H2AFZ (Ab-11) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to H2AFZ (Ab-11) Antibody

The H2AFZ (Ab-11) Antibody is a rabbit-derived polyclonal antibody targeting the lysine 11 (K11) post-translational modification site of histone H2A.Z (encoded by the H2AFZ gene). This antibody is specifically designed for detecting acetylated H2A.Z isoforms, which play critical roles in chromatin remodeling, transcriptional regulation, and cellular differentiation . Its development addresses the need for isoform-specific tools to study H2A.Z's distinct functions in normal and pathological states, particularly in cancer research .

Antibody Characteristics

Key specifications of the H2AFZ (Ab-11) Antibody are summarized below:

PropertyDetails
TargetAcetylated H2A.Z (Lys 11)
Host SpeciesRabbit
ClonalityPolyclonal
ReactivityHuman
ApplicationsELISA, Immunofluorescence/Immunocytochemistry (IF/ICC)
Recommended DilutionsIF/ICC: 1:50–1:200
ImmunogenSynthetic peptide around K11 of human H2A.Z
Storage-20°C in 50% glycerol and 0.03% Proclin-300 preservative
UniProt IDP0C0S5

This antibody has been validated for specificity in detecting acetylated H2A.Z via dot blot and immunocytochemistry .

Role in Cancer Biology

H2AFZ overexpression is linked to oncogenesis and poor prognosis across multiple cancers:

Functional Insights

  • Chromatin Regulation: H2A.Z regulates Wnt signaling and intestinal cell differentiation by suppressing CDX2/HNF1α-dependent gene activation .

  • Immune Modulation: High H2AFZ expression correlates with increased immune checkpoint molecules (e.g., PD-1, TIM3) in HCC, suggesting sensitivity to immunotherapy .

Applications in Experimental Research

The H2AFZ (Ab-11) Antibody enables:

  • Epigenetic Studies: Detects acetylation at K11, a marker of transcriptionally active chromatin .

  • Cancer Biomarker Research: Identifies H2AFZ overexpression in clinical samples (IHC) .

  • Mechanistic Investigations: Links TP53 mutations to H2AFZ-driven proliferation in HCC cell lines (EdU assays) .

Clinical Relevance

  • Prognostic Value: High H2AFZ levels predict poor survival in HCC (HR = 2.1, P < 0.01) and LUAD (HR = 1.8, P = 0.003) .

  • Therapeutic Target: H2AFZ depletion reduces HCC cell migration by 60% (P < 0.001) , highlighting its potential as a therapeutic target.

Technical Considerations

  • Buffer Composition: 0.01 M PBS (pH 7.4) with 50% glycerol for long-term stability .

  • Safety: Contains sodium azide; handle with protective equipment .

