Mono-methyl-HIST1H3A (K9) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Table 1: Applications and Working Concentrations

ApplicationConcentration RangeKey Validation Data
ChIP5–10 µg/mLEnrichment of H3K9me1 in HeLa cells (real-time PCR)
Western Blot0.2–1 µg/mLDetection in HeLa lysates and recombinant H3.3 ; observed at ~16–17 kDa
Immunocytochemistry0.5–2 µg/mLStaining of sodium butyrate-treated HeLa cells (red fluorescence)
ELISA0.2–1 µg/mLSpecific binding to K9me1 peptides
CUT&Tag1 µg per 10⁵ cellsEnrichment at MYOD1 locus in K562 cells
IHC1:500–1:1000Staining in human stomach and cervix tissues

3.1. Antibody Specificity

  • No cross-reactivity with non-methylated K9, dimethylated (K9me2), or trimethylated (K9me3) histones .

  • High specificity confirmed via dot-blot assays against synthetic peptides .

3.2. Experimental Validation

Assay TypeDetailsSource
CUT&TagEnrichment at MYOD1 promoter in K562 cells using 1 µg antibody
Western BlotClear band at ~17 kDa in HeLa lysates and calf thymus histones
ImmunocytochemistryDistinct nuclear staining in HeLa cells (Alexa Fluor® 488 conjugate)
IHCDetection in formalin-fixed stomach and colon carcinoma tissues

4.1. Epigenetic Studies

  • Chromatin Mapping: Used to identify H3K9me1-enriched regions in gene promoters and enhancers .

  • Gene Regulation: Links H3K9 monomethylation to transcriptional activation or repression in cancer models .

4.2. Biotechnological Applications

  • Recombinant Protein Production: Monitors histone methylation in CHO cell lines to optimize monoclonal antibody expression .

Comparative Analysis with Other H3K9 Antibodies

AntibodyCloneTypeReactivityApplications
RM150RM150MonoclonalHuman, VertebratesChIP, WB, ICC, IHC
EPR16989EPR16989MonoclonalHuman, Mouse, RatChIP, WB, PepArr
Ab8896PolyclonalRabbitHuman, CowChIP, WB, ICC/IF
A20734ARC2677MonoclonalHuman, Mouse, RatWB, IHC-P, CUT&Tag

Product Specs

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

The generation of the mono-methyl-HIST1H3A (K9) recombinant monoclonal antibody typically involves the integration of the HIST1H3A antibody-encoding gene into expression vectors. These vectors are subsequently introduced into host cells through polyethyleneimine-mediated transfection techniques. The host cells harboring these vectors are cultivated to produce and secrete the antibodies. Following purification via affinity chromatography, the antibodies undergo rigorous evaluation using ELISA, IHC, IF, and FC assays, demonstrating their specific binding to the human HIST1H3A protein mono-methylated at K9.

HIST1H3A mono-methylated at K9 is commonly found in genomic regions associated with heterochromatin and plays a pivotal role in epigenetic regulation. This modification contributes significantly to gene repression and the formation of transcriptionally inactive heterochromatin, ultimately impacting proper gene expression patterns and cellular identity. It has implications for various biological processes and disease states.

Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days following receipt of your order. Delivery timelines may vary depending on the mode of purchase and location. For specific delivery details, please consult your local distributor.
Synonyms
Histone H3.1 (Histone H3/a) (Histone H3/b) (Histone H3/c) (Histone H3/d) (Histone H3/f) (Histone H3/h) (Histone H3/i) (Histone H3/j) (Histone H3/k) (Histone H3/l), HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, H3FA, H3FL, H3FC, H3FB, H3FD, H3FI, H3FH, H3FK, H3FF, H3FJ
Target Names
Uniprot No.

Target Background

Function

Histone H3 constitutes a core component of nucleosomes. Nucleosomes serve to wrap and compact DNA into chromatin, thereby restricting DNA accessibility to cellular machinery that relies on DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is meticulously regulated through a complex array of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.

