Target: HIST1H2AG (Histone Cluster 1 H2A Family Member G), a core histone involved in nucleosome assembly and gene regulation .
Immunogen: Synthetic peptide sequence around K118 of human HIST1H2AG .
Host: Rabbit .
Clonality: Polyclonal .
Accession Number: UniProt P0C0S8 .
The antibody has been rigorously validated across multiple platforms:
Studies using H2A-targeting antibodies (including HIST1H2AG) reveal critical biological insights:
Vendor | Catalog # | Applications | Species Reactivity | Price Range |
---|---|---|---|---|
Biomatik | CAC15621 | ELISA, WB, IHC, IF, IP | Human | $200–$400 |
Biomatik | CAC15633 | ELISA, IHC, IF, ChIP | Human | $250–$450 |
Active Motif | 39235 | WB, ChIP | Budding yeast, Human | $500–$800 |
Cell Signaling | #2578 | WB, IP, IHC | Human, Mouse, Rat, Zebrafish | $400–$600 |
The HIST1H2AG (Ab-118) Polyclonal Antibody has been validated for multiple applications in molecular and cellular biology research. According to product documentation, this antibody has been specifically validated for ELISA (Enzyme-Linked Immunosorbent Assay), Western Blotting (WB), Immunohistochemistry (IHC), Immunofluorescence (IF), Immunoprecipitation (IP), and Chromatin Immunoprecipitation (ChIP). This versatility makes it suitable for diverse experimental paradigms investigating histone modifications, chromatin structure, and epigenetic regulation .
The HIST1H2AG (Ab-118) antibody is a rabbit-derived polyclonal IgG antibody specifically targeting the human HIST1H2AG protein. It was developed using a peptide sequence centered around the Lysine 118 residue of human Histone H2A type 1 as the immunogen. The antibody recognizes the target protein with accession number P0C0S8. For optimal results, recommended dilutions vary by application: ELISA (1:2000-1:10000) and IHC (1:10-1:100). The antibody is intended exclusively for research purposes and should not be used in diagnostic procedures .
For optimal IHC results with HIST1H2AG (Ab-118) antibody, a systematic optimization approach is recommended. Begin with antigen retrieval testing, comparing heat-induced epitope retrieval (HIER) methods using citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0) to determine which better exposes the histone epitopes. Antibody titration should start with the manufacturer's recommended dilution range (1:10-1:100) and be adjusted based on signal-to-noise ratio. For chromatin-associated proteins like histones, extended blocking steps (1-2 hours) with BSA/serum are crucial to minimize background. Include both positive controls (tissues known to express HIST1H2AG) and negative controls (primary antibody omission or non-immune IgG). Counterstain selection is also important - hematoxylin provides good nuclear contrast but should be optimized to not mask the specific antibody signal .
When conducting Chromatin Immunoprecipitation (ChIP) assays with the HIST1H2AG (Ab-118) antibody, multiple rigorous controls are essential to ensure data validity. Input control (pre-immunoprecipitated chromatin) must be processed in parallel to normalize enrichment calculations. A negative control using non-immune rabbit IgG should be included to establish background binding levels. Positive controls targeting known HIST1H2AG-enriched genomic regions should verify antibody functionality. For quantitative ChIP analyses, standard curves using serial dilutions of input DNA are necessary for accurate quantification. Technical replicates (minimum triplicate) and biological replicates (different cell preparations) are required for statistical validity. Finally, a spike-in normalization approach using chromatin from a different species can correct for technical variations between samples .
Recent research has revealed an unexpected connection between histone H2A-reactive antibodies and HIV-1 neutralization. Studies have demonstrated that certain autoreactive B cells that produce antibodies against histone H2A also generate antibodies capable of neutralizing HIV-1, including multiple clades of tier 2 HIV-1 variants. This cross-reactivity appears to involve molecular mimicry between histone epitopes and HIV-1 envelope structures. Longitudinal studies of HIV-1 infected individuals showed that those generating neutralizing antibody responses often exhibit characteristics of impaired immune tolerance, including increased levels of serum autoantibodies. This suggests that H2A-reactive B cells, normally silenced by immune tolerance mechanisms, harbor the potential to produce broadly neutralizing antibodies against HIV-1 when tolerance is broken .
In healthy individuals, H2A-reactive B cells are maintained in a state of functional anergy as part of normal immune tolerance mechanisms. Flow cytometric analysis has revealed that these autoreactive B cells are present in peripheral B cell populations but display increased expression of inhibitory mediators such as CD5 and PTEN phosphatase. Functionally, they fail to mobilize calcium upon immunoreceptor stimulation, a characteristic marker of B cell anergy. This anergic state prevents potentially harmful autoantibody production. In contrast, under autoimmune conditions or when peripheral tolerance is experimentally or genetically impaired, these restrictions are lifted. Specifically, toll-like receptor stimulation or provision of CD4 T cell help can break tolerance and induce the production of H2A-reactive antibodies in vitro. This regulatory mechanism represents an important balance between preventing autoimmunity and maintaining a potentially useful reservoir of B cells that could produce broadly neutralizing antibodies against pathogens like HIV-1 .