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 your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery timeframes.
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 replaces conventional H2A in a specific subset of nucleosomes. Nucleosomes play a crucial role in packaging and compacting DNA into chromatin, thereby limiting DNA accessibility to cellular machinery that requires DNA as a template. Consequently, histones are central to the regulation of transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is intricately regulated through a complex interplay of post-translational modifications of histones, known as the histone code, and nucleosome remodeling. H2A.Z may be involved in the formation of constitutive heterochromatin and is potentially required for chromosome segregation during cell division.
Gene References Into Functions
  1. H2A.Z associates with epigenetic gene activation in prostate cancer. Acetylated H2A.Z plays a role in activating newly formed enhancers in prostate cancer. PMID: 29116202
  2. Research has shown that 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 the H2A.Z/S-phase kinase-associated protein 2/p27/p21 signaling pathway. PMID: 29532867
  3. A recent study identified 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 are associated with combinations of H2A.Z and p300/CBP at nucleosome-occupied enhancers. PMID: 28301306
  5. Results indicate that the accumulation of H2A.Z within repressed genes can also be a consequence of repressed gene transcription, rather than an active mechanism required for establishing the repression. PMID: 29036442
  6. Findings suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis, highlighting its role in accelerating cell cycle transition and epithelial-mesenchymal transition (EMT) during hepatocarcinogenesis. PMID: 26863632
  7. Crystal structure results demonstrate that the flexible nature of the H2A.Z L1 loop plays a crucial role in 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 surprising 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 driving the proliferation of breast cancer cells. PMID: 27569210
  11. Results suggest that the N-terminal tail of H2A.Z contributes distinctively 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 reveals that YL1 binding, similar to ANP32E binding, triggers an extension of the H2A.Z alphaC helix. PMID: 26974126
  14. H2A.Z removal from chromatin is the primary function of INO80 and ANP32E in promoting homologous recombination. PMID: 26142279
  15. Results demonstrate a male-selective association of the H2AFZ gene with schizophrenia, suggesting that modifications to the H2AFZ signaling pathway warrant further investigation in the context of schizophrenia pathophysiology. PMID: 25392085
  16. Dynamic modulation of H2A.Z exchange and removal by Anp32e underscores the importance of the nucleosome surface and nucleosome dynamics in processing the damaged chromatin template during DNA double-strand break repair. PMID: 26034280
  17. The findings suggest that H2A.Z.2 acts as a mediator of cell proliferation and drug sensitivity in malignant melanoma. PMID: 26051178
  18. The predictive values associated with low expressions of H2AFZ and CASP8AP2 and high white blood cell count suggest that these features could assist in identifying patients at a greater risk of relapse more accurately. PMID: 24397596
  19. Anp32e may help to 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. A study mapped H2A.Z genome-wide in embryonic stem cells and neural progenitors. H2A.Z is deposited at promoters and enhancers, and exhibits a strong correlation 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 alter DNA double-strand breaks repair parameters while affecting clonogenic ability and cell cycle distribution. PMID: 24240188
  22. A mutational analysis revealed 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 interconnected. Epigenetic mechanisms, primarily histone post-translational modifications, are likely involved and impair sirt1-mediated downregulation of H2A.Z via 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. Data demonstrate that 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. Data indicate that histone H2A.Z is a protein capable of specifically binding ST1926. PMID: 23245330
  28. Age-dependent p400 downregulation and loss of H2A.Z localization may contribute to the onset of replicative senescence through a sustained high rate 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 implicated in the transcriptional regulation of genes associated with oncogenesis through 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 allowing 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. This nucleosome shifts during mitosis, resulting in occupancy at the transcriptional start site and a reduced nucleosome-depleted region. PMID: 20864037
  36. This review provides a concise overview of H2A.Z biology and presents hypotheses that could reconcile contradictory reports in the literature 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 initially 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. Results demonstrate that 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 are likely to participate in 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 remaining 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. Studies demonstrate that upon gene induction, human H2A.Z associates with gene promoters and aids 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 referred to as H2A.Z.1) is one of the three isoforms of the H2A.Z histone variant in humans, alongside H2A.Z.2.1 and H2A.Z.2.2. Unlike canonical histones, H2A.Z plays critical roles in both gene activation and repression through its strategic positioning at nucleosomes surrounding the nucleosome-depleted regions of active promoters . H2AFZ is evolutionarily conserved from yeast to humans, underscoring its fundamental importance in chromatin architecture and gene regulation. The significance of H2AFZ in epigenetic research stems from its dynamic incorporation into chromatin by specialized ATP-dependent chromatin remodeling complexes like SRCAP and p400, which can dramatically alter the transcriptional potential of associated genes . Beyond transcriptional control, H2AFZ participates in DNA damage repair mechanisms and genetic stability maintenance, making it a multifunctional component of the epigenome worthy of detailed investigation .

How do I optimize immunofluorescence protocols using H2AFZ (Ab-11) Antibody?

For optimal immunofluorescence results with H2AFZ (Ab-11) Antibody:

  • Fixation: Use freshly prepared 4% paraformaldehyde for 15 minutes at room temperature. Over-fixation can mask epitopes and reduce signal intensity.

  • Permeabilization: Implement a gentle permeabilization with 0.2% Triton X-100 for 10 minutes to maintain nuclear structure while allowing antibody access.

  • Blocking: Use 5% BSA or normal serum from the species of your secondary antibody for 1 hour to minimize background.

  • Primary antibody dilution: Test multiple dilutions (typically 1:100-1:500) based on H2AFZ expression levels in your experimental system. The optimal dilution may differ between cell types due to varying H2AFZ expression levels across tissues .

  • Incubation conditions: For nuclear proteins like H2AFZ, overnight incubation at 4°C generally yields better specificity and signal-to-noise ratio than shorter incubations.

  • Controls: Include both negative controls (secondary antibody only) and positive controls (cell types known to express H2AFZ, such as PDAC cell lines which show high H2AFZ expression) .

  • Counterstaining: When co-staining with other histone marks, consider sequential rather than simultaneous staining to prevent epitope competition.

  • Mounting media: Use anti-fade mounting media with DAPI to visualize nuclear localization of H2AFZ while preserving fluorescent signal.

How can I validate the specificity of H2AFZ (Ab-11) Antibody in my experimental system?

