Target: Histone H2A.Z acetylated at lysine 7 (H2A.Z K7ac)
Immunogen: Synthetic peptide corresponding to residues surrounding acetylated K7 in human H2A.Z .
Host Species: Rabbit
Clonality: Polyclonal
Reactivity: Human (primary), with cross-reactivity reported in mouse and rat for some batches .
Purification: Antigen affinity-purified .
Storage: PBS with 0.03% ProClin 300 or 0.02% sodium azide; stable at -20°C for long-term storage .
| Parameter | Details |
|---|---|
| Molecular Weight | 14 kDa (observed) |
| Gene ID | 3015 (H2AFZ) |
| UniProt ID | P0C0S5 |
| Applications | WB, IHC, IF/ICC, ELISA, ChIP |
| Recommended Dilution | WB: 1:500–5,000; IHC: 1:20–200; IF/ICC: 1:50–200 |
Localizes H2A.Z in formalin-fixed paraffin-embedded (FFPE) tissues, including human skin cancer, testis, and small intestine .
Antigen retrieval recommended (citrate buffer pH 6.0 or TE buffer pH 9.0) .
Identifies H2A.Z-enriched regions at transcriptional start sites (TSS) of memory-related genes (e.g., Fos, Arc) .
H2A.Z eviction from the +1 nucleosome near TSS correlates with activation of memory-associated genes (Npas4, Egr1) and suppression of Ppp3ca .
Depletion of H2A.Z enhances fear memory consolidation in mice, linked to increased Fos expression and reduced Ppp3ca levels .
Acetylation at K7 (detected by H2AFZ Ab-7) modulates chromatin accessibility and interaction with bromodomain-containing proteins .
Competing regulators include DNA methylation, SIRT1 deacetylase, and H3K56 acetylation .
No cross-reactivity with non-acetylated H2A.Z or other acetylated histones in ELISA .
Recognizes 14 kDa band in WB across species (human, mouse, zebrafish) .
AAV-mediated H2afz knockdown in hippocampal CA1 neurons reduced H2A.Z protein by 55.8%, improving long-term fear memory .
H2AFZ (also known as H2A.Z.1) is one of two vertebrate H2A.Z histone hypervariants, with H2A.Z.2 being the other. Despite differing by only three amino acids, these hypervariants have distinct and context-specific roles in gene regulation. H2AFZ is encoded by the H2afz gene, while H2A.Z.2 is encoded by H2afv. These genes are located on different chromosomes and are driven by independent promoters . H2AFZ is significant in epigenetic research because it functions as a critical regulator of eukaryotic gene transcription, playing roles in both activation and repression of genes depending on cellular context. Research has demonstrated that H2AFZ is essential for embryonic development, as H2A.Z.1-/- mice exhibit embryonic lethality, indicating its unique functions that cannot be compensated by H2A.Z.2 .
Although H2AFZ and H2A.Z.2 differ by only three amino acids positioned far apart on their polypeptide chains, these subtle differences can lead to significant functional variations:
Structural differences: Crystal structures reveal subtle variations in the L1 loop region between H2AFZ and H2A.Z.2 nucleosomes .
Functional specificity: Microarray analyses show that H2AFZ and H2A.Z.2 regulate largely non-overlapping gene sets in neurons, particularly genes encoding synaptic proteins .
Context-dependent roles: In response to neuronal activity, rapid transcription of immediate early genes like Arc is differentially regulated by these hypervariants. For example, H2A.Z.2 depletion impairs Arc transcription under certain conditions, while both hypervariants are required in other contexts (e.g., after 48 hours of tetrodotoxin treatment) .
Developmental requirements: H2AFZ cannot be functionally replaced by H2A.Z.2 during embryonic development, highlighting non-redundant roles .
The methodological approach to studying these differences typically involves hypervariant-specific RNAi combined with gene expression analyses to distinguish their individual contributions to cellular processes.
H2AFZ antibodies are particularly valuable for:
Chromatin immunoprecipitation (ChIP) studies to identify genomic regions where H2AFZ is incorporated.
Western blotting to assess protein expression levels across different tissues or experimental conditions.
Immunofluorescence microscopy to examine nuclear localization and distribution patterns.
Flow cytometry for cell-specific analysis of H2AFZ expression.
When studying H2AFZ specifically, it's crucial to note that most commercial antibodies cannot distinguish between H2AFZ and H2A.Z.2 due to their highly similar sequences. As noted in the literature, "there are currently no antibodies available to distinguish between H2A.Z.1 and H2A.Z.2" due to the high structural similarity of their respective amino acid differences . Therefore, complementary techniques such as isoform-specific qPCR for mRNA detection should be used alongside antibody-based methods to distinguish between these hypervariants.
