Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production and Validation

The antibody is produced via a multi-step process:

  1. Gene Cloning: Heavy and light chain genes encoding HIST1H3A-specific antibodies are cloned .

  2. Vector Integration: Genes are inserted into expression vectors optimized for mammalian cell systems .

  3. Host Cell Transfection: HEK293F cells or rabbit cells are transfected to express and secrete antibodies .

  4. Purification: Affinity chromatography ensures high purity .

  5. Validation: Rigorous testing via ELISA, Western blot (WB), immunocytochemistry (ICC), immunofluorescence (IF), and immunohistochemistry (IHC-P) .

Applications and Dilution Recommendations

The antibody is validated for diverse assays, with species reactivity spanning human, mouse, and rat tissues .

ApplicationRecommended DilutionKey Findings
Western Blot1:500–1:2000 Detects a 15 kDa band in HeLa, NIH/3T3, and U2OS cell lysates .
ICC/IF1:50–1:500 Nuclear staining in HeLa cells, with DAPI counterstain .
IHC-P1:2000 Nuclear staining in human liver, mouse cerebral cortex, and rat pancreas .
ELISANot specifiedUsed in peptide array validation to confirm specificity .

Validation Data

  • Western Blot:

    • Observed band size: 15 kDa (matches histone H3.1) .

    • No cross-reactivity with unmodified or symmetrically di-methylated R17 peptides .

  • Immunofluorescence:

    • Confocal imaging shows nuclear localization in HeLa cells, co-stained with tubulin (red) and DAPI (blue) .

  • Immunohistochemistry:

    • Detects nuclear methylation in formalin-fixed paraffin-embedded (FFPE) tissues, including human liver and mouse cerebral cortex .

Species Reactivity

SpeciesReactivityApplications
HumanYesWB, ICC, IHC-P, IF
MouseYesWB, IHC-P
RatYesIHC-P

Biological Relevance

Mono-methylation at H3.1 R17 is implicated in:

  • Transcriptional Regulation: Facilitates chromatin accessibility for transcription machinery .

  • Epigenetic Signaling: Modulates DNA repair and cellular identity .

  • Cancer Pathology: Linked to chromatin remodeling in malignancies, though direct antibody applications focus on detection rather than therapeutic use .

Product Specs

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

This mono-methyl-Histone H3.1 (R17) recombinant monoclonal antibody is produced through a meticulous process. Initially, genes encoding the HIST1H3A antibody, encompassing both heavy and light chains, are cloned. These cloned genes are then integrated into expression vectors engineered for optimal performance. Subsequently, these modified expression vectors are introduced into host cells via transfection, where the host cells synthesize and secrete the antibody. The antibody undergoes purification using affinity chromatography to ensure its purity and functionality. To guarantee its efficacy, the antibody is rigorously tested in various applications, including ELISA, WB, ICC, and IF, specifically tailored for the precise detection of the human HIST1H3A protein mono-methylated at R17.

Mono-methylation of Histone H3.1 at arginine 17 (R17) plays a crucial role in transcriptional regulation, chromatin structure, DNA repair, cellular identity, and epigenetic signaling. This modification has significant implications in the development and progression of various diseases.

Form
Liquid
Lead Time
Typically, we are able to dispatch the products within 1-3 working days after receiving your orders. The delivery time may vary depending on the chosen purchase method and location. For precise delivery information, kindly consult your local distributors.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function

Histone H3.1 is a core component of the nucleosome, a fundamental structural unit of chromatin. Nucleosomes wrap and compact DNA, restricting DNA accessibility to cellular machinery that utilizes DNA as a template. Consequently, histones play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex interplay of post-translational modifications of histones, collectively known as the histone code, along with nucleosome remodeling.

Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer occurs through 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 a valuable indicator for determining whether tumors are heterochronous. PMID: 29482987
  3. This research reports that JMJD5, a Jumonji C (JmjC) domain-containing protein, functions as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stress conditions leading to a DNA damage response. PMID: 28982940
  4. This study suggests that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) presents an alternative proliferative marker. PMID: 29040195
  5. The data suggests that cytokine-induced histone 3 lysine 27 trimethylation serves as a mechanism to stabilize gene silencing in macrophages. PMID: 27653678
  6. This research indicates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, the histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with BRAF-V600E mutation and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. This data demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral, and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments reveal 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 describes, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation in adult cerebellar HGG is not rare. PMID: 28547652
  14. This research demonstrates that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest, while the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. This 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. Conversely, 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. This 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 the Raji cells treated by staurosporin. PMID: 26032366
  28. The research concludes that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. This data indicates that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. This research indicates 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. This data suggests that histone H3 lysine methylation (H3K4me3) plays a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. This data indicates that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. This research indicates 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 Mono-methyl-Histone H3.1 (R17) and why is it significant in epigenetic research?

Mono-methyl-Histone H3.1 (R17) refers to histone H3.1 protein that is mono-methylated at the arginine 17 position. This post-translational modification is involved in transcriptional regulation, chromatin structure modulation, DNA repair processes, cellular identity determination, and epigenetic signaling pathways . The significance of this modification lies in its role in controlling gene expression through altering chromatin accessibility. The recombinant monoclonal antibody targeting this specific modification allows researchers to study these processes with high specificity and sensitivity, making it an essential tool for epigenetic research. When designing experiments, researchers should consider that this modification exists in a complex network of histone modifications that collectively regulate gene expression.

What applications has the Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody been validated for?

The Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody has been validated for multiple applications in molecular and cellular biology research. According to available data, this antibody has been successfully used in ELISA, Western Blot (WB), Immunocytochemistry (ICC), and Immunofluorescence (IF) applications . For Western Blot, the recommended dilution ranges from 1:500 to 1:2000; for Immunofluorescence, the recommended dilution is 1:50 to 1:500; and for Immunocytochemistry, the recommended dilution is also 1:50 to 1:500 . These dilution ranges should be optimized by researchers depending on their specific experimental conditions, sample types, and detection methods employed. The versatility of this antibody across multiple applications makes it valuable for researchers investigating histone modifications from different methodological approaches.

What is the species reactivity of this antibody and how does this influence experimental design?

The Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody has been specifically designed to react with human (Homo sapiens) proteins . This specificity means that researchers working with human cell lines, tissues, or clinical samples will achieve optimal results. When designing experiments, researchers should be aware that this antibody might not recognize the equivalent modifications in other species or might show reduced affinity, which could lead to weaker signals or false negatives in non-human samples. For cross-species studies, validation experiments should be performed to confirm reactivity, or species-specific antibodies should be considered as alternatives. The human-specific nature of this antibody makes it particularly valuable for studies focusing on human diseases and development where epigenetic modifications play important roles.

What are the recommended storage conditions for maintaining antibody activity?

To maintain optimal activity of the Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody, proper storage conditions are essential. The antibody should be aliquoted upon receipt to avoid repeated freeze-thaw cycles and stored at -20°C . The antibody is typically supplied in PBS (pH 7.4) containing 150 mM NaCl, 0.02% sodium azide, and 50% glycerol as a stabilizer . When working with the antibody, it should be thawed on ice or at 4°C rather than at room temperature to preserve its binding capacity. Repeated freeze-thaw cycles can lead to protein denaturation and loss of antibody specificity and sensitivity. For long-term storage of working dilutions, addition of carrier proteins such as BSA (0.1-1%) may help maintain stability. Antibody activity should be monitored periodically, especially after extended storage periods, to ensure consistent experimental results.

How does H3.1 function as a chromatin redox sensor and what implications does this have for interpreting results?

Recent research has revealed that histone H3.1 functions as a chromatin-embedded redox sensor due to its unique cysteine residue at position 96 (Cys96), which is absent in other H3 variants . This residue can be oxidized in response to hydrogen peroxide (H₂O₂), particularly nuclear H₂O₂ (nH₂O₂), leading to nucleosome instability and subsequent exchange of H3.1 for the H3.3 variant . Studies have demonstrated that nH₂O₂-dependent oxidation of H3.1 Cys96 promotes this histone exchange, which is associated with chromatin decompaction, increased accessibility at promoter regions, and activation of gene expression, particularly genes involved in epithelial-mesenchymal transition (EMT) .