Validating H2AFZ (Ab-11) Antibody specificity requires multiple complementary approaches:

  • Western blot analysis: Confirm a single band at approximately 15 kDa with minimal non-specific bands. Compare this with lysates from cells where H2AFZ has been knocked down using siRNA or CRISPR-Cas9, which should show reduced signal intensity .

  • Peptide competition assay: Pre-incubate the antibody with excess H2AFZ peptide (the immunogen) before immunostaining or Western blotting. Signal elimination confirms specificity.

  • Genetic validation: Generate H2AFZ knockout or knockdown cells using CRISPR-Cas9 techniques as described in literature . Significant signal reduction in these cells confirms antibody specificity.

  • Cross-reactivity assessment: Test the antibody against recombinant H2A.Z.2 isoforms, as H2AFZ differs from H2A.Z.2 by only three amino acids . This is critical for experiments requiring isoform-specific detection.

  • Chromatin immunoprecipitation validation: Perform ChIP-qPCR on known H2AFZ-enriched genomic regions such as active promoters. Enrichment at these sites compared to control regions confirms functional specificity.

  • Mass spectrometry validation: For absolute confidence, perform immunoprecipitation followed by mass spectrometry to identify the exact protein(s) recognized by the antibody .

How can I distinguish between H2AFZ (H2A.Z.1) and other H2A.Z isoforms using antibodies?

Distinguishing between H2A.Z isoforms presents significant challenges due to their high sequence similarity—H2A.Z.1 (encoded by H2AFZ) differs from H2A.Z.2 by only three amino acids . For precise isoform discrimination:

  • Epitope selection: Confirm that the H2AFZ (Ab-11) Antibody targets regions containing isoform-specific amino acids (particularly the T14A difference between H2A.Z.1 and H2A.Z.2) .

  • Validation using isoform-specific knockdowns: Generate cell lines with CRISPR-mediated knockdown of specific H2A.Z isoforms as detailed in the literature, where researchers have successfully targeted H2AFZ and H2AFV (encoding H2A.Z.2) individually . Test antibody reactivity in these lines to confirm isoform specificity.

  • Recombinant protein controls: Use purified recombinant H2A.Z.1, H2A.Z.2.1, and H2A.Z.2.2 proteins as standards in Western blots to assess cross-reactivity.

  • Complementary techniques: Supplement antibody-based detection with mRNA analysis (RT-qPCR) using isoform-specific primers to correlate protein with transcript levels.

  • Mass spectrometry analysis: For definitive identification, perform immunoprecipitation followed by mass spectrometry, which can detect the subtle amino acid differences between isoforms .

  • Functional validation: Since H2A.Z isoforms can have distinct interactors (e.g., PHF14 preferentially interacts with H2A.Z.1 while SIRT1 associates more with H2A.Z.2) , co-immunoprecipitation with these known interactors can provide functional confirmation of isoform identity.

What are the key considerations when performing ChIP-seq with H2AFZ (Ab-11) Antibody?

ChIP-seq with H2AFZ (Ab-11) Antibody requires careful optimization:

  • Crosslinking conditions: Standard 1% formaldehyde for 10 minutes works for most H2AFZ applications, but dual crosslinking with additional DSG (disuccinimidyl glutarate) may improve capture of H2AFZ and associated protein complexes.

  • Chromatin fragmentation: Aim for 200-300bp fragments for optimal resolution. H2AFZ is typically enriched at the -1 and +1 nucleosomes surrounding transcription start sites, requiring precise fragment sizes to accurately map these positions .

  • Antibody amount optimization: Titrate antibody amounts (2-5μg per IP is typical) and validate with ChIP-qPCR at known H2AFZ-enriched regions before proceeding to sequencing.

  • Controls: Include IgG controls and, ideally, H2AFZ-depleted samples as negative controls. Input normalization is essential for accurate peak calling.

  • Library preparation considerations: H2AFZ is enriched at specific genomic features rather than broadly distributed, so deeper sequencing (30-50 million reads) may be necessary for comprehensive coverage.

  • Data analysis specifics: During analysis, focus on +1 and -1 nucleosome positions around transcription start sites where H2AFZ is typically enriched . Consider using nucleosome-positioning algorithms that account for the 147bp DNA footprint of nucleosomes.

  • Validation strategy: Confirm key ChIP-seq findings with ChIP-qPCR and correlate with gene expression data, as H2AFZ can both activate and repress transcription in a context-dependent manner .

