Mono/Di/Tri-methyl-Histone H3.1 (K14) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Definition and Purpose

This recombinant monoclonal antibody targets methylated lysine 14 residues on histone H3.1, a core component of nucleosomes. Histone H3 methylation at K14 influences DNA accessibility, transcriptional activity, and chromatin remodeling . The antibody is engineered to recognize all three methylation states (mono-, di-, and tri-) at this site, making it valuable for studying context-dependent epigenetic regulation .

Development and Production

The antibody is generated through a multi-step process:

  • Gene Cloning: Genes encoding heavy and light chains of the HIST1H3A antibody are cloned into expression vectors .

  • Host Cell Transfection: Vectors are transfected into host cells for antibody production and secretion .

  • Purification: Affinity chromatography ensures high purity (>95%) .

  • Validation: Functionality is confirmed via ELISA, Western blot (WB), immunofluorescence (IF), and peptide array .

Western Blot Performance

Sample TypeObserved Bands (kDa)Predicted Bands (kDa)Dilution
NIH/3T3 (Mouse)15, 25, 13015, 25, 1021:1000
HeLa (Human)15, 25, 13015, 25, 1021:1000

Data from , using peroxidase-conjugated secondary antibodies.

Immunofluorescence

  • Nuclear Localization: Confocal imaging in HeLa cells shows strong nuclear staining (green) with DAPI counterstain (blue) .

  • Controls: Specificity confirmed via negative controls using mismatched primary/secondary antibody pairs .

Dot Blot Specificity

PeptideSignal Intensity (Relative to Unmodified)
H3K14me1++++
H3K14me2+++
H3K14me3++
Unmodified H3K14-

Data from , validated using peptide arrays and 5% BSA/TBST blocking buffer.

Biological Relevance of HK14 Methylation

  • Mono-methylation (K14me1): Linked to transcriptional activation or repression, depending on chromatin context .

  • Di-methylation (K14me2): Associated with developmental gene regulation and cell cycle control .

  • Tri-methylation (K14me3): Implicated in genome stability and cellular identity maintenance .

Limitations and Considerations

  • Species Restrictions: Validated only for human and mouse samples .

  • Buffer Composition: Contains sodium azide, requiring careful handling .

  • Band Discrepancies: Observed vs. predicted molecular weights may vary due to post-translational modifications .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The Mono/Di/Tri-methyl-Histone H3.1 (K14) Recombinant Antibody is produced through a robust process that begins with cloning the genes encoding the HIST1H3A antibody, encompassing both heavy and light chains. These cloned genes are then integrated into expression vectors, which are subsequently introduced into host cells using transfection. The host cells are responsible for the production and secretion of the antibody. Following purification using affinity chromatography to ensure its purity, the antibody undergoes rigorous functionality testing through ELISA, ensuring its ability to accurately detect the human HIST1H3A protein mono/di/tri-methylated at K14.

Mono-methylation of H3.1 at K14 is associated with both transcriptional activation and repression, depending on the cellular context. Di- or tri-methylation at H3.1 K14 is often linked to transcriptional repression. Di-methylation at H3.1 K14 can participate in the regulation of genes involved in development, differentiation, and cell cycle control. Tri-methylation at H3.1 K14 plays a crucial role in regulating genes essential for cellular identity, development, and genome stability.

Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors 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 H3.1 is a core component of the nucleosome, the fundamental unit of chromatin. Nucleosomes wrap and compact DNA, limiting its accessibility to cellular machinery that requires DNA as a template. As a result, histones play a central role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is meticulously controlled through a complex array of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.

Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer may be mediated by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The detection of increased expression of H3K27me3 during a patient's clinical course can be a valuable tool for determining whether tumors are heterochronous. PMID: 29482987
  3. Recent studies have shown 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. Evidence suggests that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) presents an alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data reveals 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 demonstrates that the histone H3-K27M mutation is mutually exclusive with the IDH1-R132H mutation and EGFR amplification, rarely co-occurs with the BRAF-V600E mutation, and is commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Notably, among these K27M+ diffuse midline gliomas... PMID: 26517431
  8. Data suggests that the histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral, and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments have demonstrated 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 correlated with poor outcome 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. Research shows 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. Data suggests that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. 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 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. In contrast, 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 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 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 Raji cells treated with 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) decrease 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 the incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Data suggests 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 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 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 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 biological significance of H3.1 K14 methylation?

