H2AFZ (Ab-7) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Properties

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 .

ParameterDetails
Molecular Weight14 kDa (observed)
Gene ID3015 (H2AFZ)
UniProt IDP0C0S5
ApplicationsWB, IHC, IF/ICC, ELISA, ChIP
Recommended DilutionWB: 1:500–5,000; IHC: 1:20–200; IF/ICC: 1:50–200

Western Blot (WB)

  • Detects endogenous H2A.Z in human brain, kidney, and cancer tissues .

  • Validated in lysates from HeLa, HepG2, and HL60 cells .

Immunohistochemistry (IHC)

  • 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) .

Immunofluorescence (IF)

  • Labels H2A.Z in nuclear compartments of HeLa cells .

Chromatin Immunoprecipitation (ChIP)

  • Identifies H2A.Z-enriched regions at transcriptional start sites (TSS) of memory-related genes (e.g., Fos, Arc) .

Role in Transcriptional Regulation

  • 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 .

Epigenetic Modifications

  • 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 .

Specificity

  • 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) .

Functional Studies

  • AAV-mediated H2afz knockdown in hippocampal CA1 neurons reduced H2A.Z protein by 55.8%, improving long-term fear memory .

Limitations

  • Limited reactivity in non-human primates and zebrafish .

  • Batch-dependent variability in IHC performance noted .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery information.
Synonyms
H2A histone family member Z antibody; H2A.z antibody; H2A/z antibody; H2afz antibody; H2AZ antibody; H2AZ_HUMAN antibody; Histone H2A.Z antibody; MGC117173 antibody
Target Names
Uniprot No.