  • Isoform considerations: Be aware that standard ChIP-seq with H2AFZ antibodies may capture both H2A.Z.1 and potentially H2A.Z.2 due to their high sequence similarity. Consider complementary approaches to distinguish isoform-specific genomic locations .

How does post-translational modification of H2AFZ affect antibody recognition and experimental outcomes?

Post-translational modifications (PTMs) of H2AFZ can significantly impact antibody recognition:

  • Critical modification sites: The N-terminal tail of H2AFZ contains a basic patch subject to multiple modifications, including acetylation of lysines (K4, K7, K11, K13, K15), ubiquitylation, and potential phosphorylation at T14 (a residue unique to H2A.Z.1) . These modifications can mask or alter epitopes.

  • Antibody epitope mapping: Determine whether the H2AFZ (Ab-11) Antibody epitope includes regions susceptible to PTMs. Antibodies targeting the N-terminal tail (residues 1-19) may show variable reactivity depending on the modification state.

  • Modification-specific outcomes: Acetylation of H2AFZ has been linked to transcriptional activation , so antibodies with differential sensitivity to acetylated vs. non-acetylated H2AFZ will produce disparate ChIP-seq profiles correlating with gene activity states.

  • Experimental strategies:

    • For total H2AFZ detection: Use antibodies targeting epitopes outside PTM-rich regions

    • For modification-specific analysis: Complement H2AFZ (Ab-11) with modification-specific antibodies (e.g., acetyl-H2AFZ)

    • For comprehensive analysis: Consider sequential ChIP (re-ChIP) to identify differentially modified H2AFZ subpopulations

  • Validation approaches: Test antibody reactivity against synthetic peptides with defined modifications and in cells treated with HDAC inhibitors (which increase H2AFZ acetylation) or phosphatase inhibitors to confirm modification sensitivity.

  • Functional correlations: The PHF14 complex, which preferentially interacts with H2AFZ, contains multiple PHD fingers that may function as readers of H2AFZ PTMs , providing a functional readout for modification states.

What role does H2AFZ play in DNA damage repair, and how can this be investigated using H2AFZ (Ab-11) Antibody?

H2AFZ plays significant roles in DNA damage repair that can be investigated using specialized approaches:

  • Mechanistic basis: H2AFZ facilitates DNA damage repair primarily through the exchange of H2A/H2A.Z at DNA damage sites . The FBXL10-RNF68-RNF2 ubiquitin ligase complex mediates H2A monoubiquitylation, facilitating this exchange process .

  • Experimental design for studying H2AFZ in DNA damage:

    • Induce DNA damage using UV, ionizing radiation, or chemical agents (e.g., etoposide)

    • Track H2AFZ recruitment to damage sites using immunofluorescence with H2AFZ (Ab-11) Antibody

    • Perform time-course ChIP experiments to measure dynamic H2AFZ incorporation at damage sites

    • Use laser microirradiation combined with live-cell imaging of tagged H2AFZ to monitor real-time recruitment

  • Key controls and validations:

    • Confirm damage induction using γH2AX staining

    • Include H2AFZ-depleted cells as negative controls

    • Monitor recruitment of known DNA damage factors (53BP1, BRCA1) in parallel

  • Advanced techniques:

    • Proximity ligation assays (PLA) to detect interactions between H2AFZ and DNA repair proteins

    • CRISPR-Cas9 editing of H2AFZ to introduce mutations in potential phosphorylation sites (e.g., T14) that might regulate its function in DNA repair

    • Sequential ChIP to determine if DNA damage-associated H2AFZ carries specific modifications

  • Functional readouts:

    • Measure DNA repair efficiency following H2AFZ depletion

    • Investigate cell cycle checkpoint activation in relation to H2AFZ levels

    • Assess chemosensitivity changes when H2AFZ is depleted, as observed in pancreatic cancer cells that show increased sensitivity to gemcitabine upon H2AFZ knockdown

How is H2AFZ expression altered in cancer, and what are the implications for using H2AFZ antibodies in cancer research?

H2AFZ expression shows significant alterations in cancer with important research implications:

  • Expression patterns: H2AFZ is frequently overexpressed in multiple cancer types, notably in pancreatic ductal adenocarcinoma (PDAC) where high expression correlates with poor prognosis . This overexpression extends to all three H2A.Z isoforms (H2A.Z.1, H2A.Z.2.1, and H2A.Z.2.2) .