Since antibodies (including Ab-7) typically cannot distinguish between H2AFZ and H2A.Z.2 proteins, a combinatorial approach is recommended:
Hypervariant-specific RNA interference: Design shRNAs or siRNAs targeting unique regions of H2AFZ (H2afz) and H2A.Z.2 (H2afv) mRNAs. Validate knockdown efficiency using hypervariant-specific qPCR primers as shown in this example table from published research:
| Target | Forward primer | Reverse primer |
|---|---|---|
| H2Afz mRNA | GAAGAAAGGACAACAGAAGACTGT | CAGCTGTTAAGAGTATTTAGAGTCC |
| H2Afv mRNA | ACCCTATGCTCCCGTGTGTTAGAA | AGGCAAAGATCAGCACCAACTCTG |
Rescue experiments: After knockdown, perform rescue experiments by expressing recombinant H2AFZ or H2A.Z.2 with silent mutations that prevent targeting by RNAi to confirm specificity.
Genomic approaches: Employ CRISPR/Cas9-mediated gene editing to introduce modifications specifically in either H2afz or H2afv genes.
mRNA expression analysis: Use the hypervariant-specific primers listed above to quantify expression levels of each isoform independently .
This multi-faceted approach allows for the functional separation of these highly similar hypervariants in your experimental system.
For rigorous ChIP experiments with H2AFZ antibodies, include the following controls:
Input control: Reserve 5-10% of chromatin before immunoprecipitation to normalize for differences in starting material.
IgG control: Use an isotype-matched IgG to assess non-specific binding.
Positive control regions: Include genomic regions known to be enriched for H2AFZ, such as transcriptionally active promoters.
Negative control regions: Include regions where H2AFZ is typically absent, such as certain heterochromatic regions.
Antibody validation controls:
Knockdown verification: Perform ChIP after H2AFZ knockdown to demonstrate reduced signal.
Peptide competition: Pre-incubate antibody with excess H2AFZ peptide to block specific binding.
Hypervariant specificity controls: If attempting to distinguish H2AFZ from H2A.Z.2, validate results using complementary genetic approaches such as hypervariant-specific knockdown followed by ChIP with a general H2A.Z antibody.
Cross-validation with multiple antibodies: When possible, compare results using different H2AFZ antibodies targeting distinct epitopes.
These controls help ensure the specificity and reliability of ChIP data, particularly important given the challenge of distinguishing between highly similar histone hypervariants.
For optimal detection of H2AFZ in neuronal tissues:
Tissue collection and fixation:
For ChIP: Use 1% formaldehyde for 10-15 minutes at room temperature, followed by quenching with 125mM glycine.
For protein extraction: Flash-freeze tissue in liquid nitrogen immediately after collection.
Chromatin preparation:
For neuronal cultures: Following protocols established in neuronal studies, such as bicuculline (50μM) and 4-aminopyridine (75μM) treatment to induce gene transcription via synaptic circuits, or tetrodotoxin (1-2μM) treatment to block neuronal activity .
Cross-link cells with 1% formaldehyde for 10 minutes.
Lyse cells in appropriate buffer containing protease inhibitors.
Sonicate to obtain chromatin fragments of 200-500bp.
Protein extraction:
Use specialized extraction methods for histones, such as acid extraction (0.2N HCl).
Include protease inhibitors and histone deacetylase inhibitors to preserve post-translational modifications.
Consider subcellular fractionation to enrich nuclear proteins.
Western blotting considerations:
Use 15-18% SDS-PAGE gels to adequately resolve histone proteins.
Include appropriate loading controls such as total H2A or H3.
Immunofluorescence:
Fix neurons with 4% paraformaldehyde.
Perform antigen retrieval if necessary.
Include permeabilization step with 0.2% Triton X-100.
Block with 5% BSA or appropriate blocking solution.
This protocol has been optimized based on published neuronal studies examining H2A.Z variants and their roles in neuronal gene expression .
To analyze H2AFZ dynamics during activity-dependent transcription:
Temporal ChIP analysis:
Perform ChIP-qPCR or ChIP-seq at multiple timepoints following neuronal stimulation.
Focus on promoters and gene bodies of immediate early genes (IEGs).
Use primer sets targeting both promoter regions and gene bodies to track H2AFZ occupancy changes.
Combined approaches:
Couple ChIP with nascent RNA analysis (e.g., nuclear run-on assays or pre-mRNA qPCR) to correlate H2AFZ occupancy with transcriptional activity.