When interpreting results involving H3.1 modifications, researchers should consider that oxidative stress conditions might alter the chromatin landscape by promoting H3.1 exchange, potentially influencing other histone modifications including R17 methylation. Experimental designs should account for redox states in the cellular environment, and controls for oxidative conditions should be included. The dynamic interplay between redox signaling and histone modifications represents an important layer of epigenetic regulation that may affect experimental outcomes and interpretation of results involving H3.1 modifications.

What methodological approaches can be used to distinguish between different methylation states of H3.1 R17?

Distinguishing between different methylation states of H3.1 R17 (unmethylated, mono-methylated, di-methylated, and tri-methylated) requires careful methodological approaches. The Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody is specifically designed to recognize only the mono-methylated form at R17 . To ensure specificity, researchers should:

  • Perform peptide competition assays using unmethylated, mono-methylated, di-methylated, and tri-methylated peptides containing the R17 residue.

  • Include positive controls (cells or tissues known to have high levels of H3.1R17me1) and negative controls (cells with CARM1/PRMT4 knockdown, as these are the primary methyltransferases for this site).

  • Use mass spectrometry-based approaches for unbiased detection of different methylation states.

  • Consider employing parallel antibodies specific to other methylation states for comparative analysis.

  • Validate results using orthogonal methods such as ChIP-seq followed by mass spectrometry.

These methodological approaches help ensure that the signals detected truly represent mono-methylation at R17 rather than other methylation states or modifications at nearby residues, increasing the reliability of experimental findings.

How can ChIP-seq experiments be optimized when using the Mono-methyl-Histone H3.1 (R17) antibody?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) is a powerful technique for mapping histone modifications genome-wide. When using the Mono-methyl-Histone H3.1 (R17) antibody for ChIP-seq, several optimization strategies should be considered:

  • Cross-linking optimization: Standard 1% formaldehyde for 10 minutes at room temperature works for most histone modifications, but optimal cross-linking conditions may vary for R17 methylation.

  • Sonication parameters: Optimize sonication conditions to yield DNA fragments between 200-600 bp, while avoiding excessive heat that could affect epitope recognition.

  • Antibody validation: Confirm antibody specificity using peptide competition assays specifically for ChIP applications.

  • Input controls: Always include input DNA controls and IgG negative controls.

  • Sequential ChIP: Consider sequential ChIP (ChIP-reChIP) to identify genomic regions containing both H3.1R17me1 and other histone marks of interest.

Based on research findings in the field, ChIP-seq experiments with H3.1 variant-specific antibodies have successfully identified differential occupancy patterns at transcription start sites, with H3.1 showing decreased occupancy and H3.3 showing increased occupancy after hydrogen peroxide treatment . This indicates that the H3.1 methylation patterns may be dynamically regulated in response to cellular stressors, which should be considered when designing ChIP-seq experiments with the Mono-methyl-Histone H3.1 (R17) antibody.

What is the relationship between H3.1 R17 methylation and other histone modifications in the histone code?

H3.1 R17 mono-methylation exists within a complex network of histone modifications collectively known as the histone code. The relationship between H3.1 R17 methylation and other modifications is multifaceted:

  • Cross-talk with nearby modifications: Methylation at R17 may influence or be influenced by other modifications on the H3 tail, particularly those at K9, K14, and R2. For example, acetylation at K14 may facilitate enzyme access to R17 for methylation.

  • Exclusivity with certain modifications: Some modifications may be mutually exclusive with R17 methylation due to steric hindrance or enzyme competition.

  • Sequential modification patterns: R17 methylation may be part of sequential modification patterns, where one modification leads to another in a specific order.