  • Functional consequences: H2AFZ overexpression contributes to cancer progression through:

    • Suppression of senescence pathways

    • Promotion of cell cycle progression

    • Downregulation of cyclin-dependent kinase inhibitors like CDKN2A/p16

    • Altered gene expression in fatty acid biosynthesis pathways

    • Enhanced DNA damage repair leading to chemoresistance

  • Research applications:

    • Diagnostic potential: H2AFZ (Ab-11) Antibody can be used in immunohistochemistry to assess H2AFZ levels as a potential diagnostic biomarker, particularly in PDAC where expression correlates with outcomes

    • Therapeutic target validation: Monitor changes in H2AFZ levels in response to experimental therapies

    • Mechanistic studies: Investigate how H2AFZ overexpression contributes to senescence evasion and chemoresistance

  • Technical considerations:

    • Higher background may occur in cancer tissues due to elevated H2AFZ levels; optimization of antibody dilutions is critical

    • Include normal adjacent tissue controls to establish baseline expression

    • Consider dual staining with proliferation markers to correlate H2AFZ levels with proliferative capacity

    • For immunoprecipitation studies, account for potentially higher H2AFZ protein abundance in cancer samples when optimizing protocols

  • Validation approaches:

    • Correlate protein levels detected by antibody with mRNA expression

    • Compare antibody staining patterns with public database expression data

    • Validate findings using H2AFZ knockdown models in cancer cell lines

How can I investigate the interplay between H2AFZ isoforms in disease models using antibodies?

Investigating H2AFZ isoform interplay requires sophisticated experimental design:

  • Isoform-specific knockdown models: Generate cell lines with selective depletion of H2A.Z isoforms using CRISPR-Cas9 technology targeting H2AFZ (H2A.Z.1) or H2AFV (H2A.Z.2) as demonstrated in research studies . These models can reveal compensatory mechanisms and unique functions.

  • Rescue experiments: After knocking down all H2A.Z isoforms, perform selective rescue experiments by reintroducing individual isoforms (H2A.Z.1, H2A.Z.2.1, or H2A.Z.2.2). Research has shown that H2A.Z.1 and H2A.Z.2.1 can more effectively restore oncogenic phenotypes compared to H2A.Z.2.2 in PDAC models .

  • Antibody application strategies:

    • Use H2AFZ (Ab-11) Antibody in combination with isoform-specific antibodies when available

    • Implement epitope-tagged isoforms to circumvent antibody specificity limitations

    • Perform sequential immunoprecipitation to isolate specific isoform complexes

  • Analysis of isoform-specific interactors:

    • Co-immunoprecipitation combined with mass spectrometry to identify differential protein interactions

    • Focus on known isoform-specific partners: PHF14 preferentially interacts with H2A.Z.1, while SIRT1 associates more with H2A.Z.2

    • Validate interactions through reciprocal co-immunoprecipitation and proximity ligation assays

  • Functional readouts in disease models:

    • Measure isoform-specific effects on gene expression using RNA-Seq after selective knockdown

    • Assess phenotypic outcomes like senescence (SA-β-galactosidase activity), cell cycle progression, and chemosensitivity

    • Monitor changes in inflammatory mediators like interleukin-8, which increases upon H2A.Z depletion in PDAC models

  • Analysis of genomic distribution:

    • Compare ChIP-seq profiles of differentially tagged isoforms to identify unique and shared genomic targets

    • Focus on genes showing antagonistic regulation by different H2A.Z isoforms

What are the critical optimization steps for Western blotting with H2AFZ (Ab-11) Antibody?

Optimizing Western blot protocols for H2AFZ detection requires attention to several critical factors:

  • Sample preparation:

    • Use histone extraction protocols with sulfuric acid or Triton extraction to enrich for nuclear proteins

    • Include protease inhibitors and phosphatase inhibitors to preserve H2AFZ and its modifications

    • For total protein extracts, ensure complete nuclear lysis to release chromatin-bound H2AFZ

  • Gel electrophoresis parameters:

    • Use high percentage (15-18%) SDS-PAGE gels or specialized Triton-Acid-Urea (TAU) gels for better separation of histone variants

    • Load appropriate positive controls (e.g., recombinant H2AFZ, nuclear extracts from cells known to express high H2AFZ levels like PDAC cell lines)

    • Consider including isoform controls if distinguishing between H2A.Z variants

  • Transfer conditions:

    • Optimize transfer time and voltage for small proteins (~15 kDa)

    • Use PVDF membranes with small pore size (0.22μm) to prevent small protein loss

    • Consider semi-dry transfer systems which can be more efficient for histone proteins

  • Blocking and antibody incubation:

    • Test different blocking agents (BSA often works better than milk for histone detection)

