HIST1H2AG (Histone Cluster 1 H2A Family Member G) is a core component of nucleosomes, playing critical roles in:
The Ab-118 epitope corresponds to residues near K118, a site implicated in histone crosstalk and epigenetic signaling .
Vendor | Catalog # | Applications Validated |
---|---|---|
Biomatik | CAC15621 | ELISA, WB, IHC, IF, IP |
Biorbyt | orb517097 | ELISA, IF, IHC, IP, WB |
Assay Genie | PACO60624 | ELISA, IHC (1:2,000–1:10,000) |
BAP1-deficient B-cells showed dysregulated H2AK119ub levels, impairing antibody production and germinal center dynamics .
The antibody helped map genome-wide H2AK119ub changes linked to defective humoral immunity .
Overexpression observed in hepatocellular carcinoma (HCC) and colon cancer tissues .
IHC staining revealed elevated HIST1H2AG in breast cancer vs. normal tissue .
No cross-reactivity in negative controls using PBS or mismatched secondary antibodies .
Lot-specific variability noted; users must verify datasheets for batch consistency .
The HIST1H2AG (Ab-118) Polyclonal Antibody has been validated for multiple research applications including ELISA, Western Blot (WB), Immunoprecipitation (IP), Chromatin Immunoprecipitation (ChIP), Immunohistochemistry (IHC), and Immunofluorescence (IF). These applications provide researchers with versatile options for detecting histone H2A type 1 in various experimental contexts . The antibody's broad application profile makes it suitable for both protein quantification and localization studies, with particular strength in chromatin-based research applications.
For optimal results, researchers should use the following application-specific dilution ranges:
Western Blot: 1:500-5000
Immunohistochemistry (paraffin-embedded sections): 1:20-200
These ranges should be considered starting points for optimization, as actual optimal dilutions may vary depending on sample type, preparation method, and detection system employed. Preliminary titration experiments are recommended when working with new sample types.
The HIST1H2AG (Ab-118) Antibody has confirmed reactivity with human (Homo sapiens) samples across multiple applications . Some product variants also demonstrate reactivity with mouse (Mus musculus) samples . The cross-species reactivity reflects the highly conserved nature of histone proteins across mammalian species, though researchers should verify reactivity when working with species not explicitly listed in the product specifications.
This polyclonal antibody was generated using a peptide sequence around the site of Lysine 118 derived from Human Histone H2A type 1 . The specific immunogen design targeting this region allows for detection of HIST1H2AG protein in its native and denatured forms, making it suitable for both structural and functional studies of histone H2A type 1.
To maintain optimal activity, the HIST1H2AG (Ab-118) Antibody should be stored refrigerated at 2-8°C for short-term use (up to 2 weeks). For long-term storage, it should be kept at -20°C in small aliquots to prevent freeze-thaw cycles that could degrade the antibody . The antibody is supplied in a buffer containing 50% glycerol and 0.03% Proclin 300 as a preservative, which helps maintain stability during storage .
This antibody serves as a valuable tool in epigenetic research through its validated use in Chromatin Immunoprecipitation (ChIP) assays . Researchers can employ it to investigate histone H2A distribution patterns across the genome, histone modifications associated with specific genomic regions, and changes in chromatin structure during cellular processes like differentiation or disease progression. When combined with sequencing (ChIP-seq) or qPCR techniques, this antibody enables genome-wide mapping of H2A-associated regulatory elements and their dynamics during biological processes.
When designing multi-parameter histone studies, researchers should consider potential epitope masking effects where histone modifications may interfere with antibody binding. Since this antibody targets a specific region around Lysine 118, researchers should verify that post-translational modifications near this site (such as methylation, acetylation, or phosphorylation) won't affect antibody recognition. For co-detection studies with other histone markers, antibody compatibility in multiplexed assays should be tested, particularly regarding species of origin to avoid cross-reactivity in secondary detection systems.