  • Functional consequences: The combination of R17 methylation with other modifications determines functional outcomes such as transcriptional activation or repression.

When investigating H3.1 R17 methylation, researchers should consider analyzing co-occurring modifications to understand the broader epigenetic context. Methods such as sequential ChIP or mass spectrometry-based approaches can help identify modification patterns that co-exist with R17 methylation. Understanding these relationships is crucial for interpreting the biological significance of R17 methylation in different cellular contexts and disease states.

What are common issues encountered in Western blot applications with this antibody and how can they be resolved?

When using the Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody in Western blot applications, researchers may encounter several challenges. Here are common issues and their solutions:

IssuePotential CausesSolutions
Weak or no signalInsufficient antibody concentration; Epitope masking due to sample preparation; Degradation of target proteinIncrease antibody concentration within recommended range (1:500-1:2000) ; Use fresh samples with protease inhibitors; Include phosphatase inhibitors in lysis buffer
High backgroundExcessive antibody concentration; Insufficient blocking; Non-specific bindingOptimize antibody dilution; Extend blocking time or try different blocking agents; Include 0.1-0.3% Tween-20 in wash buffers
Multiple bandsCross-reactivity with other methylated histones; Protein degradation; Post-translational modificationsValidate with peptide competition assay; Use freshly prepared samples; Consider dephosphorylation treatment
Inconsistent resultsBatch-to-batch variation; Inconsistent sample preparation; Variable transfer efficiencyUse consistent antibody lots; Standardize sample preparation protocols; Monitor and standardize transfer efficiency

When optimizing Western blots with this antibody, researchers should be particularly attentive to extraction methods that preserve histone modifications, as harsh conditions may affect the methylation status. Additionally, including appropriate positive controls (such as recombinant H3.1 with verified R17 mono-methylation) and negative controls (such as samples treated with demethylase enzymes) can help validate the specificity of the observed signals.

How can specificity of the antibody be verified in different experimental contexts?

Verifying the specificity of the Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody across different experimental contexts is crucial for reliable results. Here are methodological approaches for verification:

  • Peptide competition assays: Pre-incubate the antibody with excess mono-methylated H3.1 R17 peptide before application. A specific antibody will show significantly reduced signal when pre-bound to its target epitope.

  • Methyltransferase inhibition or knockout: Treat cells with inhibitors of arginine methyltransferases (particularly CARM1/PRMT4) or use cells with knocked-out methyltransferases. Reduced signal confirms specificity for the methylated form.

  • Demethylase overexpression: Overexpress histone arginine demethylases that target R17. Decreased signal supports antibody specificity.

  • Mass spectrometry validation: Immunoprecipitate with the antibody and analyze by mass spectrometry to confirm that the captured proteins contain the expected modification.

  • Cross-reactivity testing: Test the antibody against a panel of similar histone modifications, especially di- and tri-methylated R17, as well as methylations at other arginine residues (R2, R8, R26) to ensure specificity.

  • Dot blot analysis: Perform dot blots with modified and unmodified peptides to establish binding specificity without the complications of protein denaturation or complex sample matrices.

These validation approaches should be applied within the specific experimental context (cell type, treatment conditions, etc.) being studied, as antibody performance can vary across different biological systems and experimental conditions.

What considerations should be made when using this antibody in immunofluorescence applications?

When using the Mono-methyl-Histone H3.1 (R17) Recombinant Monoclonal Antibody for immunofluorescence applications, several important considerations should be addressed:

  • Fixation method: Different fixation methods can affect epitope accessibility. For histone modifications, 4% paraformaldehyde (10-15 minutes) is generally recommended, but methanol fixation may provide better nuclear penetration. Compare both methods to determine optimal conditions.

  • Permeabilization: Adequate permeabilization is crucial for nuclear antigens. Try 0.1-0.5% Triton X-100 for 10 minutes, but optimize based on cell type.

  • Antibody dilution: Begin with the recommended dilution range (1:50-1:500) and optimize. Too high concentration may lead to non-specific binding, while too low may result in weak signals.