    • Determine optimal H2AFZ (Ab-11) Antibody dilution through titration (typically 1:500-1:2000)

    • Extend primary antibody incubation time (overnight at 4°C) for improved specificity

  • Detection systems:

    • Use high-sensitivity ECL systems due to the relatively low abundance of specific histone variants

    • Consider fluorescent secondary antibodies for more quantitative analysis and multiplexing with other histone markers

  • Validation approaches:

    • Include H2AFZ-depleted samples as negative controls

    • Perform peptide competition assays to confirm signal specificity

    • Run H2A.Z.1 and H2A.Z.2 recombinant proteins in parallel to assess cross-reactivity

  • Quantification considerations:

    • Normalize to total histone H3 or H4 rather than housekeeping proteins like GAPDH or β-actin

    • Account for potential post-translational modifications that might affect antibody recognition or protein mobility

How do I design experiments to study the antagonistic functions of H2AFZ and other H2A.Z isoforms in gene regulation?

Designing experiments to investigate H2AFZ's antagonistic functions requires sophisticated approaches:

  • Experimental model development:

    • Generate cell lines with individual or combined depletion of H2A.Z isoforms using CRISPR-Cas9

    • Create cells expressing epitope-tagged isoforms at endogenous levels using homology-directed repair as described in the literature

    • Develop inducible expression systems to control the relative levels of different isoforms

  • Gene target selection:

    • Focus initial studies on genes known to be antagonistically regulated by H2A.Z isoforms, such as PLAT and AKAP12

    • Perform RNA-Seq after isoform-specific depletions to identify additional targets showing differential regulation

    • Select genes from diverse functional categories to capture the full spectrum of H2A.Z regulatory roles

  • Mechanistic investigation strategy:

    • Perform ChIP-seq with isoform-specific antibodies or epitope tags to map genomic distribution

    • Analyze nucleosome stability using MNase-seq in cells with altered isoform ratios

    • Assess recruitment of transcriptional regulators using sequential ChIP or CUT&RUN approaches

  • Interactome analysis:

    • Isolate protein complexes associated with specific H2A.Z isoforms using immunoprecipitation followed by mass spectrometry

    • Focus on known differential interactors like PHF14 (preferentially associates with H2A.Z.1) and SIRT1 (preferentially associates with H2A.Z.2)

    • Use proximity-based labeling techniques (BioID, APEX) to capture transient interactions

  • Functional validation experiments:

    • Perform co-depletion experiments involving H2A.Z isoforms and their specific interactors (e.g., SIRT1, PHF14) to test dependency relationships

    • Analyze changes in chromatin accessibility using ATAC-seq when isoform ratios are altered

    • Investigate post-translational modification patterns specific to each isoform and their functional impacts

  • Quantitative analytical approaches:

    • Use RT-qPCR to precisely measure expression changes in target genes

    • Implement ChIP-qPCR to quantify isoform occupancy at specific regulatory elements

    • Apply mathematical modeling to understand how changes in isoform ratios affect gene expression outcomes

What controls and validation steps are essential when using H2AFZ (Ab-11) Antibody in proximity ligation assays (PLA)?

For rigorous proximity ligation assays with H2AFZ (Ab-11) Antibody:

  • Essential antibody controls:

    • Single primary antibody controls to establish background signal levels

    • Isotype-matched irrelevant antibody controls to assess non-specific interactions

    • Peptide competition controls to confirm H2AFZ specificity

    • H2AFZ-depleted cell controls using CRISPR or siRNA knockdown systems

  • Biological validation controls:

    • Positive interaction controls: pair H2AFZ antibody with antibodies against known interactors (e.g., SRCAP complex components, p400 complex members)

    • Negative interaction controls: pair H2AFZ antibody with antibodies against proteins not expected to interact

    • Stimulus-dependent controls: examine interactions known to increase or decrease under specific conditions (e.g., DNA damage)

  • Technical optimization parameters:

    • Fixation method: Optimize between crosslinking fixatives (formaldehyde) and organic solvent fixation (methanol) based on epitope accessibility

    • Antibody concentration: Titrate both H2AFZ antibody and partner antibody independently to minimize background

    • Proximity probe selection: Test different proximity probes (PLUS/MINUS) for each antibody combination to optimize signal-to-noise ratio

    • Amplification duration: Adjust rolling circle amplification time to balance sensitivity and specificity

  • Validation through orthogonal methods:

    • Confirm key interactions using co-immunoprecipitation followed by Western blotting

    • Correlate PLA signals with co-localization observed in super-resolution microscopy