Gene References Into Functions
  1. Evidence suggests a mechanism for epigenetic regulation in cancer 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 instrumental in determining whether the tumors are heterochronous. PMID: 29482987
  3. Here, we report that JMJD5, a Jumonji C (JmjC) domain-containing protein, functions as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions, inducing a DNA damage response. PMID: 28982940
  4. Our findings indicate that the Ki-67 antigen proliferative index exhibits significant limitations, making phosphohistone H3 (PHH3) a viable alternative proliferative marker. PMID: 29040195
  5. These results identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data reveals that, in the early developing human brain, HIST1H3B constitutes the most abundant H3.1 transcript 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 commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Notably, among these K27M+ diffuse midline gliomas... PMID: 26517431
  8. Our findings demonstrate 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 exhibits specific binding to DNA and 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. We describe, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3), which correlates with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. The occurrence of H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Our data demonstrate 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 exhibited the highest prevalence when the Dbf4 transcription level was at its peak, whereas 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. Our findings suggest 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) necessitates molecular recognition of secondary structure motifs within the H3 tail, potentially representing an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results showcase 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 induced 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 the regulation of gene transcription of DNA damage response genes. 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. Conversely, histone 3 mutations appear to be absent in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Our data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to facilitate 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. PMID: 25961932
  27. Collectively, we verified that histone H3 is a genuine substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. Our findings indicate that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Our data reveal 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. Our data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for the incorporation of histone H3.1-H4 into chromatin but is crucial for the stability of H3.1-H4. PMID: 26167883
  31. Our findings suggest that 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. Our data indicate that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Our findings demonstrate 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-HIST1H3A (K9) and why is it significant in epigenetic research?

Mono-methyl-HIST1H3A (K9) refers to the monomethylation of lysine 9 on histone H3, a core component of nucleosomes. This epigenetic modification plays a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. Histones are subject to complex post-translational modifications, collectively known as the histone code, which regulate DNA accessibility to cellular machinery . Mono-methylation at K9 specifically is linked to transcriptional repression mechanisms, making it a critical target for researchers studying chromatin dynamics and gene regulation.

How does a Mono-methyl-HIST1H3A (K9) antibody differ from antibodies targeting other histone H3 methylation states?

While all targeting histone H3 modifications, these antibodies recognize distinct epitopes:

Antibody TargetRecognition SpecificityAssociated FunctionMolecular Weight
H3K9me1Mono-methylation at lysine 9Transcriptional repression15-18 kDa
H3K9me2/me3Di/tri-methylation at lysine 9Heterochromatin formation15-18 kDa
H3K36me1Mono-methylation at lysine 36Transcriptional elongation15-18 kDa
H3K18me1Mono-methylation at lysine 18Context-dependent regulation15-18 kDa

Specificity testing confirms that mono-methyl K9 antibodies do not cross-react with di-methyl or tri-methyl K9 modifications, nor with modifications at other lysine residues such as K27 or K4 . This specificity is critical for accurate interpretation of experimental results.

What are the key specifications of commercially available Mono-methyl-HIST1H3A (K9) Recombinant Monoclonal Antibodies?

Recombinant monoclonal antibodies targeting H3K9me1 typically have the following specifications:

  • Host/Isotype: Rabbit IgG

  • Clonality: Monoclonal (recombinant)

  • Applications: Western Blot (WB), Immunofluorescence (IF), Immunocytochemistry (ICC), ChIP, ELISA

  • Reactivity: Human, mouse, rat

  • Molecular Weight: Typically detected at 15-18 kDa (predicted 15 kDa)

  • Storage Conditions: -20°C in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3)

What are the validated applications for Mono-methyl-HIST1H3A (K9) antibodies and their optimal working dilutions?

Based on validation data from manufacturers, these antibodies perform reliably in multiple applications:

ApplicationRecommended DilutionIncubation ConditionsExpected Result
Western Blot1:2000-1:5000Overnight at 4°CBand at 15-18 kDa
IF/ICC1:200-1:8001 hour at 25°C or overnight at 4°CNuclear localization
ChIP1-5 μg per reactionOvernight at 4°CEnrichment at target loci
ELISAAssay-dependentAssay-dependentSpecific detection

Experimental validation has confirmed reactivity in multiple cell lines including HeLa, HEK-293, THP-1, K-562, Jurkat, NIH/3T3, C2C12, HSC-T6, and PC-12 cells .

What is the optimal Western blot protocol for detecting Mono-methyl-HIST1H3A (K9)?

For optimal Western blot results:

  • Sample Preparation: Extract nuclear proteins or isolate histones using acid extraction.

  • Gel Electrophoresis: Run 10-30 μg of nuclear lysate on a 5-20% SDS-PAGE gel at 70V (stacking)/90V (resolving) for 2-3 hours .

  • Transfer: Transfer proteins to nitrocellulose membrane at 150 mA for 50-90 minutes .

  • Blocking: Block membrane with 5% non-fat milk in TBS for 1.5 hours at room temperature .