  • Blocking conditions: Use 5% normal serum from the same species as the secondary antibody. BSA (3-5%) can also be effective. Block for at least 1 hour at room temperature.

  • Controls: Include a negative control (no primary antibody) and positive controls (cells known to express H3.1R17me1). Consider using peptide competition controls to demonstrate specificity.

  • Signal detection optimization: Adjust exposure times to capture specific signals while avoiding saturation. Use appropriate filter sets matched to your secondary antibody fluorophores.

  • Co-localization studies: When performing co-localization with other nuclear markers, select secondary antibodies with well-separated emission spectra to avoid bleed-through.

  • Quantification approaches: For quantitative analyses, establish consistent imaging parameters and use appropriate software for signal quantification.

By addressing these considerations methodically, researchers can achieve reliable and reproducible immunofluorescence results with the Mono-methyl-Histone H3.1 (R17) antibody.

How does the exchange between H3.1 and H3.3 variants influence gene expression and cellular phenotypes?

The exchange between histone variants H3.1 and H3.3 represents a dynamic mechanism for regulating chromatin structure and gene expression. Research has revealed several key aspects of this process:

  • Differential incorporation mechanisms: H3.1 incorporation is primarily replication-dependent, whereas H3.3 incorporation is replication-independent . This difference allows for dynamic regulation of chromatin structure throughout the cell cycle.

  • Chromatin decompaction: The replacement of H3.1 with H3.3 is associated with chromatin decompaction and increased accessibility at promoter regions . Electron microscopy studies have shown that this exchange reduces heterochromatin content, particularly near the nuclear envelope .

  • Transcriptional activation: H3.3 incorporation correlates with transcriptional activation . The exchange from H3.1 to H3.3 at promoter regions facilitates the binding of transcription factors and the assembly of the transcriptional machinery.

  • Redox sensitivity: The unique cysteine residue (Cys96) in H3.1 serves as a redox sensor, with oxidation promoting H3.1 exchange for H3.3 . This mechanism links oxidative signaling to epigenetic reprogramming.

  • EMT regulation: Research has shown that the exchange of H3.1 for H3.3 precedes and is required for epithelial-mesenchymal transition (EMT), a process crucial for cancer metastasis . This exchange activates EMT gene expression programs driven by nuclear hydrogen peroxide.

  • Temporal dynamics: The exchange shows specific temporal dynamics, with peak H3.3 incorporation at promoters occurring around 4 hours after stimulus, followed by a return toward baseline levels by 24 hours . This indicates a transitional role in gene activation rather than a permanent change.

Understanding the interplay between H3.1 R17 methylation and these variant exchange dynamics presents an important area for future research, as both processes contribute to epigenetic regulation of gene expression and cellular phenotypes.

What role does H3.1 R17 mono-methylation play in disease processes and potential therapeutic targets?

H3.1 R17 mono-methylation plays significant roles in various disease processes, particularly through its influence on gene expression regulation:

  • Cancer progression: Aberrant histone arginine methylation, including at R17, has been implicated in cancer development and progression. Changes in methylation patterns can lead to inappropriate activation or silencing of genes involved in cell proliferation, apoptosis, and metastasis.

  • Inflammatory diseases: Dysregulation of histone arginine methylation contributes to aberrant inflammatory responses by affecting the expression of cytokines and other immune-related genes.

  • Neurodegenerative disorders: Altered histone methylation patterns have been observed in various neurodegenerative conditions, potentially affecting neuronal gene expression programs.

  • Developmental disorders: As histone modifications are crucial for proper gene expression during development, abnormal R17 methylation may contribute to developmental abnormalities.

From a therapeutic perspective, several approaches targeting histone arginine methylation are being explored:

  • Methyltransferase inhibitors: Compounds targeting the enzymes responsible for R17 methylation (primarily CARM1/PRMT4) could normalize aberrant methylation patterns.

  • Demethylase activators: Molecules that enhance the activity of arginine demethylases might counter excessive methylation at R17.