    • Compare interaction profiles with published mass spectrometry datasets

  • Contextual validation approaches:

    • Test interactions in different cell types with varying H2AFZ expression levels

    • Examine interaction differences between normal and disease states (e.g., cancer cells with H2AFZ overexpression)

    • Assess interactions at different cell cycle stages or following specific treatments

  • Quantification and statistical analysis:

    • Score PLA signals in multiple cells (>100) across independent experiments

    • Implement automated spot counting with defined intensity thresholds

    • Apply appropriate statistical tests with corrections for multiple comparisons when screening numerous interactions

How can I accurately quantify H2AFZ occupancy at specific genomic loci in different experimental conditions?

Accurately quantifying H2AFZ occupancy requires rigorous methodology:

  • Chromatin immunoprecipitation approaches:

    • Standard ChIP-qPCR using H2AFZ (Ab-11) Antibody for targeted loci analysis

    • ChIP-seq for genome-wide occupancy patterns

    • CUT&RUN or CUT&Tag for improved signal-to-noise and reduced sample input requirements

    • Sequential ChIP (re-ChIP) to identify H2AFZ populations with specific modification patterns

  • Control strategies:

    • Input normalization to account for chromatin preparation biases

    • IgG negative controls to establish background signals

    • Spike-in normalization using foreign chromatin (e.g., Drosophila) for quantitative comparisons between conditions

    • Positive control regions with established H2AFZ enrichment (e.g., active promoters)

  • Technical optimization:

    • Crosslinking conditions: Standard 1% formaldehyde for 10 minutes is typical, but optimization may be required

    • Sonication parameters: Achieve consistent fragment size distribution (200-300bp) across samples

    • Antibody amounts: Titrate antibody to chromatin ratio to ensure complete immunoprecipitation

    • Washing stringency: Balance between reducing background and maintaining specific interactions

  • Quantification approaches:

    • Percent input method for ChIP-qPCR to calculate enrichment

    • Fold enrichment over IgG for comparing occupancy between conditions

    • For ChIP-seq: use spike-in normalization factors or house-keeping regions for scaling between samples

    • Implement appropriate statistical tests for comparing occupancy changes

  • Genomic context considerations:

    • Analyze H2AFZ occupancy at specific functional elements (promoters, enhancers, gene bodies)

    • Examine nucleosome positioning in relation to transcription start sites (-1 and +1 nucleosomes)

    • Compare occupancy with gene expression data, as H2AFZ can both activate and repress transcription

    • Correlate with other histone modifications to understand chromatin environment

  • Validation strategies:

    • Confirm key findings using orthogonal techniques like CUT&RUN

    • Perform targeted validation in H2AFZ-depleted cells

    • Correlate occupancy changes with functional outcomes in gene expression

How should I interpret conflicting results between H2AFZ antibody-based detection methods and mRNA expression data?

When facing discrepancies between H2AFZ protein and mRNA data:

  • Technical considerations:

    • Antibody specificity: Verify whether the H2AFZ (Ab-11) Antibody cross-reacts with H2A.Z.2 isoforms, which differ by only three amino acids .

    • Epitope accessibility: Post-translational modifications can mask epitopes, particularly in the N-terminal region where acetylation, methylation, or phosphorylation may occur .

    • mRNA splicing: Confirm that mRNA detection methods capture all relevant transcript variants.

  • Biological explanations:

    • Post-transcriptional regulation: H2AFZ protein levels may be regulated by microRNAs or RNA-binding proteins independent of mRNA abundance.

    • Protein stability differences: H2AFZ incorporation into chromatin may stabilize the protein compared to free histone pools.

    • Isoform compensation: Knockdown of one H2A.Z isoform can lead to compensatory increases in other isoforms .

  • Temporal dynamics:

    • mRNA and protein expression may be temporally offset due to translation rates and protein turnover.

    • Consider time-course experiments to capture the relationship between transcription and protein accumulation.

  • Cellular compartmentalization:

    • Total cellular mRNA measurements may not reflect nuclear histone pools.

    • Use fractionation approaches to compare cytoplasmic mRNA with nuclear protein levels.

  • Resolution strategies:

    • Implement epitope-tagged H2AFZ expression to circumvent antibody limitations .

    • Use isoform-specific RT-qPCR to distinguish between H2A.Z variants at the mRNA level.

    • Perform protein half-life measurements to assess differential stability.

    • Consider ribosome profiling to measure translation efficiency as an intermediate between mRNA and protein.

