Mono-methyl-H2AFZ (K7) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Mono-methyl-H2AFZ (K7) Antibody

The Mono-methyl-H2AFZ (K7) antibody is a specialized immunological tool designed to detect the monomethylation of lysine 7 (K7) on histone H2AFZ, a variant of the canonical H2A histone. H2AFZ (also known as H2AZ) is evolutionarily conserved and plays critical roles in chromatin dynamics, including nucleosome stability, gene regulation, and cellular differentiation . This antibody specifically recognizes the post-translational modification of H2AFZ at K7, enabling researchers to study its functional implications in epigenetic regulation.

Mechanism of Action and Biological Significance

The Mono-methyl-H2AFZ (K7) antibody binds to H2AFZ that has undergone monomethylation at lysine 7, a modification catalyzed by the lysine methyltransferase SETD6 . This modification is dynamically regulated during cellular processes such as stem cell differentiation, where it interacts with repressive chromatin marks like H3K27me3 to silence differentiation genes .

Key biological insights:

  • SETD6-mediated methylation: In vitro and in vivo studies confirm that SETD6 exclusively monomethylates H2AFZK7, with no evidence of di- or trimethylation .

  • Role in gene silencing: H2AFZK7me1 colocalizes with H3K27me3 at promoters of differentiation markers (e.g., Fgf5, FoxA2) in embryonic stem cells (ESCs), suggesting a cooperative role in maintaining pluripotency .

  • Dynamic regulation: During retinoic acid (RA)-induced differentiation, H2AFZK7me1 levels decrease, coinciding with loss of H3K27me3 and activation of differentiation genes .

Research Findings and Applications

ApplicationMethodKey FindingsCitations
ChIP-SeqChromatin immunoprecipitation sequencingH2AFZK7me1 localizes to promoters of differentiation genes in ESCs; depletion of SETD6 disrupts self-renewal, leading to premature differentiation .
Western BlotImmunoblottingIncreased H2AFZK7me1 levels detected upon RA-induced differentiation; SETD6 knockdown reduces methylation .
Mass SpectrometryIn vitro KMT assaysConfirmed SETD6’s specificity for H2AFZK7 monomethylation; secondary methylation at K4 observed .
Cellular DifferentiationshRNA-mediated depletionSETD6 depletion in mESCs causes loss of H2AFZK7me1, H3K27me3, and self-renewal capacity .

Comparative Analysis with Other Antibodies

While the Mono-methyl-H2AFZ (K7) antibody is specialized for K7 methylation, other antibodies target distinct modifications of H2AFZ:

Antibody TargetModificationApplicationsKey Difference
H2AFZ (General)Unmethylated H2AFZChIP, WB, IFDetects total H2AFZ, not specific to K7 methylation .
Acetyl-H2AFZK7Acetylation at K7WBRecognizes acetylation, not methylation .
H2AFZK7me1Monomethylation at K7ChIP, ChIP-Seq, WBExclusive to K7 methylation; validated for differentiation studies .

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the method of purchase or location. For specific delivery times, please consult your local distributor.
Synonyms
Histone H2A.Z (H2A/z), H2AFZ, H2AZ
Target Names
Uniprot No.