Histone H3.1 K14 methylation serves as a key epigenetic mark that regulates chromatin structure and gene expression. The biological significance varies by methylation state: mono-methylation at K14 is associated with both transcriptional activation and repression depending on cellular context; di-methylation regulates genes involved in development, differentiation, and cell cycle control; while tri-methylation primarily governs genes essential for cellular identity, development, and genome stability . These modifications contribute to the "histone code" that controls DNA accessibility to transcriptional machinery, thereby playing a central role in transcription regulation, DNA repair, replication, and chromosomal stability .

How does H3.1 K14 methylation differ from other histone H3 modifications?

H3.1 K14 methylation is distinctive in its variable turnover rate compared to other histone modifications. While some histone marks like H3K4me3 exhibit rapid turnover (half-life of approximately 6.8 hours), methylation at K14 demonstrates intermediate stability . Furthermore, K14 methylation often works in combination with adjacent modifications, particularly acetylation at K9, to create specific chromatin environments. The positioning of K14 within the histone tail makes it strategically important for interactions with chromatin-modifying complexes and transcription factors, distinguishing it from modifications at other lysine residues that may serve different regulatory functions .

What are the distinct characteristics of mono-, di-, and tri-methylation at H3.1 K14?

Each methylation state at H3.1 K14 serves unique functions in chromatin regulation:

Methylation StatePrimary FunctionsAssociated ProcessesTypical Chromatin Regions
Mono-methylationContext-dependent activation or repressionGene poising, transcriptional memoryEnhancers, promoter regions
Di-methylationPrimarily repressiveDevelopment, differentiation, cell cycle controlGene bodies, facultative heterochromatin
Tri-methylationStrong repressionCellular identity maintenance, genome stabilityConstitutive heterochromatin

This functional divergence makes antibodies that can distinguish between these methylation states particularly valuable for understanding the complex regulatory mechanisms governing gene expression .

What criteria should be considered when selecting a Mono/Di/Tri-methyl-Histone H3.1 (K14) antibody for specific applications?

When selecting an antibody for H3.1 K14 methylation studies, researchers should consider several critical factors. First, determine whether the experimental question requires discrimination between methylation states or detection of all states collectively. For methylation-state-specific detection, choose antibodies validated for specificity against mono-, di-, or tri-methylation exclusively. For applications requiring detection of all methylation states, pan-methyl antibodies like the recombinant monoclonal H3 mono+di+tri methyl K14 antibody are appropriate .

Second, consider the intended application: for Western blotting, prioritize antibodies validated for strong band specificity at the expected 17 kDa size ; for immunofluorescence, select antibodies with demonstrated nuclear localization and minimal background. Recombinant monoclonal antibodies generally offer higher reproducibility across experiments than polyclonal alternatives. Finally, cross-reactivity with other histone modifications should be thoroughly assessed, particularly between K14 methylation and nearby modifications such as K9 acetylation, which can affect epitope recognition .

How can specificity of a Mono/Di/Tri-methyl-Histone H3.1 (K14) antibody be validated?

Validating antibody specificity for H3.1 K14 methylation requires a multi-faceted approach:

  • Peptide competition assays: Compare antibody binding in the presence and absence of competing synthetic peptides containing specific methylation states of K14. A significant reduction in signal when the target-specific peptide is present confirms specificity .

  • Methyltransferase/demethylase manipulations: Conduct experiments with knockdown or overexpression of enzymes that regulate K14 methylation. Changes in antibody signal corresponding to expected methylation changes support specificity.

  • Histone peptide arrays: Utilize arrays containing various histone modifications to identify potential cross-reactivity with other histone modifications, particularly those at adjacent residues like K9 .

  • Mass spectrometry correlation: Compare antibody-based detection with mass spectrometry quantification of modification abundance across different conditions to verify that the antibody accurately reflects true biological changes .

  • Knockout controls: Where possible, use cells with genetic manipulation of histone genes to create modification-free controls that demonstrate antibody specificity.

Documentation of these validation steps provides confidence in experimental results and enables accurate interpretation of biological phenomena .

What are the optimal conditions for using Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies in Western blotting?

For optimal Western blotting with Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies, careful attention to protocol details is essential. Begin with proper sample preparation by extracting histones using acid extraction methods to enrich for basic proteins. For cell lysates, use RIPA buffer supplemented with protease inhibitors, deacetylase inhibitors (such as sodium butyrate), and demethylase inhibitors to preserve modification states.

