Mono-methyl-HIST1H3A (K27) Antibody

Shipped with Ice Packs
In Stock

Description

Target Specificity

  • Recognizes H3K27me1, a mono-methylated histone H3 variant encoded by the HIST1H3A gene.

  • Does not cross-react with di- or trimethylated H3K27 (H3K27me2/me3) or unmodified H3K27 .

Western Blot Analysis

  • Detects a band at ~15–17 kDa corresponding to mono-methylated histone H3 in nuclear extracts .

  • Validated in human HeLa cells and mouse tissues .

Chromatin Immunoprecipitation (ChIP)

  • Identifies H3K27me1-enriched genomic regions, such as promoters of actively transcribed genes .

  • Example: ChIP-qPCR in HeLa cells showed enrichment at the GAPDH promoter .

Immunofluorescence (IF)

  • Localizes H3K27me1 to euchromatic regions in nuclei of human osteosarcoma (U2OS) cells .

  • Specificity confirmed by blocking experiments with methylated peptides .

Dot Blot Validation

  • Distinguishes H3K27me1 from other methylation states (e.g., 0.2–100 pmol peptide sensitivity) .

Selectivity Assays

  • Peptide Competition: Pre-incubation with H3K27me1 peptides abolishes signal, while di-/trimethylated peptides do not .

  • Species Cross-Reactivity: Confirmed in human, mouse, rat, and monkey samples .

Key Experimental Results

Study DesignFindingsSource
ChIP-seq in HeLa cellsH3K27me1 associated with active enhancers and transcription start sites (TSS)
IF in U2OS cellsNuclear staining reduced upon siRNA-mediated depletion of H3K27 methyltransferases
WB in mouse embryonic cellsH3K27me1 levels increased during differentiation, contrasting with H3K27me3 repression

Transcriptional Activation

  • H3K27me1 marks active enhancers and gene bodies, facilitating RNA polymerase II recruitment .

  • In Drosophila, H3K27me1 is broadly distributed in euchromatin, while H3K27me3 is restricted to Polycomb-repressed loci .

Disease Relevance

  • Dysregulation linked to cancers (e.g., ovarian, leukemia) and developmental disorders .

  • Example: Elevated H3K27me1 in polycystic ovary syndrome (PCOS) correlates with aberrant gene expression .

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 of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
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 are fundamental structures responsible for wrapping and compacting DNA into chromatin. This process limits DNA accessibility to cellular machinery that requires DNA as a template, including transcription, repair, replication, and chromosomal stability. Histones play a central role in regulating these processes. DNA accessibility is meticulously controlled through a complex system of post-translational modifications of histones, often referred to as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests a mechanism for epigenetic regulation in cancer involving 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 can be valuable for determining whether tumors are heterochronous. PMID: 29482987
  3. Studies demonstrate 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. Data indicates that the Ki-67 antigen proliferative index has significant limitations, making phosphohistone H3 (PHH3) an alternative marker for proliferation. PMID: 29040195
  5. These findings suggest that cytokine-induced histone 3 lysine 27 trimethylation is a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. Research indicates that in the early developing human brain, HIST1H3B comprises the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This series of 47 diffuse midline gliomas reveals that the histone H3-K27M mutation is mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurs with BRAF-V600E mutation, and is frequently associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Data demonstrate that 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 show that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl), functioning 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 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). This mutation is correlated with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data suggest 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 immediately after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Research indicates 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 could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. These 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. Studies examine 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 is highlighted. PMID: 27996159
  21. hTERT promoter mutations are prevalent in medulloblastoma and are associated with older patients, prone to recurrence, and located in the right cerebellar hemisphere. Histone 3 mutations, however, 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 efficiently control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggests 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 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. The functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK is demonstrated. PMID: 25961932
  27. The authors confirm that histone H3 is a genuine substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. Circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Double mutations on the residues in the interface (L325A/D328A) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. MCM2 binding is not required for the incorporation of histone H3.1-H4 into chromatin but is essential for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a critical 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. Lower-resolution mass spectrometry instruments can be effectively used for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevents IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. De novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are 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 the specificity of Mono-methyl-HIST1H3A (K27) Antibody?

