HIST3H2A Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HIST3H2A Antibody

HIST3H2A (Histone Cluster 3 H2A) is a replication-dependent histone variant involved in chromatin structure regulation, DNA repair, and transcriptional control. Antibodies targeting HIST3H2A are critical tools for studying its role in epigenetic regulation, cancer biology, and immune response modulation. These antibodies enable detection of HIST3H2A in diverse applications, including Western blot (WB), immunohistochemistry (IHC), and chromatin immunoprecipitation (ChIP) .

3.1. HIST3H2A in Pancreatic Cancer Prognosis

HIST3H2A is overexpressed in pancreatic cancer tissues compared to normal tissues (P<0.001P < 0.001) and correlates with poor prognosis. Key findings include:

ParameterLow HIST3H2AHigh HIST3H2AP-value
CD8+ T cells49.4%50.6%0.007
Activated CD4+ T cells47.1%52.9%0.001
M0 Macrophages34.1%65.9%0.002

3.2. Epigenetic Regulation

  • Gene Expression: HIST3H2A expression is linked to histone H3K9 acetylation, which enhances transcriptional activity in replication-dependent genes .

  • Pathway Enrichment: High HIST3H2A expression activates the JAK-STAT pathway, while low expression enriches IgA production and chemokine signaling pathways .

Immune Cell Infiltration and Pathway Analysis

CIBERSORT analysis of pancreatic cancer tissues revealed distinct immune profiles:

  • High HIST3H2A: Associated with M0 macrophages (P=0.002P = 0.002) and activated NK cells (P=0.006P = 0.006) .

  • Low HIST3H2A: Correlates with anti-tumor immune cells (e.g., CD8+ T cells), suggesting HIST3H2A as a modulator of tumor immunity .

5.1. Validation Metrics

  • Western Blot: Detects a 14–18 kDa band in human brain tissue .

  • Immunohistochemistry: Validated in formalin-fixed paraffin-embedded pancreatic cancer samples .

  • Cross-Reactivity: Confirmed in zebrafish (Danio rerio) and rat models .

5.2. Limitations

  • Species Specificity: Some antibodies (e.g., MA3-047) are human-specific .

  • Post-Translational Modifications: May require epitope-specific validation for acetylation or phosphorylation studies .

Product Specs

Buffer
The antibody is provided in PBS buffer containing 0.1% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically ship orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery information.
Synonyms
HIST3H2AHistone H2A type 3 antibody
Target Names
Uniprot No.

Target Background

Function
Histone H2A is a core component of the nucleosome. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that utilizes DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Database Links

HGNC: 20507

OMIM: 615015

KEGG: hsa:92815

STRING: 9606.ENSP00000355656

UniGene: Hs.26331

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is HIST3H2A and what cellular functions does it perform?

HIST3H2A (Histone Cluster 3, H2a) is part of the histone family responsible for the nucleosome structure of chromosomal fiber in eukaryotes. It is primarily located in the nucleus and is notably enriched in brain tissue. Histones are basic nuclear proteins that form the structural unit of chromatin called the nucleosome, consisting of approximately 146 bp of DNA wrapped around a histone octamer composed of pairs of each of the four core histones (H2A, H2B, H3, and H4) . HIST3H2A plays a critical role in DNA packaging, gene regulation, and chromosome stability. Recent research has identified its involvement in cancer progression, particularly in prostate and pancreatic cancers, where it regulates cell proliferation, migration, invasion, and the epithelial-mesenchymal transition (EMT) process .

What applications is the HIST3H2A antibody suitable for?

The HIST3H2A antibody is compatible with multiple experimental applications, making it versatile for diverse research needs. The primary validated applications include:

ApplicationRecommended DilutionsNotes
Western Blot (WB)0.01-2 μg/ml or 1:500-1:1000Successfully tested on human brain tissue and fetal human brain tissue
Immunohistochemistry (IHC)5-20 μg/mlFor tissue section analysis
Immunofluorescence/Immunocytochemistry (IF/ICC)5-20 μg/mlFor cellular localization studies
Immunoprecipitation (IP)VariableFor protein-protein interaction studies
ELISAVariableFor quantitative detection

Researchers should optimize these recommended dilutions for their specific experimental conditions to achieve optimal results .