When running gels, use 15-18% polyacrylamide to achieve good separation of the low molecular weight (approximately 17 kDa) histone proteins . After transfer to membranes (PVDF preferred for histones), block with 5% BSA in TBST rather than milk, as milk contains proteins that can cross-react with some histone antibodies .

For primary antibody incubation, dilute the Mono/Di/Tri-methyl-Histone H3.1 (K14) antibody to 1 μg/mL in blocking buffer and incubate overnight at 4°C for optimal signal-to-noise ratio . Include appropriate controls such as recombinant histones with defined modification states or peptide competition assays to confirm specificity of detected bands. The expected band size for H3 is approximately 17 kDa, with potential additional bands at 37 kDa representing histone dimers that have not been fully reduced .

How can Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies be effectively used in immunofluorescence studies?

For successful immunofluorescence with Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies, sample preparation is critical. Begin with cell fixation using 4% paraformaldehyde for 10 minutes followed by permeabilization with 0.2% Triton X-100. For histone modifications, an additional antigen retrieval step may improve epitope accessibility: incubate fixed cells in 10 mM citrate buffer (pH 6.0) at 95°C for 10 minutes.

To minimize background and ensure specificity, block with 3% BSA in PBS for at least 1 hour at room temperature. Dilute the primary antibody (e.g., rabbit recombinant monoclonal H3 mono+di+tri methyl K14 antibody) according to validated ratios for immunocytochemistry/immunofluorescence (ICC/IF) . Overnight incubation at 4°C generally provides the best signal-to-noise ratio.

For visualization, use species-appropriate fluorophore-conjugated secondary antibodies and include DAPI staining to visualize nuclei. When interpreting results, H3.1 K14 methylation typically appears as punctate nuclear staining with patterns that vary depending on cell type and the specific methylation state being detected. Different methylation states may show distinct nuclear localization patterns, with tri-methylation often appearing in dense heterochromatic regions while mono-methylation might show broader distribution .

What approaches can be used to study the dynamics of H3.1 K14 methylation in living cells?

Studying H3.1 K14 methylation dynamics in living cells requires specialized approaches that go beyond fixed-sample antibody techniques. One powerful method involves stable isotope labeling with amino acids in cell culture (SILAC) combined with quantitative mass spectrometry. This approach allows tracking of methylation turnover by labeling both the histone (with heavy arginine) and methyl groups (with heavy methionine) .

For temporal resolution of methylation changes, researchers can employ:

  • Inducible enzyme systems: Generate cell lines with inducible expression of methyltransferases or demethylases that target K14, coupled with time-course sampling for antibody-based detection or mass spectrometry analysis.

  • FRAP (Fluorescence Recovery After Photobleaching): Using fluorescently tagged histone H3.1 constructs and live-cell imaging to monitor turnover rates of histones carrying specific modifications.

  • Targeted degradation approaches: Employing degron-tagged histone modifying enzymes that can be rapidly depleted to observe acute effects on K14 methylation status.

  • ChIP-seq time courses: Following stimulus application with sequential chromatin immunoprecipitation to map genome-wide changes in K14 methylation patterns over time.

These approaches have revealed that different histone modifications exhibit significantly different turnover rates, with H3K4me3 showing relatively rapid turnover (half-life of 6.8 hours) compared to more stable modifications like H3K9me3 and H3K27me3 (half-lives spanning multiple cell divisions) .

How can inconsistent signals in Western blotting with Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies be resolved?

Inconsistent Western blot signals when using Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies can stem from several sources. First, ensure proper histone extraction and preservation of modifications by using acid extraction methods and including deacetylase and demethylase inhibitors in all buffers. If signals are weak or variable, optimize antibody concentration through titration experiments (typically ranging from 0.5-2 μg/mL) .

For cross-reactivity issues, consider the following troubleshooting approaches:

ProblemPotential CausesSolution Approaches
Multiple unexpected bandsCross-reactivity with other histone modificationsPerform peptide competition assays with modified and unmodified peptides
Weak target bandModification level too low in sampleCompare with positive control samples known to have the modification
High backgroundNon-specific bindingIncrease blocking time, use 5% BSA instead of milk, optimize antibody dilution
Inconsistent results between experimentsLot-to-lot antibody variationSwitch to recombinant monoclonal antibodies for greater consistency
Signal at incorrect molecular weightSample degradation or aggregationUse fresh samples, ensure complete reduction of samples

Additionally, always include proper loading controls such as total H3 antibodies to normalize for variations in histone content between samples. When comparing different cell types or treatments, be aware that global levels of histone modifications can vary significantly, making relative quantification essential .