The HIST1H2AG (Ab-118) Antibody has demonstrated utility in cancer research, as evidenced by its successful application in immunohistochemistry of paraffin-embedded human breast cancer and glioma tissues . Researchers can use this antibody to investigate alterations in histone H2A expression or localization in tumor samples compared to normal tissues. This may provide insights into epigenetic dysregulation associated with tumorigenesis or cancer progression. The antibody can also be valuable in studying histone dynamics in cancer cell lines using techniques like immunofluorescence, as demonstrated in HeLa cells .
To optimize ChIP experiments using the HIST1H2AG (Ab-118) Antibody, researchers should:
Perform careful crosslinking optimization (typically 1% formaldehyde for 10-15 minutes)
Ensure thorough sonication to generate DNA fragments of 200-500 bp
Include appropriate blocking agents to reduce non-specific binding
Optimize antibody concentration through preliminary titration experiments
Include proper controls (IgG negative control and positive control using a well-characterized histone antibody)
Consider using specialized ChIP-grade protease inhibitors to preserve epitope integrity
These methodological refinements can significantly improve signal-to-noise ratio and reproducibility in ChIP experiments targeting histone H2A.
High background in IHC applications with the HIST1H2AG (Ab-118) Antibody may result from several factors:
Insufficient blocking: Extend blocking time or use alternative blocking reagents (5% BSA, 5% normal serum)
Excessive primary antibody concentration: Adhere to the recommended dilution range (1:20-200) and optimize through titration
Inadequate washing: Increase wash duration or frequency between incubation steps
Endogenous peroxidase/phosphatase activity: Ensure proper quenching steps prior to antibody incubation
Non-specific binding to highly charged tissues: Consider adding 0.1-0.3M NaCl to antibody diluent
Tissue fixation artifacts: Optimize fixation protocols or consider antigen retrieval methods
Each potential source should be systematically addressed to improve signal specificity in IHC applications.
Researchers should validate the specificity of the HIST1H2AG (Ab-118) Antibody through multiple approaches:
Western blot analysis showing a band of the expected molecular weight (around 14 kDa for histone H2A)
Peptide competition assays where pre-incubation with the immunizing peptide blocks specific signal
Genetic knockdown/knockout validation where signal reduction corresponds to decreased target expression
Correlation with orthogonal detection methods (e.g., mass spectrometry)
Comparison with other validated antibodies targeting different epitopes of the same protein
These validation strategies help ensure experimental results reflect genuine biological phenomena rather than antibody artifacts.
For optimal immunohistochemical detection using the HIST1H2AG (Ab-118) Antibody, heat-induced epitope retrieval (HIER) methods are recommended. The most effective approaches include:
Citrate buffer (pH 6.0) heating: 10mM sodium citrate buffer heated to 95-100°C for 20 minutes
EDTA buffer (pH 8.0) retrieval: 1mM EDTA heated to 95-100°C for 20 minutes
Tris-EDTA (pH 9.0) method: 10mM Tris, 1mM EDTA heated to 95-100°C for 20 minutes
The optimal method should be determined empirically, as histone epitopes can respond differently to retrieval conditions depending on fixation parameters and tissue type. Generally, EDTA-based methods may provide better results for nuclear antigens like histones.
Due to the high sequence homology between histone H2A variants, cross-reactivity may occur with this antibody. Researchers can address this by:
Performing parallel detection with variant-specific antibodies to identify potential overlapping signals
Utilizing recombinant histone H2A variants in competition assays to assess relative binding affinities
Implementing mass spectrometry validation to definitively identify detected protein species
Conducting siRNA knockdown experiments targeting specific H2A variants to determine contribution to observed signals
Comparing immunoblot patterns with predicted molecular weights of different H2A variants
These approaches help distinguish between specific detection of HIST1H2AG and potential cross-reactivity with related histone variants.