How should the HIST3H2A antibody be stored and handled to maintain its efficacy?

For optimal antibody performance and longevity, HIST3H2A antibody should be stored at -20°C in aliquots to avoid repeated freeze/thaw cycles which can degrade antibody quality . The typical storage buffer consists of PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . When properly stored, the antibody remains stable for one year after shipment. For working solutions, it's recommended to briefly centrifuge the antibody before opening the tube and to maintain sterile conditions during handling. The antibody should not be exposed to prolonged periods at room temperature, and any unused portions should be promptly returned to proper storage conditions .

How can I validate HIST3H2A antibody specificity in my experimental system?

Validating antibody specificity is critical for ensuring reliable research outcomes. For HIST3H2A antibody validation, implement the following multi-step approach:

First, perform Western blot analysis using positive control samples (human brain tissue or fetal human brain tissue are recommended controls) . The expected molecular weight range for HIST3H2A is 14-18 kDa . Include protein ladder markers to confirm accurate size.

Second, include comparative samples with HIST3H2A knockdown or knockout (using shRNA or CRISPR-Cas9) alongside wild-type controls. This approach was effectively employed in prostate cancer cell lines, where HIST3H2A was successfully interfered within 22RV1 cells showing altered protein expression patterns compared to controls .

Third, incorporate peptide competition assays where pre-incubation of the antibody with the immunizing peptide (HIST3H2A Met1-Lys130) should abolish specific signals .

Finally, cross-validate with an alternative antibody targeting a different epitope of HIST3H2A or employ orthogonal methods such as mass spectrometry to confirm target identity in immunoprecipitated samples.

What are the recommended protocols for studying HIST3H2A's role in cancer cell proliferation and invasion?

Based on recent research methodologies, the following protocols are recommended for investigating HIST3H2A's role in cancer:

For proliferation studies, the EdU incorporation assay has proven effective. Following transfection with either HIST3H2A expression vector or shRNA (48 hours), seed cells in 24-well plates at 2 × 10^5 cells per well and incubate with 20 mM EdU for 2 hours. After fixation with 3.7% formaldehyde (15 min) and permeabilization with 0.25% Triton X-100 (15 min), process cells according to the EdU detection kit protocol. Calculate the uptake rate by determining the ratio of EdU-positive cells (red) to total Hoechst 33,258-stained cells (blue) .

For invasion and migration assessment, the Transwell assay is recommended. This has successfully demonstrated that HIST3H2A overexpression promotes invasion and migration in PC3 cells, while HIST3H2A interference inhibits these processes in 22RV1 cells .

Western blot analysis should be used to examine the expression of proliferation-related proteins (PCNA, CDK2, CDK6, cyclinD1, cyclinE1, P21) and invasion-related genes (MMP-2, MMP-9, E-cadherin, N-cadherin, Vimentin) . Changes in these markers correlate with HIST3H2A expression levels and provide mechanistic insights into its role in cancer progression.

How can I investigate the relationship between HIST3H2A expression and cancer prognosis?

To establish correlations between HIST3H2A expression and cancer prognosis, implement the following comprehensive approach:

First, analyze HIST3H2A expression levels in paired tumor and normal tissues using techniques such as qRT-PCR, Western blot, and immunohistochemistry to establish baseline expression differences .

Conduct univariate and multivariate Cox regression analyses to determine whether HIST3H2A expression is an independent prognostic factor after adjusting for clinicopathological variables such as age, gender, tumor stage, and grade .

For a deeper understanding of HIST3H2A's role in cancer biology, perform Gene Set Enrichment Analysis (GSEA) to identify enriched signaling pathways associated with differential HIST3H2A expression. Previous studies identified JAK-STAT signaling pathway enrichment in HIST3H2A high expression phenotypes in pancreatic cancer .

Finally, analyze immune cell infiltration patterns in relation to HIST3H2A expression using computational methods like CIBERSORT to estimate the relative proportions of 22 immune cell types. This approach revealed that CD8 T cells (P = .007), activated CD4 memory T cells (P = .001), and monocytes (P = .002) were more abundant in lower HIST3H2A expression groups in pancreatic cancer .

What are the common technical challenges when using HIST3H2A antibody in IHC and how can they be addressed?