What factors might contribute to false positive or false negative results when using these antibodies?

Several factors can lead to false positive or false negative results when working with Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies:

False positives may result from:

  • Antibody cross-reactivity with similar epitopes, particularly adjacent modifications like K9 or K18 methylation or acetylation

  • Non-specific binding to other proteins with similar sequences or charge properties

  • Insufficient blocking leading to background signal interpreted as positive

  • Sample preparation methods that create artifacts or epitope modifications

  • Excessive antibody concentration leading to non-specific binding

False negatives may result from:

  • Epitope masking due to protein-protein interactions or adjacent modifications

  • Degradation of the methylation mark during sample preparation

  • Insufficient antigen retrieval in fixed tissues or cells

  • Antibody lot variability or degradation

  • Excessively stringent washing conditions removing specific binding

To minimize these issues, researchers should validate antibodies using multiple approaches, including peptide competition assays, western blotting combined with known positive and negative controls, and correlation with orthogonal techniques like mass spectrometry . Additionally, understanding the turnover dynamics of histone methylation is crucial—rapidly cycling modifications like H3K4me3 may be more susceptible to false negatives if samples are not processed quickly .

How should conflicting results between ChIP-seq and immunofluorescence data using the same antibody be interpreted?

When ChIP-seq and immunofluorescence results using the same Mono/Di/Tri-methyl-Histone H3.1 (K14) antibody conflict, systematic analysis is required to resolve discrepancies. These techniques operate at different scales and under different conditions, which can explain divergent outcomes:

  • Epitope accessibility differences: In ChIP-seq, chromatin is fragmented and partially denatured, potentially exposing epitopes that remain masked in the more native chromatin environment of immunofluorescence. Test whether different fixation or permeabilization methods for immunofluorescence alter the results.

  • Antibody concentration effects: ChIP typically uses higher antibody concentrations than immunofluorescence, which may reveal different epitope recognition profiles. Perform antibody titration experiments in both techniques to determine optimal concentrations.

  • Chromatin context sensitivity: Some antibodies recognize their target modification differently depending on adjacent modifications or DNA sequences. Check antibody behavior on peptide arrays containing combinatorial modifications .

  • Quantitative vs. qualitative interpretation: ChIP-seq provides genome-wide quantitative data, while immunofluorescence offers spatial information with less quantitative precision. A modification might be present at specific genomic loci (detected by ChIP-seq) but at levels too low for immunofluorescence detection.

  • Batch effects: Different antibody lots can show variation in specificity profiles. Whenever possible, use the same antibody lot for comparative analyses.

To resolve conflicting results, consider orthogonal approaches such as targeted mass spectrometry of specific genomic regions, alternative antibodies recognizing the same modification, or genetic manipulation of the enzymes responsible for establishing or removing the modification .

How can Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies be integrated into multiplexed epigenetic profiling pipelines?

Integrating Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies into multiplexed epigenetic profiling requires strategic planning to maximize data quality and interpretability. Advanced multiplexing approaches include:

  • Sequential ChIP (re-ChIP): Performing consecutive immunoprecipitations with antibodies against different modifications, including H3.1 K14 methylation, to identify genomic regions containing specific combinations of marks. This approach reveals the co-occurrence of K14 methylation with other modifications like K9 acetylation .

  • Mass cytometry (CyTOF): Using metal-conjugated antibodies against various histone modifications for single-cell analysis of epigenetic states. This technique can incorporate antibodies against different methylation states of H3.1 K14 to classify cell populations based on their epigenetic profiles.

  • Combinatorial indexed ChIP-seq: Employing barcoding strategies to perform ChIP-seq for multiple histone modifications, including H3.1 K14 methylation states, across numerous samples simultaneously, dramatically increasing throughput.

  • Spatial epigenomics: Combining immunofluorescence detection of H3.1 K14 methylation with other histone marks using spectrally distinct fluorophores, followed by high-resolution imaging to map the spatial organization of different chromatin states within the nucleus.

  • Single-molecule approaches: Utilizing techniques like single-molecule FISH combined with immunofluorescence to correlate H3.1 K14 methylation states with transcriptional activity at specific genomic loci.

When designing multiplexed experiments, careful antibody panel design is essential to avoid spectral overlap in fluorescence-based detection or antibody cross-reactivity issues. Validation of each antibody individually and in the multiplexed context is critical before proceeding with large-scale experiments .