  • Reader domain inhibitors: Blocking the proteins that recognize and bind to methylated R17 could interrupt downstream signaling without altering the methylation itself.

  • Combination epigenetic therapies: Targeting multiple epigenetic modifications simultaneously may provide synergistic effects in diseases with complex epigenetic dysregulation.

Researchers using the Mono-methyl-Histone H3.1 (R17) antibody can contribute to this field by characterizing methylation patterns in disease models, identifying genes regulated by this modification, and evaluating the effects of potential therapeutic compounds on R17 methylation levels and distribution.

How do the functions of H3.1 R17 mono-methylation compare with other arginine methylation sites on histones?

H3.1 R17 mono-methylation is one of several arginine methylation sites on histones, each with distinct and sometimes overlapping functions. Understanding these comparisons provides important context for research:

Methylation SitePrimary EnzymesPrimary FunctionsGenomic LocalizationAssociated Transcriptional Effect
H3R17me1CARM1/PRMT4Transcriptional activation, Hormone signalingPromoters and enhancersGenerally activating
H3R2me1/2PRMT6Transcriptional repression, Cell proliferationPromotersGenerally repressive
H3R8me1/2PRMT2, PRMT5Transcriptional regulationVariedContext-dependent
H3R26me1/2CARM1/PRMT4Transcriptional activationEnhancersGenerally activating
H4R3me1/2PRMT1, PRMT5Transcriptional regulation, DNA damage responsePromotersSymmetric dimethylation: repressive; Asymmetric dimethylation: activating

Key functional comparisons include:

  • Cooperation and antagonism: H3R17 methylation often cooperates with H3R26 methylation (both mediated by CARM1/PRMT4) but may antagonize H3R2 methylation (mediated by PRMT6) in some contexts.

  • Cross-talk with other modifications: Unlike H3R2 methylation, which can inhibit H3K4 methylation, H3R17 methylation often positively correlates with nearby activating modifications like H3K4 methylation and H3K9 acetylation.

  • Temporal dynamics: H3R17 methylation often occurs rapidly in response to signaling events (particularly hormone signaling), while other arginine methylations may show different temporal patterns.

  • Reader proteins: Different methylarginine sites are recognized by distinct reader proteins, leading to recruitment of different downstream effector complexes.

Researchers studying H3.1 R17 mono-methylation should consider these comparative aspects when designing experiments and interpreting results, as the specific biological context may influence the functional outcomes of this modification.

What emerging technologies can enhance the detection and functional analysis of H3.1 R17 mono-methylation?

Several cutting-edge technologies are expanding our capabilities for studying H3.1 R17 mono-methylation:

  • CUT&RUN and CUT&Tag: These techniques offer advantages over traditional ChIP-seq by providing higher signal-to-noise ratios and requiring fewer cells. They can be optimized for the Mono-methyl-Histone H3.1 (R17) antibody to map genomic distributions with greater sensitivity.

  • Single-cell epigenomics: Techniques such as single-cell CUT&Tag allow for analysis of histone modification heterogeneity at the single-cell level, revealing cell-to-cell variation in H3.1 R17 methylation patterns within populations.

  • Proximity ligation assays (PLA): These can detect interactions between H3.1 R17 methylation and other chromatin-associated proteins in situ, providing spatial context within the nucleus.

  • Live-cell imaging of histone modifications: Using techniques such as FRAP (Fluorescence Recovery After Photobleaching) with specific readers of H3.1 R17 methylation fused to fluorescent proteins can reveal dynamics of this modification in living cells.

  • Mass spectrometry-based approaches: Advanced mass spectrometry methods such as MALDI-TOF MS/MS and quantitative proteomics can identify and quantify combinatorial patterns of histone modifications co-occurring with H3.1 R17 methylation.

  • CRISPR-based epigenome editing: Tools like dCas9 fused to CARM1/PRMT4 can induce site-specific H3.1 R17 methylation, allowing for causal studies of this modification's effects on gene expression and cellular phenotypes.