  • Primary Antibody: Incubate with anti-H3K9me1 antibody at 1:500-1:5000 dilution overnight at 4°C .

  • Washing: Wash membrane with TBS-0.1% Tween 3 times, 5 minutes each .

  • Secondary Antibody: Incubate with HRP-conjugated anti-rabbit IgG at 1:5000-1:10000 for 1-1.5 hours at room temperature .

  • Detection: Develop using enhanced chemiluminescence (ECL) detection system .

The expected band for mono-methyl histone H3 (K9) appears at approximately 15-18 kDa .

What protocols yield optimal results for immunofluorescence detection of H3K9me1?

For successful IF/ICC staining:

  • Fixation: Fix cells with either 4% paraformaldehyde (5-10 min) or 100% methanol (5 min) .

  • Permeabilization: Permeabilize with 0.05-0.3% Triton X-100 in PBS or TBS .

  • Blocking: Block with 2-10% normal goat serum and/or 1% BSA in PBS for 30-60 minutes .

  • Primary Antibody: Incubate with anti-H3K9me1 antibody at 1:100-1:800 dilution for 1 hour at room temperature or overnight at 4°C .

  • Secondary Antibody: Incubate with fluorophore-conjugated anti-rabbit IgG (e.g., Alexa Fluor 488) at 1:200-1:1000 dilution for 30-60 minutes at room temperature .

  • Counterstaining: Counterstain nuclei with DAPI and cell membranes with WGA if desired .

  • Visualization: Observe using a fluorescence microscope with appropriate filter sets .

H3K9me1 typically shows nuclear localization with potential enrichment in specific nuclear domains.

What are common challenges when working with Mono-methyl-HIST1H3A (K9) antibodies and how can they be addressed?

ChallengePossible CausesSolutions
Weak WB signalInsufficient antibody concentration, low target abundanceIncrease antibody concentration (1:500-1:2000), enrich for histones using acid extraction
High background in IFInsufficient blocking, excessive antibodyExtend blocking time (2 hours), reduce antibody concentration, include 0.1% BSA in wash buffer
Cross-reactivityAntibody specificity issuesVerify antibody specificity with peptide competition assays, use recombinant antibodies with validated specificity
No signal in ChIPInefficient crosslinking, epitope maskingOptimize crosslinking time, try alternative fixation methods, adjust sonication conditions
Variable resultsBatch-to-batch variationUse recombinant monoclonal antibodies for consistency, maintain consistent experimental conditions

Peptide competition assays demonstrate that H3K9me1 antibodies can be successfully blocked by the immunizing peptide (mono-methyl K9) but not by peptides containing di-methyl K9, tri-methyl K9, or other modified residues .

How can I verify the specificity of a Mono-methyl-HIST1H3A (K9) antibody?

To confirm antibody specificity:

  • Peptide Competition: Perform Western blot with antibody pre-incubated with mono-methyl K9 peptide versus other methylated states. A specific antibody will be blocked only by the mono-methyl K9 peptide but not by di-methyl K9, tri-methyl K9, mono-methyl K27, or unmodified peptides .

  • Knockout/Knockdown Controls: Test antibody reactivity in cells with knockdown of enzymes responsible for H3K9 monomethylation (e.g., G9a). Western blot analysis shows reduced H3K9me1 signal in G9a siRNA-treated cells compared to untreated controls .

  • Cross-Reactivity Testing: Examine reactivity against a panel of modified histone peptides using dot blots or ELISA to confirm specific recognition of H3K9me1.

  • Application-Specific Validation: For ChIP applications, validate enrichment at known H3K9me1-marked regions versus negative control regions.

How can Mono-methyl-HIST1H3A (K9) antibodies be used for ChIP-seq applications?

For successful ChIP-seq experiments:

  • Crosslinking: Fix cells with 1% formaldehyde for 10 minutes at room temperature.

  • Chromatin Preparation: Lyse cells, isolate nuclei, and sonicate to generate 200-500 bp DNA fragments.

  • Immunoprecipitation: Incubate chromatin with 2-5 μg anti-H3K9me1 antibody overnight at 4°C.

  • Bead Capture: Add protein A/G magnetic beads, incubate 2-4 hours at 4°C.

  • Washing: Perform stringent washes to remove non-specific binding.

  • Elution and Reversal: Elute bound chromatin and reverse crosslinks.

  • Library Preparation: Purify DNA and prepare sequencing libraries.

  • Data Analysis: Align reads, call peaks, and analyze genomic distribution.