What computational approaches are recommended for analyzing ChIP-seq data generated using Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies?

Analyzing ChIP-seq data for H3.1 K14 methylation requires specialized computational approaches to account for the unique characteristics of this modification:

  • Peak calling optimization: Standard peak callers (MACS2, SICER) should be optimized specifically for the distribution pattern of H3.1 K14 methylation, which may differ between mono-, di-, and tri-methylation states. Tri-methylation typically shows sharper, more defined peaks associated with heterochromatin regions, while mono-methylation may display broader distributions .

  • Differential methylation analysis: Tools like DiffBind or MAnorm should be employed to identify regions with statistically significant changes in K14 methylation between conditions, with careful normalization to account for global changes in modification levels.

  • Integration with other epigenetic marks: Correlation analysis with datasets for other histone modifications helps place K14 methylation in context. Given the known interplay between H3.1 K14 methylation and acetylation at K9/K14, co-occurrence analysis is particularly informative .

  • Turnover rate correction: When comparing ChIP-seq data across time points, consider the differential turnover rates of histone modifications. K14 methylation may show intermediate turnover compared to rapidly cycling modifications like H3K4me3 (half-life ~6.8 hours) or very stable marks like H3K9me3 .

  • Machine learning approaches: Supervised classification algorithms can identify chromatin signatures incorporating H3.1 K14 methylation states that predict functional outcomes like transcriptional activity or replication timing.

  • Motif analysis: Identify DNA sequence motifs enriched in regions marked by specific K14 methylation states to uncover potential relationships with transcription factor binding sites or chromatin regulators.

For proper interpretation, researchers should also consider the differences in antibody performance between methylation states, as some antibodies may have different efficiencies for mono-, di-, or tri-methylation, potentially biasing comparative analyses .

How can researchers study the interplay between H3.1 K14 methylation and other histone modifications?

Investigating the interplay between H3.1 K14 methylation and other histone modifications requires multi-dimensional approaches:

  • Sequential ChIP (re-ChIP): This technique involves successive immunoprecipitations with antibodies against different modifications. First precipitate with anti-H3.1 K14 methylation antibodies, then subject the enriched material to a second round of ChIP using antibodies against other modifications of interest, such as H3K9ac or H3K4me3. This reveals genomic regions containing both modifications simultaneously .

  • Mass spectrometry of histone PTMs: Quantitative mass spectrometry can identify and quantify combinatorial histone modifications on the same histone tail. This approach has revealed that the turnover rates of different methylation marks vary significantly, with K4me3 having a half-life of approximately 6.8 hours, while K9 and K27 methylation are stable across multiple cell divisions .

  • Biochemical enzyme assays: In vitro assays can determine how existing modifications affect the activity of enzymes that establish or remove K14 methylation. For example, testing whether K9 acetylation enhances or inhibits the activity of K14 methyltransferases.

  • Genetic manipulation of modifying enzymes: By overexpressing or knocking down enzymes that establish specific modifications, researchers can observe the consequent effects on K14 methylation patterns. This approach helps establish causal relationships between modifications.

  • Structural biology approaches: Crystallography or cryo-EM studies of histone modifying complexes with differentially modified histone tail peptides can reveal how one modification physically influences the recognition of the K14 residue by methyltransferases or demethylases.

  • Combinatorial antibody profiling: Using antibodies that specifically recognize combinatorial modifications (e.g., dual modified histones with both K9ac and K14me2) can reveal the prevalence and distribution of specific modification combinations .

These approaches have revealed that different methylation states at K14 have distinct associations with other modifications: mono-methylation often co-occurs with active marks in euchromatin, while tri-methylation is frequently associated with repressive marks in heterochromatic regions .

How are Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies being applied in single-cell epigenomic studies?

Single-cell epigenomic studies represent a frontier in understanding cellular heterogeneity, and Mono/Di/Tri-methyl-Histone H3.1 (K14) antibodies are increasingly being incorporated into these approaches:

  • Single-cell CUT&Tag/CUT&RUN: These techniques adapt chromatin profiling for single-cell resolution, allowing researchers to map K14 methylation patterns across individual cells within heterogeneous populations. Antibody quality is particularly critical in these applications, with recombinant monoclonal antibodies generally providing better consistency than polyclonal alternatives .

  • scCHiC (single-cell Chromatin Immunocleavage): This technique combines chromatin immunoprecipitation with single-cell Hi-C to simultaneously map a histone modification and chromatin conformation. Application with H3.1 K14 methylation antibodies can reveal how this modification correlates with 3D genome organization at the single-cell level.