Target Background

Function
Histone variant H2A.Z is a replacement for conventional H2A in a subset of nucleosomes. Nucleosomes function in wrapping and compacting DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling. H2A.Z may be involved in the formation of constitutive heterochromatin and could be essential for chromosome segregation during cell division.
Gene References Into Functions
  1. H2A.Z is associated with epigenetic gene activation in prostate cancer. Acetylated H2A.Z plays a role in the activation of newly formed enhancers in prostate cancer. PMID: 29116202
  2. H2A.Z is overexpressed in intrahepatic cholangiocarcinoma (ICC), and its expression correlates with poor prognosis in ICC patients. H2A.Z regulates cell proliferation in vitro and in vivo through H2A.Z/S-phase kinase-associated protein 2/p27/p21 signaling. PMID: 29532867
  3. This study identifies GAS41 as a histone acetylation reader that promotes histone H2A.Z deposition in non-small cell lung cancer. PMID: 29437725
  4. Two possible modes of pioneering associated with combinations of H2A.Z and p300/CBP at nucleosome-occupied enhancers. PMID: 28301306
  5. Results indicate that accumulation of H2A.Z within repressed genes can also be a consequence of the repression of gene transcription rather than an active mechanism required to establish the repression. PMID: 29036442
  6. Findings suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis, where it plays a role in accelerating cell cycle transition and epithelial-mesenchymal transition (EMT) during hepatocarcinogenesis. PMID: 26863632
  7. Crystal structure results show that the flexibility of the H2A.Z L1 loop is crucial for forming the stable heterotypic H2A.Z/H2A nucleosome. PMID: 27358293
  8. Monoubiquitination of histone H2B prevents the eviction of histone variant H2A.Z from inducible enhancers. PMID: 27692985
  9. PWWP2A is a novel H2A.Z-specific multivalent chromatin binder, establishing a link between H2A.Z, chromosome segregation, and organ development. PMID: 28645917
  10. SMYD3-mediated H2A.Z.1K101 dimethylation activates cyclin A1 expression and contributes to the proliferation of breast cancer cells. PMID: 27569210
  11. Results suggest that the N-terminal tail of H2A.Z makes unique contributions to epigenetic events. PMID: 26833946
  12. The H2AFZ gene may confer a risk for schizophrenia and contribute to the impairment of executive function in Han Chinese patients with schizophrenia. PMID: 26246156
  13. The 2.7-A-resolution crystal structure of the human YL1-H2A.Z-H2B complex shows that YL1 binding, similar to ANP32E binding, triggers an extension of the H2A.Z alphaC helix. PMID: 26974126
  14. The primary function of INO80 and ANP32E in promoting homologous recombination is the removal of H2A.Z from chromatin. PMID: 26142279
  15. Results demonstrate a male-selective association of the H2AFZ gene with schizophrenia, indicating that modifications of the H2AFZ signaling pathway warrant further investigation in terms of the pathophysiology of schizophrenia. PMID: 25392085
  16. Dynamic modulation of H2A.Z exchange and removal by Anp32e highlights the importance of the nucleosome surface and nucleosome dynamics in processing the damaged chromatin template during double-strand break (DSB) repair. PMID: 26034280
  17. H2A.Z.2 is a mediator of cell proliferation and drug sensitivity in malignant melanoma. PMID: 26051178
  18. The predictive values regarding low expressions of H2AFZ and CASP8AP2 and high white blood cell count suggest that these features could help identify patients at greater risk of relapse. PMID: 24397596
  19. Anp32e may help resolve non-nucleosomal H2A.Z aggregates and facilitate the removal of H2A.Z at the +1 nucleosomes, potentially aiding RNA polymerase II in overcoming the first nucleosomal barrier. PMID: 24613878
  20. H2A.Z is deposited at promoters and enhancers in embryonic stem cells and neural progenitors, correlating strongly with H3K4 methylation. H2A.Z is present at poised promoters with bivalent chromatin and at active promoters with H3K4 methylation, but is absent from stably repressed promoters enriched for H3K27 trimethylation. PMID: 23034477
  21. Depletion of H2A.Z in the osteosarcoma U2OS cell line and in immortalized human fibroblasts does not affect DNA double-strand breaks repair parameters while impacting clonogenic ability and cell cycle distribution. PMID: 24240188
  22. Mutational analysis reveals that the amino-acid difference at position 38 is at least partially responsible for the structural polymorphism in the L1 loop region of H2A.Z.1 and H2A.Z.2. PMID: 24311584
  23. Sirt1 and H2A.Z deregulation in prostate cancer are related. Epigenetic mechanisms, primarily histone post-translational modifications, likely impair sirt1-mediated downregulation of H2A.Z through proteasome-mediated degradation. PMID: 24127549
  24. H2A.Z-dependent crosstalk between enhancer and promoter regulates cyclin D1 expression. PMID: 23108396
  25. SETD6 monomethylates H2AZ on lysine 7. PMID: 23324626
  26. Histone deacetylase inhibitors (HDACi) induce p21 transcription and reduce cell proliferation of MDA-MB231, an ERalpha-negative mammary tumor cell line, in an H2A.Z-dependent manner. PMID: 23349794
  27. Histone H2A.Z binds specifically to ST1926. PMID: 23245330
  28. Age-dependent p400 downregulation and loss of H2A.Z localization may contribute to the onset of replicative senescence through sustained high rates of p21 transcription. PMID: 23146670
  29. H2A.Z exchange promotes specific patterns of histone modification and reorganization of the chromatin architecture, leading to the assembly of a chromatin template that is an efficient substrate for the DNA double-strand break repair machinery. PMID: 23122415
  30. ZNF24 may be involved in transcriptional regulation of genes associated with oncogenesis through its interaction with H2A.Z. PMID: 22678762
  31. Incorporation of the histone variant H2A.Z at the promoter regions of PPARgamma target genes by p400/Brd8 is essential for fat cell differentiation. PMID: 23064015
  32. Nucleosomes containing H2AZ are primarily composed of H4 K12ac and H3 K4me3 but not H3 K36me3. PMID: 22393239
  33. The short forms of H2A.Z in both yeast and human cells are more loosely associated with chromatin than the full-length proteins, indicating a conserved function for the H2A.Z C-terminal tail in regulating the association of H2A.Z with nucleosomes. PMID: 22493515
  34. Acetylation of H2A.Z is a key modification associated with gene activity in normal cells and epigenetic gene deregulation in tumorigenesis. PMID: 21788347
  35. H2A.Z is maintained during mitosis and marks the +1 nucleosome of active genes, which shifts during mitosis, resulting in occupancy at the transcriptional start site and a reduced nucleosome-depleted region. PMID: 20864037
  36. This review provides an overview of H2A.Z biology and presents hypotheses that could reconcile contradictory reports regarding the influence of H2A.Z on nucleosome stability. PMID: 20364108
  37. Estrogen Receptor alpha directly associates with the H2A.Z promoter, consequently modulating its expression. PMID: 20023423
  38. Chromatin remodeling at the c-myc gene involves the local exchange of histone H2A.Z. PMID: 15878876
  39. Neither H2AZ itself nor other features of the H2AZ-containing nucleosome spread to neighboring nucleosomes in vivo, arguing against a role for H2AZ as a self-perpetuating epigenetic mark. PMID: 16809769
  40. The essential histone variant H2A.Z is identified as a new structural component of the centromere. PMID: 17194760
  41. Monoubiquitylation of H2A.z distinguishes its association with euchromatin or facultative heterochromatin. PMID: 17636032
  42. Upon DNA damage, histone H2A.Z is first evicted from the p21 promoter, followed by the recruitment of the Tip60 histone acetyltransferase to activate p21 transcription. PMID: 17671089
  43. Histone variant H2A.Z is associated with breast cancer progression. PMID: 18414489
  44. H2A.Z nucleosomes protect only approximately 120 bp of DNA from MNase digestion and exhibit specific sequence preferences, suggesting a novel mechanism of nucleosome organization for the H2A.Z variant. PMID: 19246569
  45. Both genetic and epigenetic features likely contribute to targeting H2A.Z to distinct chromatin loci. PMID: 19261190
  46. The nucleosome destabilizing effect of H2A.Z acetylation occurs synergistically with the acetylation of the rest of the core histones. PMID: 19385636
  47. H2A.Z is incorporated into the promoter regions of estrogen receptor (ERalpha) target genes only upon gene induction, and that, in a cyclic pattern. PMID: 19515975
  48. Upon gene induction, human H2A.Z associates with gene promoters and assists in recruiting the transcriptional machinery. PMID: 19834540
  49. Both H2A.Z and H3.3 affect nucleosome positioning, either creating new positions or altering the relative occupancy of existing nucleosome position space. Only H2A.Z-containing nucleosomes exhibit altered linker histone binding. PMID: 19856965