Example gene targets and primers for pre-mRNA analysis from published research:
| Target | Forward primer | Reverse primer |
|---|---|---|
| Arc pre-mRNA | GAATTTGCTATGCCAACTCACGGG | AGTCATGGAGCCGAAGTCTGCTTT |
| cFos pre-mRNA | ACAGCCTTTCCTACTACCATTCCC | CTGCACAAAGCCAAACTCACCTGT |
| Npas4 pre-mRNA | GTTGCATCAACTCCAGAGCCAAGT | ACATTTGGGCTGGACCTACCTTCA |
Context-specific stimulation paradigms:
Hypervariant-specific analysis:
Combine H2AFZ or H2A.Z.2 knockdown with ChIP-seq using a general H2A.Z antibody to identify hypervariant-specific occupancy patterns.
Monitor gene expression changes using microarray or RNA-seq following hypervariant-specific knockdown.
Analysis of chaperone interactions:
This comprehensive approach allows for detailed characterization of the dynamic relationship between H2AFZ chromatin occupancy and activity-dependent gene transcription in neurons.
Resolving contradictions between ChIP-seq and immunofluorescence data requires systematic troubleshooting:
Antibody considerations:
Epitope accessibility: Certain epitopes may be differentially accessible in fixed chromatin versus immunofluorescence preparations.
Antibody specificity: Validate antibody specificity in both applications using knockdown controls.
Cross-reactivity: Test for potential cross-reactivity with H2A.Z.2 or other histone variants.
Technical validation approaches:
Use multiple antibodies targeting different H2AFZ epitopes.
Compare results with tagged H2AFZ expression systems.
Perform ChIP-seq with alternative fixation methods or native ChIP.
Biological explanations:
Different pools: Discrepancies may reflect distinct nuclear pools of H2AFZ (e.g., nucleoplasmic versus chromatin-bound).
Post-translational modifications: Consider whether modifications affect epitope recognition differentially between methods.
Resolution differences: ChIP-seq provides genome-wide binding sites, while immunofluorescence shows nuclear distribution patterns at lower resolution.
Integration strategies:
Perform ChIP-seq and immunofluorescence under identical experimental conditions.
Use cell fractionation to distinguish soluble versus chromatin-bound H2AFZ.
Combine with super-resolution microscopy for higher-resolution localization data.
Correlate with functional genomics data to determine biological relevance of discrepant findings.
By systematically evaluating technical and biological factors, apparent contradictions can often be resolved or explained, leading to a more complete understanding of H2AFZ biology.
When interpreting H2AFZ ChIP-seq data comparing cancer and normal tissues:
Research has shown that all three H2A.Z isoforms (H2A.Z.1/H2AFZ, H2A.Z.2.1, and H2A.Z.2.2) are highly expressed in PDAC cell lines and patients, and their expression correlates with poor prognosis . This suggests that careful interpretation of cancer-specific patterns of H2AFZ distribution is essential for understanding its role in oncogenesis.
Common pitfalls and their solutions in H2AFZ antibody experiments:
Cross-reactivity with H2A.Z.2:
Epitope masking by post-translational modifications:
Challenge: H2AFZ undergoes various modifications that may mask antibody epitopes.
Solution: Use antibodies targeting different epitopes; consider modification-specific antibodies when relevant.
Poor signal-to-noise ratio in ChIP:
Challenge: High background or weak enrichment.
Solution: Optimize crosslinking time, sonication conditions, and antibody concentration; increase wash stringency; use filtered buffers.
Inconsistent results across experimental replicates:
Challenge: Variable enrichment patterns or western blot signal.
Solution: Standardize protocols for sample preparation, implement quality control metrics, and use internal controls.
Misinterpretation of functional redundancy:
Inadequate validation of knockdown efficiency:
By anticipating these common challenges and implementing appropriate controls and validation strategies, researchers can significantly improve the reliability and interpretability of H2AFZ antibody-based experiments.
Optimizing ChIP-seq for low cell number samples when studying H2AFZ:
Crosslinking optimization:
Use lower formaldehyde concentrations (0.5-0.8%) to improve chromatin fragmentation.
Shorter crosslinking times (5-8 minutes) may improve epitope accessibility.
Chromatin preparation:
Sonication: Use microTUBE devices for low-volume sonication.
Enzymatic fragmentation: Consider MNase digestion as an alternative to sonication.
Target fragment size of 200-300bp for optimal resolution.
Immunoprecipitation strategies:
Carrier approach: Add "carrier" chromatin from another species to reduce non-specific loss.
Bead optimization: Use lower bead volume and protein-low-bind tubes.
Sequential ChIP: For very low cell numbers, perform two rounds of immunoprecipitation.
Library preparation:
Use library preparation kits optimized for low input (e.g., NEBNext Ultra II).
Reduce adapter concentration to minimize adapter dimer formation.
Increase PCR cycles but monitor for amplification bias.
Bioinformatic considerations:
Implement stringent quality control.