  • Imaging-based approaches: Mass cytometry (CyTOF) and multiplexed immunofluorescence allow quantification of multiple histone modifications, including H3.1 K14 methylation states, in thousands of individual cells. These approaches facilitate the identification of epigenetically distinct subpopulations within apparently homogeneous tissues.

  • Single-cell multi-omics: Emerging methods that combine chromatin accessibility, histone modification profiling, and transcriptomics in the same cell provide unprecedented insights into how H3.1 K14 methylation states directly correlate with gene expression patterns at single-cell resolution.

These single-cell approaches have revealed that H3.1 K14 methylation patterns can vary significantly between seemingly identical cells, contributing to cell-to-cell variability in gene expression and potentially influencing cell fate decisions in development and disease contexts .

What is known about the stability and turnover rates of different methylation states at H3.1 K14, and how does this impact experimental design?

Understanding the turnover dynamics of H3.1 K14 methylation is crucial for proper experimental design and data interpretation. Histone methylation turnover varies significantly by site and methylation state:

  • Differential stability by methylation state: Research using stable isotope labeling and quantitative mass spectrometry has demonstrated that histone methylation marks have varying half-lives. While H3K4me3 shows rapid turnover with a half-life of approximately 6.8 hours, other methylation marks like H3K9me3 and H3K27me3 are far more stable, with half-lives spanning multiple cell divisions . K14 methylation stability varies by methylation state, with tri-methylation generally being more stable than mono-methylation.

  • Impact on experimental timing: The differential stability of methylation states necessitates careful consideration of time points in experiments. For studying dynamic changes in K14 mono-methylation, sampling at shorter intervals (hours) may be necessary, while changes in tri-methylation may require longer timeframes (days) to observe significant changes.

  • Cell cycle effects: The dilution of existing histone modifications during DNA replication can confound interpretation of turnover data. Experiments measuring K14 methylation dynamics should account for cell cycle effects, potentially through cell synchronization or single-cell approaches that incorporate cell cycle markers .

  • Enzyme inhibitor studies: Inhibition of histone demethylases has revealed that different methylation sites have different sensitivities to enzymatic removal. K9 methylation shows larger increases upon demethylase inhibition compared to other sites, suggesting site-specific regulation of turnover rates .

  • Transcription-dependent turnover: The turnover of histone methylation is often coupled to transcriptional activity, with marks associated with active transcription (like H3K4me3) showing faster turnover. This relationship should be considered when studying K14 methylation in genes with different transcriptional states .

These dynamics have important implications for experimental design, particularly for time-course studies, drug treatments, or genetic perturbations targeting the enzymes responsible for establishing or removing K14 methylation .

What role does H3.1 K14 methylation play in disease contexts, and how are antibodies being used to investigate these connections?

H3.1 K14 methylation has emerging roles in various disease contexts, with antibody-based studies providing critical insights:

  • Cancer epigenetics: Altered patterns of H3.1 K14 methylation have been observed in multiple cancer types. Recombinant monoclonal antibodies against different methylation states are being used in ChIP-seq studies to map genome-wide changes in methylation patterns between normal and cancer tissues. These studies have revealed that the balance between different methylation states at K14 is often disrupted in cancer, with aberrant tri-methylation associated with inappropriate gene silencing .

  • Neurodegenerative disorders: In models of neurodegenerative diseases, changes in histone methylation turnover have been observed. Using antibodies against specific K14 methylation states in combination with mass spectrometry, researchers have identified altered methylation dynamics that may contribute to dysregulated gene expression in conditions like Alzheimer's and Huntington's disease .

  • Developmental disorders: Mutations in enzymes that regulate H3.1 K14 methylation have been implicated in developmental disorders. Immunofluorescence studies using methylation-specific antibodies in patient-derived cells or model organisms have helped characterize the epigenetic consequences of these mutations on chromatin organization and gene expression .

  • Inflammatory conditions: Changes in histone methylation, including at K14, have been linked to inflammatory gene regulation. Antibody-based ChIP studies in models of chronic inflammation have revealed dynamic changes in K14 methylation states at promoters of inflammatory response genes.

  • Drug development opportunities: The enzymes responsible for establishing and removing K14 methylation represent potential therapeutic targets. Antibodies against different K14 methylation states are being used in high-throughput screens to identify compounds that can modulate these modifications, with implications for epigenetic therapies .

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 2024 Thebiotek. All Rights Reserved.