When using HIST3H2A antibody for immunohistochemistry, researchers commonly encounter several technical challenges that can impact results. Here are solutions to address these issues:

For high background staining, implement more stringent blocking procedures using 5-10% normal serum from the same species as the secondary antibody for at least 1 hour at room temperature. Additionally, optimize the primary antibody concentration by testing a dilution series between 5-20 μg/ml as recommended for HIST3H2A antibody .

For weak or absent signals, employ heat-induced epitope retrieval (HIER) methods using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) to unmask antigens that may be masked during fixation. Since HIST3H2A is a nuclear protein, ensure sufficient permeabilization of the nuclear membrane using appropriate detergents like Triton X-100 .

For non-specific binding, use careful antibody titration and implement additional washing steps with PBS containing 0.05-0.1% Tween-20. Consider using protein-free blocking reagents if serum-based blockers produce excessive background.

For inconsistent results across experimental replicates, standardize tissue processing protocols, including fixation time, temperature, and reagent quality. Maintain consistent antibody lots for long-term studies and implement positive and negative controls in each experimental run.

How do I reconcile conflicting data regarding HIST3H2A's role in different cancer types?

Resolving contradictory findings about HIST3H2A's function across cancer types requires a systematic approach:

First, perform comprehensive meta-analysis of existing literature and datasets to identify consistent patterns versus context-dependent effects. Compare methodologies, cell lines, and experimental conditions across studies to identify potential sources of variability.

Second, design comparative studies using multiple cancer cell lines representing different tissue origins under identical experimental conditions. This approach helps distinguish cancer-specific from universal effects of HIST3H2A. Current research has established HIST3H2A's role in prostate cancer through studies in PC3 and 22RV1 cell lines and in pancreatic cancer through bioinformatics analyses of TCGA data .

Third, investigate tissue-specific interaction partners of HIST3H2A through co-immunoprecipitation followed by mass spectrometry or yeast two-hybrid screening. Different protein interaction networks may explain diverse functions across cancer types. Co-expression analysis in pancreatic cancer identified DCST1-AS1, HIST1H2BD, and SLC12A9-AS1 as genes positively correlated with HIST3H2A expression , which may differ in other cancers.

Fourth, examine the impact of HIST3H2A on different signaling pathways across cancer types using pathway reporter assays and inhibitor studies. The JAK-STAT pathway has been implicated in HIST3H2A's role in pancreatic cancer , but other pathways may be relevant in different cancer contexts.

Finally, consider epigenetic and post-translational modifications of HIST3H2A that may vary between cancer types, potentially leading to different functional outcomes despite similar expression levels.

How can HIST3H2A be targeted therapeutically in cancer and what assays are needed to evaluate potential inhibitors?

Developing therapeutic strategies targeting HIST3H2A requires a multi-faceted approach:

For direct targeting strategies, design and screen small molecule inhibitors that disrupt HIST3H2A's interaction with key binding partners identified through structural studies and protein-protein interaction analyses. Evaluate candidates using fluorescence polarization assays, surface plasmon resonance, or alphascreen technology to measure binding affinity and specificity.

For gene expression modulation approaches, develop siRNA, shRNA, or antisense oligonucleotides specifically targeting HIST3H2A. Evaluate knockdown efficiency through qRT-PCR and Western blotting, followed by functional assays including proliferation, migration, and invasion assessments. Previous research successfully employed shRNA to interfere with HIST3H2A expression in 22RV1 cells, resulting in decreased proliferation and invasion capacity .

For pathway-based interventions, target the JAK-STAT signaling pathway, which has been linked to HIST3H2A's function in pancreatic cancer . Test combinations of HIST3H2A inhibition with established JAK-STAT inhibitors using cell viability assays, phospho-protein analysis, and reporter gene assays.

To evaluate therapeutic efficacy, conduct xenograft studies in immunodeficient mice using cancer cell lines with modulated HIST3H2A expression. Monitor tumor growth, metastatic potential, and survival outcomes. Additionally, assess immune infiltration changes in immunocompetent mouse models, given that HIST3H2A expression levels correlate with altered immune cell populations in pancreatic cancer .

What is the role of HIST3H2A in immune regulation within the tumor microenvironment and how can it be investigated?