The Mono-methyl-HIST1H3A (K27) antibody is highly specific for histone H3 proteins that are mono-methylated at the lysine 27 position. According to product specifications, these antibodies detect endogenous levels of histone H3 only when mono-methylated at Lys27 (K27me1) . The antibodies are typically raised against synthetic peptides derived from within residues 1-100 of human histone H3, specifically containing the mono-methylated K27 modification . Specificity is a critical parameter for histone modification antibodies as cross-reactivity with other methylation states (di- or tri-methylation) or other lysine residues can lead to misinterpretation of results. Most manufacturers validate specificity through peptide array analysis, dot blot tests with modified and unmodified peptides, and Western blot validation with appropriate controls . For research requiring absolute specificity confirmation, it is advisable to conduct peptide competition assays where the antibody is pre-incubated with the mono-methylated peptide before application in your experimental system.

What applications is the Mono-methyl-HIST1H3A (K27) Antibody suitable for?

Mono-methyl-HIST1H3A (K27) antibodies have been validated for multiple applications in epigenetic research. Based on the product information, these antibodies are suitable for Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF), immunoprecipitation (IP), chromatin immunoprecipitation (ChIP), and ChIP sequencing (ChIPseq) . For Western blotting applications, the recommended dilution is typically around 1:1000, though this may vary between manufacturers and should be optimized for specific experimental conditions . In ChIP applications, these antibodies are particularly valuable for mapping the genomic distribution of H3K27me1, which is often associated with active enhancers and regions poised for transcriptional activation. For immunohistochemistry and immunofluorescence, these antibodies enable visualization of the nuclear distribution patterns of H3K27me1 in tissue sections or cultured cells. When using these antibodies for ChIP-seq, special attention should be paid to optimizing chromatin fragmentation, antibody concentration, and wash conditions to ensure high signal-to-noise ratios in the resulting data.

How should Mono-methyl-HIST1H3A (K27) Antibody be stored and handled?

Proper storage and handling of Mono-methyl-HIST1H3A (K27) antibodies are essential to maintain their specificity and sensitivity over time. According to product specifications, these antibodies are typically supplied as a liquid formulation in PBS with 0.1% sodium azide and 50% glycerol . The recommended storage temperature is -20°C, where the antibody remains stable for at least 12 months from the date of receipt . Repeated freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of antibody activity. If frequent use is anticipated, it is advisable to prepare working aliquots upon receipt and store the main stock at -20°C. When handling the antibody, always use clean pipette tips and sterile tubes to prevent contamination. Prior to use, the antibody solution should be gently mixed by inverting or flicking the tube rather than vortexing, which can lead to antibody denaturation. For long-term storage beyond the recommended shelf life, stability should be verified by functional testing before use in critical experiments.

What is the biological significance of H3K27 mono-methylation in epigenetic regulation?

H3K27 mono-methylation (H3K27me1) plays a distinct role in epigenetic regulation compared to its di- and tri-methylated counterparts. While H3K27 tri-methylation (H3K27me3) is associated with gene silencing and is deposited by the Polycomb Repressive Complex 2 (PRC2), H3K27me1 has been linked to active enhancers and is often found in euchromatic regions . Genome-wide studies have shown that H3K27me1 is enriched at the promoters of actively transcribed genes and distal enhancer elements, suggesting its role in maintaining an open chromatin state conducive to transcriptional activation. This modification is dynamically regulated by methyltransferases (such as G9a and GLP) and demethylases (including KDM7 family members), allowing for responsive changes to cellular signaling and developmental cues. The distinct genomic distribution patterns of H3K27me1 compared to H3K27me3 highlight the importance of studying specific methylation states rather than general modification sites. In developmental contexts, the transition between different methylation states at H3K27 has been implicated in cell fate decisions, lineage commitment, and cellular differentiation processes.

How can I optimize ChIP-seq protocols using Mono-methyl-HIST1H3A (K27) Antibody?

