No cross-reactivity with non-methylated K9, dimethylated (K9me2), or trimethylated (K9me3) histones .
High specificity confirmed via dot-blot assays against synthetic peptides .
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 .
Recombinant Protein Production: Monitors histone methylation in CHO cell lines to optimize monoclonal antibody expression .
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.
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.
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.
While all targeting histone H3 modifications, these antibodies recognize distinct epitopes:
Antibody Target | Recognition Specificity | Associated Function | Molecular Weight |
---|---|---|---|
H3K9me1 | Mono-methylation at lysine 9 | Transcriptional repression | 15-18 kDa |
H3K9me2/me3 | Di/tri-methylation at lysine 9 | Heterochromatin formation | 15-18 kDa |
H3K36me1 | Mono-methylation at lysine 36 | Transcriptional elongation | 15-18 kDa |
H3K18me1 | Mono-methylation at lysine 18 | Context-dependent regulation | 15-18 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.
Recombinant monoclonal antibodies targeting H3K9me1 typically have the following specifications:
Applications: Western Blot (WB), Immunofluorescence (IF), Immunocytochemistry (ICC), ChIP, ELISA
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)
Based on validation data from manufacturers, these antibodies perform reliably in multiple applications:
Application | Recommended Dilution | Incubation Conditions | Expected Result |
---|---|---|---|
Western Blot | 1:2000-1:5000 | Overnight at 4°C | Band at 15-18 kDa |
IF/ICC | 1:200-1:800 | 1 hour at 25°C or overnight at 4°C | Nuclear localization |
ChIP | 1-5 μg per reaction | Overnight at 4°C | Enrichment at target loci |
ELISA | Assay-dependent | Assay-dependent | Specific 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 .
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 .
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.
Challenge | Possible Causes | Solutions |
---|---|---|
Weak WB signal | Insufficient antibody concentration, low target abundance | Increase antibody concentration (1:500-1:2000), enrich for histones using acid extraction |
High background in IF | Insufficient blocking, excessive antibody | Extend blocking time (2 hours), reduce antibody concentration, include 0.1% BSA in wash buffer |
Cross-reactivity | Antibody specificity issues | Verify antibody specificity with peptide competition assays, use recombinant antibodies with validated specificity |
No signal in ChIP | Inefficient crosslinking, epitope masking | Optimize crosslinking time, try alternative fixation methods, adjust sonication conditions |
Variable results | Batch-to-batch variation | Use 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 .
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.
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 .
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.
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.
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).
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.
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?