  • Contextual interpretation:

    • In cancer models, evaluate whether regulatory networks are altered in ways that might uncouple mRNA and protein levels .

    • Determine if detected discrepancies correlate with specific cellular states or treatments.

What computational approaches are recommended for analyzing H2AFZ ChIP-seq data in relation to gene expression changes?

Sophisticated computational approaches for H2AFZ ChIP-seq analysis include:

  • Data processing pipeline:

    • Quality control: Implement FastQC for read quality assessment and adapter trimming

    • Alignment: Use Bowtie2 or BWA with parameters optimized for histone ChIP-seq

    • Peak calling: Apply MACS2 with histone-specific settings (broad peaks) or nucleosome-aware algorithms

    • Visualization: Generate normalized bigWig files for genome browser visualization

  • H2AFZ-specific analytical approaches:

    • Metagene analysis: Assess average H2AFZ distribution around transcription start sites, stratified by gene expression levels

    • Nucleosome positioning: Implement NucleoATAC or DANPOS for precise mapping of H2AFZ-containing nucleosomes

    • Differential binding analysis: Use DiffBind or MAnorm to compare H2AFZ occupancy between conditions

  • Integration with gene expression data:

    • Correlation analysis: Calculate Spearman correlations between H2AFZ occupancy at promoters and mRNA levels

    • Gene set enrichment: Apply GSEA to determine if H2AFZ-bound genes show coordinated expression changes

    • Machine learning approaches: Implement random forest or support vector machine models to predict gene expression from H2AFZ binding patterns

  • Multi-omic integration strategies:

    • Integrate H2AFZ ChIP-seq with other histone modifications using MultiGPS or ChromHMM

    • Combine with chromatin accessibility data (ATAC-seq) to assess nucleosome dynamics

    • Incorporate transcription factor ChIP-seq to identify co-regulatory relationships

  • Functional interpretation:

    • Motif analysis: Identify DNA sequence motifs enriched in H2AFZ-bound regions using MEME or HOMER

    • Pathway analysis: Determine if H2AFZ-regulated genes cluster in specific biological pathways

    • Network analysis: Construct gene regulatory networks focused on H2AFZ-bound genes

  • H2AFZ-specific analytical considerations:

    • Distinguish between activating and repressive roles based on genomic context and co-occurring features

    • Account for the dual functionality of H2AFZ in gene regulation when interpreting results

    • Analyze antagonistic effects of H2AFZ isoforms by comparing datasets from isoform-specific experiments

How can I determine if the H2AFZ patterns detected in my experimental system are functionally significant?

Establishing functional significance of H2AFZ patterns requires multiple lines of evidence:

  • Perturbation experiments:

    • Perform targeted knockdown of H2AFZ using siRNA or CRISPR-Cas9

    • Implement acute depletion systems (e.g., auxin-inducible degron) to study immediate consequences

    • Conduct rescue experiments with wild-type or mutant H2AFZ to establish causality

    • Target H2AFZ deposition machinery (SRCAP, p400) or removal factors (ANP32E)

  • Functional correlation analyses:

    • Compare H2AFZ occupancy patterns with transcriptional outcomes using RNA-seq

    • Correlate with chromatin accessibility (ATAC-seq) to assess impact on chromatin structure

    • Examine relationships with DNA damage response markers in repair-focused studies

    • Assess cellular phenotypes like senescence, cell cycle progression, or chemosensitivity in cancer models

  • Structural and mechanistic validation:

    • Investigate nucleosome stability changes using MNase-seq or salt fractionation

    • Perform mass spectrometry to identify co-occurring modifications and binding partners

    • Use in vitro reconstitution of H2AFZ-containing nucleosomes to test biochemical properties

  • Comparative genomics approaches:

    • Compare H2AFZ patterns across cell types or disease states

    • Evaluate evolutionary conservation of H2AFZ-occupied regions

    • Assess consistency with published H2AFZ datasets and known functions

  • Molecular intervention strategies:

    • Design experiments targeting specific post-translational modifications of H2AFZ

    • Disrupt interactions with specific partners (e.g., PHF14 for H2A.Z.1 or SIRT1 for H2A.Z.2)

    • Implement domain swaps between H2A.Z isoforms to map functional regions

  • Disease relevance assessment:

    • In cancer models, correlate H2AFZ patterns with known oncogenic pathways

    • Test whether modulating H2AFZ affects sensitivity to therapeutic agents, as observed for gemcitabine in PDAC

    • Evaluate whether H2AFZ biomarker potential correlates with clinical outcomes

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.