Optimizing ChIP-seq protocols for H3K27me1 analysis requires careful consideration of several parameters to maximize signal-to-noise ratio and data reproducibility. First, chromatin fragmentation should be optimized to generate fragments of 200-300 bp, which provides adequate resolution for H3K27me1 mapping without compromising antibody accessibility to the epitope . Sonication conditions should be empirically determined for each cell type, as chromatin compaction can vary significantly. Second, antibody concentration is critical; typically, 2-5 μg of Mono-methyl-HIST1H3A (K27) antibody per ChIP reaction is recommended, but titration experiments should be performed to determine the optimal amount for your specific experimental system . Third, implement a staged washing protocol with increasing stringency to remove non-specific binding while preserving specific interactions. Fourth, include appropriate controls such as input DNA, IgG control, and spike-in normalization standards if comparing across different conditions . For ChIP-seq library preparation, minimize PCR cycles to reduce amplification bias and consider using unique molecular identifiers (UMIs) to control for PCR duplicates. Finally, perform technical replicates and assess reproducibility through correlation analysis between replicates before proceeding to full-scale experiments.

What approaches can resolve cross-reactivity issues with other histone modifications?

Cross-reactivity between histone modification antibodies remains a significant challenge in epigenetic research, particularly for closely related modifications like mono-, di-, and tri-methylation at the same residue. Several approaches can help resolve such issues when working with Mono-methyl-HIST1H3A (K27) antibodies. First, conduct comprehensive validation using peptide arrays or dot blots with modified and unmodified peptides covering various methylation states (K27me1, K27me2, K27me3) and neighboring modified residues (e.g., K36me, K9me) . Second, perform peptide competition assays where the antibody is pre-incubated with excess competing peptides bearing different modifications before application in your experiment. Third, include appropriate biological controls such as cells with genetic knockouts of specific methyltransferases or demethylases that affect H3K27 methylation status. Fourth, consider orthogonal approaches such as mass spectrometry-based validation to quantitatively assess histone modification levels. Fifth, sequential ChIP (re-ChIP) can be employed to verify co-occurrence of modifications or distinguish between populations bearing different modifications. Additionally, emerging technologies like CUT&RUN or CUT&Tag may offer higher specificity and lower background compared to traditional ChIP for certain applications.

How can I quantitatively analyze H3K27me1 levels in different cell types or conditions?

Quantitative analysis of H3K27me1 levels across different experimental conditions requires standardized approaches to ensure reliable comparisons. For Western blot analysis, implement normalization to total histone H3 levels using a modification-independent H3 antibody on the same membrane after stripping or on a parallel blot . Densitometric analysis should be performed within the linear range of detection, and multiple biological replicates are essential for statistical validity. For immunofluorescence quantification, standardize image acquisition parameters including exposure time, gain, and offset, and analyze nuclear intensity using automated image analysis software that can segment nuclei and measure mean fluorescence intensity . For ChIP-qPCR, normalize to input DNA and consider using a reference gene region known to maintain stable H3K27me1 levels across your experimental conditions. For ChIP-seq analysis, implement appropriate normalization methods such as TMM (trimmed mean of M-values) or spike-in normalization with foreign DNA to account for global changes in modification levels . Advanced mass spectrometry-based approaches provide absolute quantification of histone modifications and can distinguish between different histone variants bearing the same modification. When comparing pathological versus normal tissues, batch effects should be carefully controlled, and appropriate statistical methods should be applied to account for heterogeneity within samples.

What are the best controls to include when using this antibody for chromatin studies?

Rigorous control strategies are essential for reliable interpretation of chromatin studies using Mono-methyl-HIST1H3A (K27) antibodies. First, include a technical negative control such as non-specific IgG from the same species as the primary antibody (rabbit) to assess background binding levels . Second, incorporate biological negative controls such as chromatin from cells where H3K27 mono-methylation has been depleted through genetic manipulation (e.g., knockout/knockdown of relevant methyltransferases like G9a) or pharmacological inhibition. Third, use positive controls such as genomic regions known to be enriched for H3K27me1 based on published datasets for your cell type. Fourth, implement peptide competition controls where the primary antibody is pre-incubated with excess H3K27me1 peptide to demonstrate binding specificity . Fifth, include input samples representing the starting chromatin material prior to immunoprecipitation for normalization purposes. For sequential ChIP experiments, additional controls should include single-antibody ChIPs performed in parallel. When integrating ChIP-seq data with other genomic datasets, include controls that account for technical biases such as chromatin accessibility, GC content variation, and mappability issues. Finally, biological replicates serve as essential controls to ensure reproducibility and distinguish biological variation from technical noise.