Show More

Hide All

Database Links

HGNC: 4741

OMIM: 142763

KEGG: hsa:3015

STRING: 9606.ENSP00000296417

UniGene: Hs.119192

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is H2AFZ and why is it significant in epigenetic research?

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 .

How do H2AFZ and H2A.Z.2 differ structurally and functionally?

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.

What are the recommended applications for H2AFZ (Ab-7) antibodies in research?

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.

How should I design experiments to specifically examine H2AFZ function as distinct from H2A.Z.2?

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:

TargetForward primerReverse primer
H2Afz mRNAGAAGAAAGGACAACAGAAGACTGTCAGCTGTTAAGAGTATTTAGAGTCC
H2Afv mRNAACCCTATGCTCCCGTGTGTTAGAAAGGCAAAGATCAGCACCAACTCTG
  • 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.

What controls should be included when using H2AFZ antibodies in ChIP experiments?

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.

What is the optimal sample preparation protocol for detecting H2AFZ in neuronal tissues?

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 .

How can I analyze H2AFZ dynamics during activity-dependent gene transcription in neurons?

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:

    TargetForward primerReverse primer
    Arc pre-mRNAGAATTTGCTATGCCAACTCACGGGAGTCATGGAGCCGAAGTCTGCTTT
    cFos pre-mRNAACAGCCTTTCCTACTACCATTCCCCTGCACAAAGCCAAACTCACCTGT
    Npas4 pre-mRNAGTTGCATCAACTCCAGAGCCAAGTACATTTGGGCTGGACCTACCTTCA
  • Context-specific stimulation paradigms:

    • Synaptic stimulation: Use bicuculline (50μM) with 4-aminopyridine (75μM).