  • Computational integration approaches: Machine learning algorithms can integrate multiple epigenomic datasets to predict functional consequences of H3.1 R17 methylation patterns across the genome.

These emerging technologies offer researchers powerful new approaches to understanding the dynamics, distribution, and functional significance of H3.1 R17 mono-methylation in various biological contexts.

What are the key experimental controls required for studying H3.1 R17 mono-methylation in different research contexts?

Robust experimental controls are essential for reliable research on H3.1 R17 mono-methylation. Here are key controls that should be implemented across different research contexts:

  • Antibody specificity controls:

    • Peptide competition assays using mono-methylated H3R17 peptides versus unmethylated and differently methylated peptides

    • Dot blots or Western blots with recombinant histones containing defined modifications

    • Use of alternative antibodies targeting the same modification for validation

  • Biological manipulation controls:

    • CARM1/PRMT4 knockdown or knockout (enzymes responsible for H3R17 methylation)

    • CARM1/PRMT4 overexpression to increase modification levels

    • Treatment with methyltransferase inhibitors

    • Demethylase overexpression

  • ChIP-seq specific controls:

    • Input DNA controls to account for genomic biases

    • IgG controls to establish background binding

    • Total H3 ChIP to normalize for nucleosome occupancy

    • Sequential ChIP to verify co-occurrence with other marks

  • Immunofluorescence controls:

    • Secondary antibody-only controls

    • Peptide competition controls

    • Positive controls (cells known to have the modification)

    • Negative controls (cells with CARM1/PRMT4 knockdown)

  • Functional analysis controls:

    • Point mutations in H3.1 at R17 (R17K or R17A) to prevent methylation

    • Time-course experiments to establish temporal dynamics

    • Dose-response studies for treatments affecting methylation levels

  • Cross-species validation:

    • Parallel experiments in different cell lines/organisms to establish conservation

    • Controls for histone variant-specific effects

Implementation of these controls helps distinguish specific effects of H3.1 R17 mono-methylation from other biological processes and technical artifacts, enhancing the reliability and interpretability of research findings.

How can the study of H3.1 R17 mono-methylation contribute to our understanding of epigenetic inheritance and cellular memory?

The study of H3.1 R17 mono-methylation offers unique insights into epigenetic inheritance and cellular memory due to several key properties:

  • Replication dynamics: As H3.1 is primarily incorporated during DNA replication, understanding how R17 methylation patterns are maintained or altered during cell division can illuminate mechanisms of epigenetic inheritance. Research questions include:

    • How are methylation patterns distributed to daughter cells during replication?

    • What role do chaperone proteins play in preserving R17 methylation during nucleosome assembly?

  • Stability and turnover: The stability of H3.1 R17 mono-methylation over time influences its potential as a carrier of epigenetic information. Studies can examine:

    • The half-life of this modification compared to other histone marks

    • The enzymes responsible for active removal versus passive dilution during replication

    • How cellular stressors affect the stability of this modification

  • Developmental programming: H3.1 R17 methylation patterns established during development may contribute to cell fate decisions and cellular memory. Researchers can investigate:

    • Changes in methylation patterns during differentiation

    • The role of R17 methylation in maintaining cell identity

    • The potential for these marks to be disrupted in disease states

  • Environmental responses: How H3.1 R17 methylation responds to environmental signals can inform our understanding of cellular adaptation and memory. Key areas include:

    • The dynamics of R17 methylation in response to signaling pathways

    • The persistence of environmentally-induced changes in methylation

    • The role of this modification in adaptive responses to repeated stimuli

  • Cross-generational inheritance: While most histone modifications are reprogrammed during gametogenesis, some may persist. Research could explore:

    • Whether R17 methylation patterns can escape reprogramming

    • The potential role of this modification in transgenerational epigenetic inheritance

    • Interactions with other epigenetic mechanisms like DNA methylation

By addressing these research areas, scientists can better understand how H3.1 R17 mono-methylation contributes to the complex landscape of epigenetic inheritance and cellular memory, with implications for development, aging, and disease.

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.