How can I resolve inconsistent Western blot results with Mono-methyl-HIST1H3A (K27) Antibody?

Inconsistent Western blot results when using Mono-methyl-HIST1H3A (K27) antibodies can stem from various technical and biological factors. First, ensure complete histone extraction by using specialized histone extraction protocols that involve acid extraction (typically with 0.2N HCl) to efficiently isolate histones from chromatin . Second, standard protein quantification methods like Bradford assay may not accurately measure histone concentrations; consider using alternative methods such as SDS-PAGE followed by Coomassie staining for normalization. Third, histone modifications can be unstable during sample preparation; include deacetylase inhibitors (e.g., sodium butyrate), phosphatase inhibitors, and protease inhibitors in lysis buffers to preserve modification status. Fourth, optimize transfer conditions specifically for low-molecular-weight proteins (~17 kDa for histones), using PVDF membranes rather than nitrocellulose and adjusting transfer time and voltage accordingly . Fifth, blocking solutions containing BSA may be preferable to milk-based blockers, as the latter can contain enzymes that affect histone modifications. If batch-to-batch antibody variability is suspected, validate each new lot against a reference sample. Additionally, ensure consistent cell culture conditions, as histone modifications can be affected by cell density, passage number, and culture media composition.

What might cause false positive or negative results in ChIP experiments?

False positive and negative results in ChIP experiments using Mono-methyl-HIST1H3A (K27) antibodies can arise from multiple sources that must be carefully controlled. False positives commonly result from insufficient washing stringency, antibody cross-reactivity with similar modifications (H3K27me2/3), or non-specific binding to highly transcribed regions due to increased chromatin accessibility rather than specific H3K27me1 enrichment . Implementing more stringent wash conditions, validating antibody specificity with peptide competition assays, and normalizing to IgG control and input can help mitigate these issues. Conversely, false negatives may occur due to epitope masking by adjacent modifications or protein binding, insufficient chromatin fragmentation limiting antibody accessibility, or suboptimal crosslinking conditions . To address these challenges, optimize crosslinking time and conditions (typically 1% formaldehyde for 10 minutes), ensure adequate chromatin fragmentation (200-300 bp average size), and consider native ChIP approaches for certain applications where crosslinking might interfere with epitope recognition. Additionally, the timing of cell harvest can significantly impact H3K27me1 levels, as this modification is dynamically regulated through the cell cycle. When designing primers for ChIP-qPCR validation, avoid regions with extreme GC content or repetitive elements that may affect PCR efficiency and specificity.

How can I distinguish between mono-, di-, and tri-methylation signals at H3K27?

Distinguishing between the different methylation states at H3K27 requires careful experimental design and validation strategies. First, use antibodies with demonstrated specificity for H3K27me1, H3K27me2, or H3K27me3, validated through peptide arrays or dot blots with all methylation states . Second, include control cell lines or treatments that alter the distribution of methylation states, such as EZH2 inhibitors (which reduce H3K27me3 levels) or G9a/GLP inhibitors (affecting H3K27me1/me2). Third, parallel ChIP-seq experiments with antibodies against each methylation state can provide genome-wide comparative maps showing distinct distribution patterns—H3K27me3 typically associates with repressed genes and Polycomb targets, H3K27me2 with intergenic regions, and H3K27me1 with active enhancers and gene bodies. Fourth, sequential ChIP (re-ChIP) can determine whether different methylation states co-occur on the same nucleosomes or represent distinct populations. Fifth, mass spectrometry-based histone analysis provides quantitative measurement of each methylation state's abundance and can detect combinatorial modifications on the same histone tail. For locus-specific analysis, calibrated ChIP-qPCR using spike-in controls allows direct comparison of enrichment levels across methylation states. When interpreting results, consider that these methylation states exist in dynamic equilibrium regulated by writers (PRC2, G9a/GLP) and erasers (KDM6A/B, KDM7 family), with their relative abundance changing during cellular processes like differentiation or disease progression.

How does H3K27me1 distribution compare with other histone marks in genome-wide studies?

