genomic features (promoters, enhancers, boundaries) reveals global patterns and relationships with functional elements. Integration with three-dimensional chromatin architecture data (Hi-C, ChIA-PET) can reveal relationships between H3K9me2 domains and higher-order chromatin organization, including topologically associating domains (TADs) and compartments. Machine learning approaches can identify predictive features of H3K9me2 enrichment and build models that predict H3K9me2 patterns from DNA sequence or other epigenetic marks. Network analysis incorporating H3K9me2 data with transcription factor binding and gene expression can reveal regulatory networks influenced by this repressive mark. For evolutionary comparisons, synteny analysis of H3K9me2 domains across species can identify conserved repressive regions that may have fundamental regulatory functions.

How can I integrate H3K9me2 data with transcriptomic data to understand gene regulation?

Integrating H3K9me2 ChIP-seq data with transcriptomic data provides comprehensive insights into gene regulatory mechanisms. Begin with correlation analysis by calculating H3K9me2 enrichment in gene bodies or promoter regions and correlating these values with gene expression levels from RNA-seq data—typically revealing negative correlations as H3K9me2 is associated with gene repression. Generate metagene profiles displaying average H3K9me2 distribution across scaled gene bodies, grouped by expression levels (high, medium, low, silent) to visualize relationships between H3K9me2 patterns and transcriptional output. For differential analysis, identify genes that show significant changes in both H3K9me2 enrichment and expression between conditions using tools like diffBind combined with DESeq2 or edgeR. Time-series integration analyzing temporal relationships between H3K9me2 changes and expression changes can reveal whether H3K9me2 deposition precedes, coincides with, or follows transcriptional silencing. Pathway and gene ontology enrichment analysis of genes marked by H3K9me2 can identify biological processes preferentially regulated by this modification. Network analysis incorporating transcription factor binding data alongside H3K9me2 and expression data can identify regulatory circuits involving repression through H3K9me2. Single-cell integration, although technically challenging, can reveal heterogeneity in H3K9me2-mediated repression across individual cells by correlating H3K9me2 patterns with single-cell transcriptomic data. Machine learning approaches can be applied to build predictive models of gene expression based on H3K9me2 enrichment patterns along with other epigenetic features. For visualizing integrated data, develop genome browser tracks showing H3K9me2 enrichment alongside RNA-seq coverage, enabling exploration of specific loci of interest in their regulatory context.

What role does aberrant H3K9me2 play in disease pathogenesis?

Aberrant H3K9me2 patterns have been implicated in numerous disease states, reflecting the critical role of this modification in maintaining proper gene regulation and genome stability. In cancer, global alterations in H3K9me2 distribution are common, with some cancers showing genome-wide loss of H3K9me2, leading to inappropriate activation of normally silenced genes, including cancer-testis antigens and endogenous retroviruses. Conversely, other cancers display abnormal gains of H3K9me2 at tumor suppressor genes, contributing to their silencing. Mutations in G9a/GLP methyltransferases responsible for H3K9me2 deposition have been identified in various malignancies, and overexpression of these enzymes correlates with poor prognosis in several cancer types. In neurodegenerative disorders, dysregulation of H3K9me2 has been observed in Alzheimer's, Huntington's, and Parkinson's diseases, potentially contributing to altered gene expression patterns in neurons. Studies have shown that environmental stressors can induce persistent changes in H3K9me2 patterns, potentially linking environmental exposures to disease risk through epigenetic mechanisms. In cardiovascular diseases, aberrant H3K9me2 patterns have been observed in vascular smooth muscle cells and cardiomyocytes under pathological conditions, contributing to inappropriate gene expression. Autoimmune conditions exhibit alterations in H3K9me2 distribution in immune cells, potentially contributing to aberrant immune responses. Developmental disorders can arise from mutations in the H3K9 methylation machinery, leading to widespread disruption of normal gene silencing patterns during development. Understanding these disease-associated alterations provides insights into pathogenic mechanisms and identifies potential targets for therapeutic intervention.

How can Di-methyl-HIST1H3A (K9) antibodies be used in clinical and diagnostic applications?

Di-methyl-HIST1H3A (K9) antibodies have emerging applications in clinical and diagnostic settings, particularly in cancer and other epigenetic disorders. For cancer diagnostics, immunohistochemistry using H3K9me2 antibodies can identify altered patterns in tumor tissues compared to adjacent normal tissues, potentially serving as prognostic or diagnostic markers. Combination panels including H3K9me2 alongside other histone modifications can improve diagnostic accuracy and provide insights into tumor subtypes. Liquid biopsy approaches analyzing circulating nucleosomes with specific H3K9me2 patterns may offer minimally invasive screening options. In pharmacodynamic monitoring, H3K9me2 antibodies can assess the effectiveness of epigenetic therapies targeting histone methyltransferases or demethylases by measuring changes in global or locus-specific H3K9me2 levels. For patient stratification, H3K9me2 patterns may help identify subgroups of patients more likely to respond to specific treatments, particularly epigenetic therapies. Multiplex imaging approaches combining H3K9me2 antibodies with other epigenetic and cellular markers can provide spatially resolved information about tumor heterogeneity and microenvironment interactions. In neurological disorders, cerebrospinal fluid analysis of nucleosomes with specific H3K9me2 patterns may serve as biomarkers for conditions like Alzheimer's disease. Development of highly specific monoclonal and recombinant antibodies, like those described in search results , provides the precision required for clinical applications. Standardization efforts for antibody validation, including rigorous testing for specificity and reproducibility, are essential for clinical implementation. As these applications advance toward clinical use, appropriate quality control measures, reference standards, and procedural standardization will be crucial for reliable and reproducible results in diagnostic settings.

What are the current approaches for targeting H3K9 methylation therapeutically?

Therapeutic targeting of H3K9 methylation represents an emerging strategy in epigenetic medicine, with several approaches under investigation. Small molecule inhibitors of H3K9 methyltransferases (particularly G9a/GLP) have been developed, including BIX-01294, UNC0638, and A-366. These compounds competitively bind to the substrate-binding pocket or cofactor-binding site, preventing methyltransferase activity and reducing H3K9me2 levels. Several of these inhibitors have shown promising results in preclinical cancer models by reactivating silenced tumor suppressor genes. For enhanced specificity, peptide-based inhibitors designed to mimic histone tails and competitively inhibit methyltransferase activity offer potentially greater selectivity than small molecules. Targeted protein degradation approaches using PROTACs (Proteolysis Targeting Chimeras) that induce proteasomal degradation of H3K9 methyltransferases represent a newer strategy with potential advantages in efficacy and specificity. Gene therapy approaches using CRISPR-based epigenome editing can target demethylases to specific genomic loci to remove H3K9me2 marks selectively. Combination therapies pairing H3K9 methyltransferase inhibitors with other epigenetic drugs (HDAC inhibitors, DNA methyltransferase inhibitors) have shown synergistic effects in reactivating silenced genes in cancer models. RNA interference or antisense oligonucleotides targeting G9a/GLP mRNA can reduce expression of these methyltransferases, thereby decreasing H3K9me2 levels. Screens for synthetic lethal interactions with H3K9 methyltransferase inhibition have identified potential combination strategies that selectively target cancer cells while sparing normal tissues. As these therapeutic approaches advance, the development of biomarkers for patient selection and response monitoring will be crucial, with H3K9me2-specific antibodies playing a central role in these assessments.

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.