Activating H2A-reactive B cells for therapeutic applications requires targeted methodologies to overcome their anergic state while controlling potential autoimmune effects. Several experimental approaches have shown promise: (1) Toll-like receptor (TLR) agonist stimulation, particularly TLR7 and TLR9 ligands, can effectively break B cell anergy and induce antibody production; (2) CD40 signaling stimulation using agonistic anti-CD40 antibodies or CD40L-expressing cells provides critical co-stimulation needed for these anergic B cells; (3) T cell help from CD4+ T cells of autoimmune-prone mice has been demonstrated to activate these cells, suggesting engineered T cells could provide controlled activation; (4) Cytokine supplementation, especially IL-4, IL-21, and BAFF, supports survival and differentiation of activated H2A-reactive B cells. For therapeutic applications, these approaches must be carefully calibrated to induce sufficient activation for HIV-1 neutralizing antibody production while incorporating regulatory checkpoints to prevent unchecked autoantibody production that could lead to autoimmune pathologies .
Detecting post-translational modifications (PTMs) of HIST1H2AG via Western blot requires specialized methodological adaptations. First, extraction protocols must preserve PTMs - use specialized histone extraction buffers containing appropriate deacetylase, phosphatase, and protease inhibitors (e.g., sodium butyrate, sodium fluoride, and PMSF). During sample preparation, avoid excessive heat or reducing conditions that may disrupt certain PTMs. For gel electrophoresis, high-percentage (15-18%) SDS-PAGE gels or specialized Triton-Acid-Urea (TAU) gels provide better resolution of histone variants and their modifications. Transfer conditions should be optimized for small, basic proteins (use PVDF membranes with 0.2μm pore size and methanol-containing transfer buffer). Blocking requires careful consideration - milk contains phosphatases and should be avoided when detecting phosphorylated histones; use 3-5% BSA instead. When probing with HIST1H2AG (Ab-118) antibody, combine with specific anti-PTM antibodies (e.g., anti-acetyl lysine) for co-localization studies to definitively identify modified HIST1H2AG. Finally, signal detection should employ highly sensitive chemiluminescent or fluorescent systems to detect potentially low-abundance modified forms .
Resolving contradictory HIST1H2AG ChIP-seq data between experimental conditions requires systematic troubleshooting and methodological standardization. Begin with antibody validation - confirm the HIST1H2AG (Ab-118) antibody maintains specificity across all experimental conditions by performing Western blots on nuclear extracts from each condition. Standardize chromatin preparation protocols, as chromatin accessibility differences can significantly impact ChIP efficiency - adjust fixation times and sonication parameters to achieve consistent chromatin fragmentation across conditions. Implement spike-in normalization using exogenous chromatin (e.g., Drosophila chromatin with Drosophila-specific antibody) to correct for technical variations. Computational analysis should include batch effect correction algorithms when comparing datasets generated at different times. Biological validation of differential binding sites using orthogonal methods such as CUT&RUN or targeted ChIP-qPCR is essential. Finally, consider the biological context - true differential binding may reflect condition-specific chromatin states rather than technical artifacts, which can be confirmed by correlating binding changes with other epigenetic marks, transcriptional activity, or chromatin accessibility data .
Designing effective multiplexed immunofluorescence experiments to study HIST1H2AG in relation to other histone modifications requires careful planning of antibody combinations and detection strategies. First, select compatible primary antibodies raised in different host species (e.g., HIST1H2AG (Ab-118) from rabbit combined with mouse anti-H3K27me3 and goat anti-H2AK119ub) to avoid cross-reactivity. For simultaneous detection of multiple rabbit-derived antibodies, implement sequential staining with tyramide signal amplification (TSA) - this allows antibody stripping while preserving amplified fluorescent signal from previous rounds. Spectral unmixing microscopy systems should be used to separate closely overlapping fluorophore emissions. For quantitative analysis, include internal calibration standards such as fluorescent beads or reference cell populations. Counterstain with DAPI and include architectural markers (e.g., anti-lamin B1) to provide nuclear context. Controls must include single-color stainings to establish bleed-through profiles and antibody competition assays to verify independent binding. For analysis, employ machine learning algorithms trained on expert-annotated images to automate colocalization quantification across large datasets, allowing robust statistical assessment of spatial relationships between HIST1H2AG and other histone modifications .