Recent findings suggest HIST3H2A plays a significant role in immune regulation within the tumor microenvironment. To investigate this relationship:

First, analyze correlations between HIST3H2A expression levels and immune cell infiltration patterns using computational deconvolution methods like CIBERSORT applied to bulk RNA-seq data. Previous research demonstrated that CD8 T cells (P = .007), activated CD4 memory T cells (P = .001), and monocytes (P = .002) were more abundant in tumors with lower HIST3H2A expression in pancreatic cancer .

Second, perform single-cell RNA sequencing of tumor samples stratified by HIST3H2A expression to identify cell-specific effects on the tumor microenvironment at higher resolution than bulk sequencing approaches.

Third, conduct multiplex immunofluorescence or immunohistochemistry on tissue microarrays to visualize and quantify spatial relationships between HIST3H2A-expressing cells and various immune cell populations.

Fourth, establish co-culture systems of cancer cells with modulated HIST3H2A expression alongside immune cells (T cells, macrophages, dendritic cells) to assess direct effects on immune cell activation, cytokine production, and effector functions. Measure outcomes through flow cytometry, ELISA, and functional killing assays.

Fifth, investigate the impact of HIST3H2A on immune checkpoint expression (PD-L1, CTLA-4) and response to checkpoint inhibitor therapy in preclinical models. This may provide insights into whether HIST3H2A expression could serve as a biomarker for immunotherapy response.

How might single-cell analysis techniques advance our understanding of HIST3H2A function in heterogeneous tumor populations?

Single-cell technologies offer unprecedented opportunities to unravel HIST3H2A's role in tumor heterogeneity:

Single-cell RNA sequencing (scRNA-seq) can determine if HIST3H2A expression varies across distinct cellular subpopulations within tumors and correlate this with specific transcriptional programs and cell states. This would reveal whether HIST3H2A drives particular cancer cell phenotypes or is associated with specific cancer stem cell populations.

Single-cell ATAC-seq (Assay for Transposase-Accessible Chromatin) can map chromatin accessibility in relation to HIST3H2A expression at the single-cell level, potentially uncovering how HIST3H2A influences genome-wide chromatin organization in different tumor subpopulations.

Mass cytometry (CyTOF) with antibodies against HIST3H2A and multiple cellular markers can quantify protein-level associations between HIST3H2A and other signaling molecules across thousands of individual cells. This would advance our understanding beyond the correlations observed in pancreatic cancer between HIST3H2A and pathway components like JAK-STAT .

Spatial transcriptomics can map HIST3H2A expression within the architectural context of the tumor, potentially revealing relationships between spatial location, HIST3H2A expression, and the proximity to specific microenvironmental features like vasculature or immune-rich regions.

To implement these approaches effectively, researchers should develop computational pipelines specifically designed to integrate multi-omic single-cell data with HIST3H2A expression as a central feature, potentially revealing new biological insights into its function in cancer progression.

What is the relationship between HIST3H2A and other histone variants in cancer development and how can this be studied systematically?

Investigating the interplay between HIST3H2A and other histone variants requires systematic approaches:

First, perform comprehensive expression correlation analyses across cancer types using TCGA, GEO, and other public databases to identify histone variants that consistently co-express with HIST3H2A. Previous research has identified correlations between HIST3H2A and HIST1H2BD in pancreatic cancer , suggesting potential functional relationships among histone family members.

Second, conduct ChIP-seq experiments for HIST3H2A alongside other histone variants to map genome-wide co-localization patterns and identify regions where these variants may work cooperatively or antagonistically. This should be complemented with sequential ChIP (re-ChIP) to determine if these variants co-occur at the same nucleosomes.

Third, implement CRISPR-based combinatorial perturbations (e.g., using CRISPR interference or activation) targeting HIST3H2A alongside other histone variants to uncover synthetic lethal or synergistic relationships. Monitor effects on cell viability, proliferation, migration, and gene expression.

Fourth, develop proximity-labeling approaches such as BioID or APEX2 fused to HIST3H2A to identify proteins that physically interact with HIST3H2A in living cells, potentially revealing connections to other histone variants or histone modifying enzymes.

Finally, utilize structural biology approaches including cryo-EM and X-ray crystallography to determine how HIST3H2A incorporation affects nucleosome structure compared to canonical H2A or other variants, providing mechanistic insights into its unique functions.

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 2025 TheBiotek. All Rights Reserved.