Use peak callers optimized for low-input samples (e.g., MACS2 with appropriate parameters).
Consider joint analysis with publicly available datasets as references.
Alternative approaches:
CUT&RUN or CUT&Tag: These techniques can work with as few as 1,000 cells with higher signal-to-noise ratio than traditional ChIP.
Single-cell approaches: For heterogeneous populations, consider single-cell CUT&Tag or similar methods.
Validation:
Confirm key findings with ChIP-qPCR on independent samples.
Compare with bulk ChIP-seq results from larger samples when available.
These optimizations can help generate reliable H2AFZ binding profiles from limited biological materials such as sorted neuronal populations or patient-derived samples.
Recent research suggests important connections between H2A.Z variants and H3K27M mutations in pediatric high-grade gliomas:
Functional interactions:
Chromatin remodeling interplay:
H3K27M mutations are known to inhibit the activity of Polycomb Repressive Complex 2 (PRC2), leading to global reduction of H3K27me3.
H2A.Z may influence the distribution or activity of residual PRC2 in H3K27M mutant cells.
The incorporation of H2A.Z might affect the spread of H3K27 acetylation, which is pervasive in H3K27M gliomas .
Therapeutic implications:
Research approaches:
ChIP-seq for H2AFZ and H3K27M in patient-derived glioma cells
Analysis of H2AFZ localization in relation to H3K27ac and H3K27me3 domains
Functional studies manipulating H2AFZ levels in H3K27M mutant cells
Testing combinatorial approaches targeting both H2AFZ-related pathways and histone acetylation
This emerging area represents a promising direction for understanding how histone variants and oncohistone mutations cooperate in driving aggressive pediatric brain tumors.
Distinguishing genome-wide localization patterns of these highly similar hypervariants requires innovative approaches:
Tagged variant expression systems:
Generate cell lines expressing epitope-tagged versions of H2AFZ or H2A.Z.2 (e.g., FLAG, HA, or biotin tags).
Ensure tagged proteins are expressed at near-endogenous levels.
Perform ChIP-seq using tag-specific antibodies.
Hypervariant depletion approach:
Perform ChIP-seq with a general H2A.Z antibody in control cells.
Specifically deplete H2AFZ or H2A.Z.2 using validated shRNAs.
Repeat ChIP-seq and identify regions with differential signal reduction.
This approach has been successfully used to identify hypervariant-specific gene regulation in neurons .
CUT&RUN or CUT&Tag with recombinant proteins:
Express and purify recombinant antibody-binding domains fused to MNase or Tn5.
Target these fusion proteins to epitope-tagged hypervariants.
These methods offer improved signal-to-noise ratio compared to traditional ChIP.
Computational integration approaches:
Correlate ChIP-seq data with hypervariant-specific transcriptome changes after knockdown.
Integrate with other epigenomic marks that may associate differentially with each hypervariant.
Apply machine learning to identify subtle differences in binding patterns.
Mass spectrometry-based approaches:
Perform chromatin enrichment for proteomics (ChEP) followed by mass spectrometry.
Identify peptides unique to each hypervariant.
Quantify hypervariant-specific peptides across different chromatin fractions.
These complementary approaches can help delineate the potentially distinct functions and genomic distributions of H2AFZ and H2A.Z.2, leading to a better understanding of their context-specific roles in gene regulation.
Research on H2AFZ has significant implications for therapeutic resistance in pancreatic cancer:
Mechanistic connections to chemoresistance:
Cellular senescence pathways:
H2A.Z knockdown induces a senescent phenotype in PDAC cells, including:
This implies that H2AFZ overexpression helps cancer cells overcome the senescence barrier.
Research methodologies:
Analyzing H2AFZ binding at genes involved in chemoresistance pathways using ChIP-seq
Profiling transcriptome changes in gemcitabine-resistant versus sensitive cells after H2AFZ modulation
Investigating combinatorial approaches targeting both H2AFZ-related pathways and conventional chemotherapy
Examining post-translational modifications of H2AFZ in resistant versus sensitive cells
Potential therapeutic strategies:
Developing small molecules that disrupt H2AFZ incorporation into chromatin
Targeting H2AFZ-specific chaperone proteins
Using H2AFZ expression as a biomarker for patient stratification
Combining H2AFZ inhibition with conventional chemotherapy or targeted agents
Experimental models:
Patient-derived xenografts with varying H2AFZ levels
Isogenic cell lines with modified H2AFZ expression
In vivo models examining tumor growth after H2AFZ depletion
Research has demonstrated that depletion of H2A.Z isoforms reduces tumor size in mouse xenograft models and sensitizes PDAC cells to gemcitabine . These findings position H2AFZ as a potential diagnostic biomarker and therapeutic target for PDAC, particularly in the context of chemoresistance.