Target Background

Function
Histone H2A.Z is a variant histone that replaces conventional H2A in a specific subset of nucleosomes. Nucleosomes are responsible for the wrapping and compacting of DNA into chromatin, thereby limiting the accessibility of DNA to cellular machinery that require DNA as a template. Histones, therefore, play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is regulated through a complex interplay of post-translational modifications of histones, known as the histone code, and nucleosome remodeling. Histone H2A.Z may be involved in the formation of constitutive heterochromatin and might 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. Research indicates that H2A.Z is overexpressed in intrahepatic cholangiocarcinoma (ICC) and its expression correlates with poor prognosis in patients with ICC. 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 study identifies GAS41 as a histone acetylation reader that promotes histone H2A.Z deposition in non-small cell lung cancer. PMID: 29437725
  4. The study identifies two possible modes of pioneering associated with combinations of H2A.Z and p300/CBP at nucleosome-occupied enhancers. PMID: 28301306
  5. Findings suggest that the 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. Results suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis and that it plays a well-established 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 inhibits 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. Findings indicate that the N-terminal tail of H2A.Z makes distinct 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 reveals that YL1 binding, similar to ANP32E binding, triggers an extension of the H2A.Z alphaC helix. PMID: 26974126
  14. The removal of H2A.Z from chromatin is the primary function of INO80 and ANP32E in promoting homologous recombination. PMID: 26142279
  15. Results demonstrated a male-selective association of the H2AFZ gene with schizophrenia, suggesting that modifications to 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 DNA double-strand break repair. PMID: 26034280
  17. Findings implicate H2A.Z.2 as 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 to more accurately identify patients at greater risk of relapse. PMID: 24397596
  19. Anp32e may help to resolve non-nucleosomal H2A.Z aggregates and also facilitate the removal of H2A.Z at the +1 nucleosomes. The latter may aid 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 strongly correlates 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 that are 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 parameters of DNA double-strand breaks repair, but it does affect clonogenic ability and cell cycle distribution. PMID: 24240188
  22. 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 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. 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 binding ST1926 specifically. 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 transcriptional regulation of genes associated with oncogenesis through its interaction with H2A.Z. PMID: 22678762
  31. The 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, which 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 found 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, and consequently modulates 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. 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 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. Studies show that 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 influence 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 Mono-methyl-H2AFZ (K7) and why is it significant in epigenetic research?

Mono-methyl-H2AFZ (K7), also known as H2AZK7me1, is a specific post-translational modification where the histone variant H2AZ is monomethylated at lysine 7 in its N-terminal tail. This modification is catalyzed primarily by the lysine methyltransferase SETD6, which has been shown to effectively monomethylate the K7 position of H2AZ in vitro and contribute to H2AZ methylation in vivo . The significance of this modification lies in its role in chromatin regulation, particularly in embryonic stem cells where it appears to function as a marker of cellular differentiation. The H2AZK7me1 mark has been found to co-localize with the repressive H3K27me3 mark and the Polycomb Repressive Complex 2 (PRC2) complex subunits near the transcription start sites of developmental genes in mouse embryonic stem cells (mESCs) . These findings suggest that H2AZK7me1 plays a crucial role in maintaining the self-renewal capabilities of stem cells and regulates gene expression during cellular differentiation.

How does H2AZK7me1 differ from other H2AZ post-translational modifications?

H2AZ is subject to various post-translational modifications that regulate its function in chromatin. While H2AZ is known to be polyacetylated within the N-terminal tail (at residues K4, K7, K11, K13, and K15) as well as monoubiquitylated at the carboxy-terminus (at residues K120 and K121), H2AZ methylation was a novel discovery . The methylation of H2AZ at lysines 4 and 7 likely prevents subsequent acetylation at these sites, thereby potentially impairing the transcriptional functions associated with H2AZ acetylation . Interestingly, mass spectrometry analysis did not detect any acetylation on endogenous H2AZ in the contexts studied, suggesting potential competition between these modifications . Additionally, immunoblotting with a methyl-specific H2AZK7me1 antibody detected a slow-migrating band that, based on its molecular weight, was suggested to be an ubiquitylated form of H2AZ, indicating possible cross-talk between methylation and ubiquitylation of H2AZ .

What methods can be used to detect H2AZK7me1 in biological samples?

Detection of H2AZK7me1 in biological samples can be accomplished through several complementary techniques:

  • Immunoblotting: Using a methyl-specific α-H2AZK7me1 antibody, researchers can detect the presence of this modification in protein lysates. This approach has been successfully employed to confirm the in vitro methylation of H2AZ by SETD6 and to detect changes in H2AZK7me1 levels during cellular differentiation .