    • Extrasynaptic stimulation: Apply tetrodotoxin (1-2μM) with phorbol ester myristate (1μM).

    • Intranuclear stimulation: Use 5,6-dichloro-1-β-D-ribofuranosyl-benzimidazole (DRB, 100μM) alone or with triptolide (250-500nM) .

  • 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:

    • Consider the role of H2A.Z chaperones like ANP32E, which may differentially affect H2AFZ versus H2A.Z.2 deposition or removal .

This comprehensive approach allows for detailed characterization of the dynamic relationship between H2AFZ chromatin occupancy and activity-dependent gene transcription in neurons.

How do I resolve contradictory data between ChIP-seq and immunofluorescence results for H2AFZ localization?

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.

What are the key considerations when interpreting H2AFZ ChIP-seq data in cancer versus normal tissues?

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.

What are common pitfalls in H2AFZ antibody-based experiments and how can I overcome them?

Common pitfalls and their solutions in H2AFZ antibody experiments:

  • Cross-reactivity with H2A.Z.2:

    • Challenge: Currently available antibodies cannot distinguish between H2AFZ and H2A.Z.2 due to their high sequence similarity .

    • Solution: Use hypervariant-specific knockdown strategies coupled with general H2A.Z antibodies, or use mRNA-based methods (qPCR) to distinguish between variants.

  • 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:

    • Challenge: Assuming H2AFZ and H2A.Z.2 have identical functions.

    • Solution: Perform hypervariant-specific knockdown and rescue experiments to identify unique roles; examine context-dependent functions across different cell types or stimulation paradigms .

  • Inadequate validation of knockdown efficiency:

    • Challenge: Incomplete validation of H2AFZ-specific depletion.

    • Solution: Use hypervariant-specific primers to confirm knockdown at the mRNA level; for protein, verify total H2A.Z reduction after hypervariant-specific knockdown .

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.

How can I optimize ChIP-seq protocols for low cell number samples when studying H2AFZ?

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.

How might H2AFZ be involved in the regulation of H3K27M mutant protein in pediatric gliomas?

Recent research suggests important connections between H2A.Z variants and H3K27M mutations in pediatric high-grade gliomas:

  • Functional interactions:

    • H2A.Z histone variants appear to facilitate HDAC inhibitor-dependent removal of H3.3K27M mutant protein in pediatric high-grade glioma cells .

    • This suggests a potentially targetable relationship between histone variants and oncohistone mutations.

  • 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:

    • The finding that HDAC inhibitors can reduce H3.3K27M protein levels (up to 80%) in multiple glioma cell lines suggests that targeting histone acetylation pathways may be therapeutically relevant .

    • Understanding how H2AFZ contributes to this process could identify new therapeutic targets.

  • 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.

What methodological approaches can distinguish the genome-wide localization patterns of H2AFZ versus H2A.Z.2?

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.

How can H2AFZ research contribute to understanding therapeutic resistance in pancreatic cancer?

Research on H2AFZ has significant implications for therapeutic resistance in pancreatic cancer:

  • Mechanistic connections to chemoresistance:

    • H2AFZ is highly expressed in pancreatic ductal adenocarcinoma (PDAC) cell lines and patients, correlating with poor prognosis .

    • Depletion of H2A.Z isoforms sensitizes PDAC cells to gemcitabine, the first-line chemotherapy .

    • This suggests that H2AFZ may contribute to chemoresistance mechanisms.

  • Cellular senescence pathways:

    • H2A.Z knockdown induces a senescent phenotype in PDAC cells, including:

      • Cell cycle arrest in G2/M phase

      • Increased expression of CDKN2A/p16

      • Enhanced SA-β-galactosidase activity

      • Increased interleukin-8 production

    • 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.

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.