To effectively study H2A dynamics throughout the cell cycle using the HIST1H2AG (Ab-118) Antibody, researchers should:
Implement cell synchronization techniques (such as double thymidine block, nocodazole arrest, or serum starvation/stimulation)
Collect samples at defined time points representing distinct cell cycle phases
Combine immunofluorescence detection of HIST1H2AG with cell cycle markers (e.g., phospho-histone H3 for mitosis)
Utilize flow cytometry with DNA content analysis to correlate H2A staining with cell cycle position
Consider ChIP-seq approaches at different cell cycle stages to map genome-wide H2A distribution changes
Include live-cell imaging with fluorescently tagged H2A to complement antibody-based fixed-cell analysis
This multi-parameter approach allows for comprehensive analysis of both spatial and temporal dynamics of histone H2A throughout cell division.
A robust ChIP experimental design using the HIST1H2AG (Ab-118) Antibody should include the following controls:
Input control: Non-immunoprecipitated chromatin representing total DNA before IP (typically 5-10%)
Negative control: IgG from the same species as the primary antibody (rabbit)
Positive control: Known regions where H2A is expected (such as actively transcribed genes)
Negative genomic regions: Areas known to have low H2A association
Technical replicates: Multiple immunoprecipitations from the same chromatin preparation
Biological replicates: ChIP performed on independently prepared samples
Antibody titration controls: To determine optimal antibody concentration
These controls ensure experimental validity and facilitate accurate interpretation of ChIP results when studying histone H2A genomic distribution.
For successful multiplexed imaging of HIST1H2AG alongside other histone modifications, researchers should:
Select antibodies raised in different host species to enable species-specific secondary detection
If using multiple rabbit antibodies, employ sequential immunostaining with complete stripping between rounds
Utilize directly conjugated primary antibodies with non-overlapping fluorophores
Implement spectral imaging and unmixing techniques to resolve closely positioned signals
Include single-stained controls to establish bleed-through parameters
Consider tyramide signal amplification for weak signals while maintaining multiplexing capability
Validate antibody combinations for potential steric hindrance effects when epitopes are in close proximity
These approaches enable simultaneous visualization of histone H2A and its associated modifications, providing spatial context for epigenetic regulation studies.
When interpreting differential HIST1H2AG staining patterns across cell types, researchers should consider:
Intrinsic biological differences in histone H2A expression levels between cell lineages
Cell-type specific chromatin compaction states affecting epitope accessibility
Variations in histone post-translational modifications that might influence antibody binding
Technical factors such as fixation efficiency differences between cell types
Cell cycle distribution differences in heterogeneous populations
Nuclear architecture variations that affect apparent signal intensity
Quantitative approaches like fluorescence intensity measurements normalized to nuclear area or DNA content can help standardize comparisons between cell types. Validation through orthogonal methods such as western blotting of sorted populations is recommended for confirming observed differences.
When confronted with discrepancies between HIST1H2AG distribution patterns in ChIP-seq versus immunofluorescence experiments, researchers should:
Evaluate fixation differences between methods (formaldehyde for ChIP vs. paraformaldehyde/methanol for IF)
Consider epitope accessibility variances in different experimental contexts
Assess ChIP resolution limitations compared to the spatial resolution of microscopy
Implement ChIP-seq normalization strategies to account for technical biases
Use alternative antibodies targeting different epitopes of the same protein
Employ super-resolution microscopy to better align microscopy data with genomic resolution
Consider complementary approaches like CUT&RUN or CUT&Tag that offer advantages of both techniques
These methodological considerations help reconcile seemingly contradictory results across platforms and provide a more complete understanding of histone H2A biology.
Advanced computational methods can significantly enhance the analysis of data generated using the HIST1H2AG (Ab-118) Antibody:
For ChIP-seq data:
Peak calling algorithms optimized for broad histone signals
Differential binding analysis across experimental conditions
Integration with other epigenomic datasets (DNA methylation, chromatin accessibility)
Motif enrichment analysis to identify co-occurring regulatory elements
For microscopy data:
Automated nuclear segmentation for high-throughput analysis
Intensity correlation analysis with other nuclear markers
Spatial statistics to quantify distribution patterns
Machine learning classification of nuclear phenotypes
These computational approaches enable extraction of biologically meaningful patterns from complex datasets, facilitating discovery of novel insights into histone H2A function and regulation.