  • Mass Spectrometry (MS): This technique provides definitive identification of the specific methylation sites. MS analyses have successfully detected monomethylation of H2AZ at both K4 and K7 in vitro and in vivo. Detailed MS2 chromatograms can highlight the monomethylation of H2AZ at specific lysine residues with precise molecular masses (e.g., 766.49 for K7 methylation) .

  • Chromatin Immunoprecipitation (ChIP): This method allows researchers to examine the genomic distribution of H2AZK7me1, particularly at promoters of interest such as developmental genes. ChIP analyses have revealed the co-localization of H2AZK7me1 with the repressive H3K27me3 mark at the promoters of differentiation marker genes .

  • Immunohistochemistry (IHC): While not specifically mentioned for H2AZK7me1 in the provided search results, IHC is commonly used to detect protein expression levels in tissue samples, as demonstrated for H2AFZ in lung adenocarcinoma tissues .

Which enzymes are responsible for the methylation and potential demethylation of H2AZK7?

The primary enzyme responsible for H2AZK7 monomethylation is SETD6, a lysine methyltransferase that has been shown to effectively methylate H2AZ at lysine 7 in vitro and contribute to H2AZ methylation in vivo . In detailed in vitro experiments, SETD6 was demonstrated to preferentially monomethylate H2AZ at K7, although it can also methylate secondary sites such as K4 at lower levels .

Another enzyme, SET7, has also been identified as a potent lysine methyltransferase that can modify H2AZ, although it appears to indiscriminately methylate multiple sites on H2AZ rather than showing the specificity that SETD6 exhibits for K7 .

What is the relationship between H2AZK7me1 and embryonic stem cell differentiation?

H2AZK7me1 has a complex relationship with embryonic stem cell differentiation that appears to be context-dependent:

  • Global Increase During Differentiation: Mass spectrometric analysis detected a significant (over 3-fold) increase in H2AZK7me1 and H2AZK4me1K7me1 following mouse embryonic stem cell (mESC) differentiation, whether induced by shRNA-mediated depletion of Nanog or by retinoic acid (RA) treatment .

  • Promoter-Specific Dynamics: Intriguingly, while global levels of H2AZK7me1 increased upon differentiation, ChIP analyses revealed decreased levels of both H2AZK7me1 and the repressive H3K27me3 mark at the promoters of differentiation marker genes (Fgf5, FoxA2, and Hand2) upon retinoic acid-induced differentiation .

  • Role in Self-Renewal: Depletion of Setd6 (the enzyme that catalyzes H2AZK7 methylation) in mESCs led to cellular differentiation, compromised self-renewal, and reduced clonogenicity, suggesting that Setd6-mediated H2AZ methylation is required for maintaining mESC self-renewal .

These findings suggest that H2AZK7me1 functions as part of an epigenetic program that regulates lineage commitment genes, potentially cooperating with H3K27me3 to maintain repression of differentiation markers in the self-renewal state .

How can researchers validate the specificity of a Mono-methyl-H2AFZ (K7) Antibody?

Validating the specificity of a Mono-methyl-H2AFZ (K7) Antibody requires a multi-faceted approach:

  • Mutational Analysis: Generate H2AZ mutants where K7 is converted to an arginine (K7R) or other non-methylatable residue. The antibody should show significantly decreased or no signal with these mutants compared to wild-type H2AZ .

  • Peptide Competition Assays: Test the antibody against a panel of synthetic peptides containing modified and unmodified versions of the target site (H2AZK7). The antibody should preferentially recognize the monomethylated K7 peptide over unmodified, di-, or tri-methylated versions .

  • Knockdown/Knockout Validation: Deplete H2AZ using shRNA or CRISPR-Cas9 approaches and confirm reduced antibody signal. In the provided search results, the H2AZK7me1 antibody detected a decrease of H2AZ in knockdown cell extracts (shH2AZ) compared to control cells .

  • Enzyme Depletion: Knockdown the methyltransferase responsible for the modification (SETD6) and demonstrate reduced antibody signal. Cells depleted of SETD6 by two independent shRNAs showed reduced levels of H2AZK7me1, confirming the antibody's specificity for the SETD6-mediated modification .