Studying HIST1H2AG dynamics during cell cycle progression requires integrated methodological approaches that synchronize cell populations while preserving native histone states. Begin with minimally disruptive synchronization protocols - serum starvation/release for G0/G1, double thymidine block for G1/S boundary, or RO-3306 for G2/M boundary - and validate synchronization efficiency using flow cytometry with propidium iodide staining. For live-cell imaging, create stable cell lines expressing fluorescently-tagged HIST1H2AG (ensuring the tag doesn't disrupt nucleosome incorporation) and combine with cell cycle markers like PCNA-RFP (S-phase) or cyclin B1-GFP (G2/M). When using fixed cells, combine HIST1H2AG (Ab-118) antibody immunofluorescence with EdU pulse-labeling (S-phase) and phospho-histone H3 (Ser10) staining (mitosis). For biochemical analyses, implement FACS-based sorting of live cells using Hoechst staining or cell-permeable Cdt1/geminin reporters to isolate pure populations at specific cell cycle stages before performing Western blot or ChIP analyses with the HIST1H2AG antibody. Quantitative image analysis should measure both total HIST1H2AG levels and subnuclear distribution patterns, as redistribution often occurs during replication and mitosis without total protein level changes .
HIST1H2AG antibodies offer valuable tools for investigating chromatin reorganization during DNA damage response (DDR). For spatiotemporal analysis, combine immunofluorescence using HIST1H2AG (Ab-118) antibody with markers of DNA damage (γH2AX, 53BP1) following controlled damage induction (ionizing radiation, laser micro-irradiation, or radiomimetic drugs). Live-cell imaging can be performed using fluorescently-tagged HIST1H2AG combined with damage sensors to track real-time chromatin dynamics at damage sites. For biochemical approaches, employ sequential ChIP (HIST1H2AG followed by DDR factors) to identify genomic regions where both co-occur. FAIRE-seq or ATAC-seq can be performed before and after damage induction to correlate HIST1H2AG occupancy with chromatin accessibility changes. High-resolution microscopy techniques such as STORM or PALM enable nanoscale visualization of HIST1H2AG redistribution relative to damage sites. Finally, mass spectrometry analysis of purified HIST1H2AG following damage can identify damage-specific post-translational modifications. These multi-dimensional approaches reveal how HIST1H2AG contributes to the chromatin environment that facilitates DNA repair processes .
Investigating HIST1H2AG in the context of three-dimensional chromatin architecture requires integration of genomic, microscopic, and computational methodologies. Chromosome Conformation Capture techniques (Hi-C, Micro-C) combined with HIST1H2AG ChIP-seq can identify correlations between HIST1H2AG enrichment and topologically associating domains (TADs) or chromatin loops. For direct visualization, implement super-resolution microscopy (STORM, PALM) using HIST1H2AG (Ab-118) antibody together with FISH probes targeting specific genomic regions to correlate HIST1H2AG distribution with spatial positioning of DNA sequences. Live-cell imaging of fluorescently-tagged HIST1H2AG can track dynamic changes in chromatin compartmentalization. Proximity ligation assays (PLA) between HIST1H2AG and architectural proteins (CTCF, cohesin components) can identify physical associations involved in chromatin looping. For functional analysis, perform HIST1H2AG depletion followed by Hi-C to determine its requirement for maintaining specific chromatin interactions. Computational integration of these datasets should employ multivariate statistical approaches to identify significant correlations between HIST1H2AG distribution and 3D genome features. These approaches collectively reveal how HIST1H2AG contributes to higher-order genome organization .
Integrating single-cell technologies with HIST1H2AG antibody-based assays enables unprecedented insights into cell-to-cell variability in histone dynamics. For single-cell genomics applications, CUT&Tag or CUT&RUN protocols can be adapted using HIST1H2AG (Ab-118) antibody and compatible with single-cell workflows, allowing genome-wide profiling of HIST1H2AG distribution in individual cells. Single-cell proteomics approaches include mass cytometry (CyTOF) with metal-conjugated HIST1H2AG antibodies to quantify protein levels alongside dozens of other cellular markers. For microscopy-based approaches, implement imaging mass cytometry or multiplexed ion beam imaging (MIBI) to visualize HIST1H2AG distribution in tissue contexts with subcellular resolution. Microfluidic platforms enable combined immunostaining and transcriptome analysis (CITE-seq) by using oligonucleotide-tagged HIST1H2AG antibodies, correlating histone variant distribution with gene expression in the same cells. Computational integration requires specialized algorithms for imputation and batch correction across modalities. These integrated approaches reveal how HIST1H2AG heterogeneity contributes to functional diversity within seemingly homogeneous cell populations .