ChIP-grade antibodies against H3K9me1 have been validated in multiple cell types and show enrichment at specific genomic regions associated with transcriptional regulation .

What experimental designs can reveal functional relationships between H3K9me1 and other chromatin modifications?

To investigate functional relationships:

  • Sequential ChIP (Re-ChIP): Perform successive immunoprecipitations with H3K9me1 antibody followed by antibodies against other modifications to identify co-occurrence.

  • Integrated Multi-Omics: Combine ChIP-seq (H3K9me1), RNA-seq, ATAC-seq, and DNA methylation analysis from the same samples to correlate modifications with gene expression and chromatin accessibility.

  • Perturbation Studies: Analyze H3K9me1 distribution following manipulation of writer/eraser enzymes or treatment with epigenetic inhibitors.

  • Time-Course Experiments: Track changes in H3K9me1 distribution during cellular differentiation, response to stimuli, or cell cycle progression.

  • Single-Cell Approaches: Combine with single-cell technologies to assess heterogeneity in H3K9me1 distribution across cell populations.

How should ChIP-seq data generated with Mono-methyl-HIST1H3A (K9) antibodies be analyzed?

For robust ChIP-seq data analysis:

  • Quality Control: Assess sequencing quality, mapping rates, library complexity, and signal-to-noise ratios.

  • Peak Calling: Use appropriate algorithms (e.g., MACS2) with parameters optimized for histone modifications.

  • Genomic Distribution: Analyze H3K9me1 enrichment relative to genomic features (promoters, enhancers, gene bodies).

  • Integration: Correlate H3K9me1 distribution with other histone marks, transcription factor binding, and gene expression.

  • Differential Binding Analysis: Compare H3K9me1 patterns between experimental conditions.

  • Motif Analysis: Identify DNA sequence motifs enriched in H3K9me1-marked regions.

  • Pathway Analysis: Perform gene ontology and pathway enrichment for genes associated with H3K9me1 marks.

  • Visualization: Create genome browser tracks and heatmaps to present spatial relationships.

What are the key considerations when interpreting Western blot data for histone modifications?

When analyzing Western blot results:

  • Appropriate Controls: Include total histone H3 as a loading control rather than typical housekeeping genes.

  • Molecular Weight Verification: Confirm the band appears at the expected size (15-18 kDa for H3) .

  • Quantification Method: Use densitometry to quantify signal intensity, normalizing to total H3.

  • Multiple Biological Replicates: Perform at least three independent experiments for statistical validity.

  • Relative Quantification: Compare the ratio of modified to total histones rather than absolute values.

  • Sample Preparation Effects: Be aware that extraction methods can affect retention of histone modifications.

  • Antibody Specificity: Consider possible cross-reactivity with similar modifications (validated by peptide competition assays).

How can Mono-methyl-HIST1H3A (K9) antibodies be integrated with cutting-edge epigenomic technologies?

Innovative applications include:

  • CUT&RUN/CUT&Tag: These techniques offer higher signal-to-noise ratios than traditional ChIP and require less starting material, making them valuable for rare cell populations.

  • Single-Cell Epigenomics: Adaptation of H3K9me1 antibodies for single-cell ChIP-seq or CUT&Tag enables analysis of cellular heterogeneity.

  • Spatial Epigenomics: Combining immunofluorescence with in situ sequencing to map H3K9me1 distribution in tissue context.

  • Live-Cell Imaging: Development of recombinant antibody fragments for real-time tracking of H3K9me1 dynamics.

  • CRISPR Epigenome Editing: Using H3K9me1 antibodies to validate targeted methylation/demethylation by CRISPR-based tools.

  • Proteomics Integration: Combining ChIP with mass spectrometry to identify proteins associating with H3K9me1-marked chromatin.

What are the current gaps in understanding H3K9me1 function that could be addressed with improved antibody-based approaches?

Key research questions include:

  • Cell-Type Specificity: How does H3K9me1 distribution vary across different cell types and tissues?

  • Temporal Dynamics: What are the kinetics of H3K9me1 deposition and removal during cellular processes?

  • Reader Proteins: Which proteins specifically recognize H3K9me1 and how do they function?

  • Disease Associations: How are H3K9me1 patterns altered in disease states?

  • Evolutionary Conservation: How conserved are H3K9me1 patterns across species?

  • Crosstalk with Other Modifications: How does H3K9me1 interact with other histone marks and DNA modifications?

  • Functional Consequences: What is the direct impact of H3K9me1 gain or loss on chromatin structure and gene expression?

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.