Genome-wide analysis of H3K27me1 distribution reveals distinct patterns compared to other histone modifications, providing insights into its functional roles. H3K27me1 shows a characteristic enrichment at active enhancers and gene bodies of actively transcribed genes, with a profile that partially overlaps with H3K4me1 but is distinct from H3K4me3 (concentrated at active promoters) and H3K27me3 (associated with repressed chromatin) . Integrative analysis of multiple ChIP-seq datasets demonstrates that H3K27me1 often co-occurs with H3K36me3 in gene bodies, suggesting a role in transcriptional elongation. Unlike the sharp peaks observed for H3K4me3 at promoters, H3K27me1 typically shows broader enrichment patterns across regulatory elements and transcribed regions. When analyzing the relationship between chromatin states and DNA accessibility, H3K27me1-enriched regions generally exhibit intermediate accessibility as measured by ATAC-seq or DNase-seq, positioning between highly accessible H3K4me3-marked promoters and inaccessible H3K27me3-marked repressed regions. Temporal analysis during cellular differentiation reveals that H3K27me1 deposition often precedes gene activation and may serve as a priming mark for future transcriptional changes. Computational approaches like chromatin state segmentation (e.g., ChromHMM or Segway) can integrate multiple histone modification datasets to define functional chromatin states, with H3K27me1 contributing to the identification of specific enhancer and transcribed region states.

What is the relationship between H3K27me1 and gene expression in different cellular contexts?

The relationship between H3K27me1 and gene expression exhibits context-dependent patterns across different cell types and physiological states. Correlation analysis of H3K27me1 ChIP-seq data with RNA-seq expression profiles generally shows a positive association between gene body H3K27me1 enrichment and transcriptional activity, particularly for housekeeping genes and tissue-specific genes active in the analyzed cell type . This contrasts with H3K27me3, which strongly anti-correlates with expression. In embryonic stem cells, the transition from H3K27me3 to H3K27me1 at developmental gene loci often accompanies lineage commitment, with H3K27me1 serving as an intermediate state during enhancer activation. The enzyme G9a (EHMT2) contributes to H3K27me1 deposition and has been linked to both gene activation and repression, depending on genomic context and interacting partners. Meta-analysis of enhancer elements classified by activity level reveals that H3K27me1 enrichment correlates with enhancer strength, often co-occurring with H3K4me1 and moderate levels of H3K27ac at active enhancers. During cellular stress responses, dynamic changes in H3K27me1 distribution can precede transcriptional reprogramming, suggesting a role in adaptation to environmental cues. Single-cell epigenomic profiling techniques have begun to reveal cell-to-cell variation in H3K27me1 patterns that correlate with expression heterogeneity within seemingly homogeneous populations. In cancer cells, altered H3K27me1 distribution has been associated with oncogene activation and tumor suppressor silencing, making it a potential biomarker for certain malignancies.

How can I integrate H3K27me1 ChIP-seq data with other epigenomic datasets?

Integrating H3K27me1 ChIP-seq data with other epigenomic datasets requires systematic computational approaches to extract meaningful biological insights. First, implement standardized preprocessing workflows including quality control, alignment to reference genome, peak calling, and signal normalization to ensure comparability across datasets . Next, utilize genome browsers like IGV or UCSC to visualize H3K27me1 patterns alongside other histone modifications, transcription factor binding sites, chromatin accessibility, and gene expression data for qualitative assessment of relationships. For quantitative integration, correlation analysis can identify histone marks or factors that co-occur with H3K27me1 across the genome, while differential binding analysis can highlight regions where H3K27me1 levels change independently of other marks. Chromatin state segmentation algorithms such as ChromHMM or Segway enable the definition of functional chromatin states based on combinatorial patterns of multiple histone modifications including H3K27me1. Network analysis approaches can connect H3K27me1-marked regulatory elements to target genes using chromatin interaction data from Hi-C or ChIA-PET experiments. To associate H3K27me1 patterns with gene expression, implement regression models that integrate multiple epigenetic features as predictors of transcriptional output. For temporal studies, trajectory analysis methods can track the dynamics of H3K27me1 and other epigenetic marks during processes like differentiation or disease progression. Finally, pathway enrichment analysis of genes associated with H3K27me1-marked regulatory elements can provide functional interpretation of the biological processes influenced by this epigenetic modification.

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.