  • Mass Spectrometry Correlation: Perform mass spectrometry analysis to confirm the presence of monomethylation at K7 in samples that show positive antibody reactivity, as was done in the reported research where MS confirmed the methylation detected by the antibody .

  • In Vitro Methylation Assays: Conduct in vitro methylation reactions with recombinant H2AZ and SETD6, then analyze the products by immunoblotting with the methyl-specific antibody. A substantial increase in signal in the presence of both H2AZ and SETD6 would indicate specificity for the enzymatically generated modification .

What controls should be included when using H2AZK7me1 antibodies in ChIP experiments?

When conducting Chromatin Immunoprecipitation (ChIP) experiments with H2AZK7me1 antibodies, the following controls are essential:

How does H2AZK7me1 co-localize with other histone marks, and what does this tell us about its function?

H2AZK7me1 exhibits specific co-localization patterns with other histone modifications that provide insights into its functional role:

  • Co-localization with H3K27me3: H2AZK7me1 co-localizes with the repressive H3K27me3 mark near the transcription start sites of developmental genes in mouse embryonic stem cells (mESCs) . This co-occurrence suggests a cooperative role in gene silencing.

  • Association with PRC2 Complex: H2AZ, which can be methylated at K7, co-localizes with the PRC2 complex subunits SUZ12 and EED in mESCs near the transcription start sites of developmental genes . This association indicates that H2AZK7me1 may be part of the Polycomb-mediated gene repression machinery.

  • Dynamic Changes During Differentiation: Upon retinoic acid-induced cellular differentiation, both H2AZK7me1 and H3K27me3 are concomitantly lost from the promoters of differentiation markers such as Fgf5, FoxA2, and Hand2 . This synchronized removal suggests a coordinated epigenetic program regulating developmental gene expression.

  • Mutual Exclusivity with Acetylation: Methylation of H2AZ at K7 likely prevents acetylation at the same residue. Mass spectrometry analysis did not detect acetylation on endogenous H2AZ in the contexts studied, suggesting potential competition between these modifications .

These co-localization patterns suggest that H2AZK7me1 functions in an epigenetic program that, together with H3K27me3 and the PRC2 complex, maintains repression of differentiation-associated genes in the self-renewal state. The loss of both marks during differentiation allows for the activation of these genes and subsequent lineage commitment .

What is known about the biological significance of dual methylation at H2AZK4 and H2AZK7?

The dual methylation of H2AZ at both K4 and K7 (H2AZK4me1K7me1) appears to have specific biological significance:

  • Co-occurrence In Vivo: Mass spectrometric analysis detected H2AZK7me1 in vivo exclusively in conjunction with H2AZK4me1, suggesting these modifications may function as a coordinated epigenetic mark .

  • Differentiation-Associated Increase: The levels of H2AZK4me1K7me1 increased noticeably (over 3-fold) in response to cellular differentiation induced by either retinoic acid or the silencing of Nanog expression . This suggests a potential role in the differentiation process.

  • Enzymatic Specificity: While SETD6 preferentially monomethylates H2AZ at K7, it can also methylate K4 at lower levels. Mass spectrometry detected low levels of simultaneous methylation on both K4 and K7 in in vitro reactions . This suggests that a single enzyme might coordinate the dual methylation pattern.

  • Potential Functional Synergy: The consistent co-occurrence of these modifications suggests they may work synergistically rather than independently. Their coordinated increase during differentiation implies a functional role in this process .

  • Distinction from Single Methylation: The dual methylation mark may have distinct functions from single methylation at either site. The research indicates that, while global levels of dual methylation increase during differentiation, locus-specific dynamics (such as at developmental gene promoters) may differ .

While the exact mechanism by which H2AZK4me1K7me1 influences gene expression remains to be fully elucidated, its differentiation-associated increase suggests it may be involved in the regulation of cell lineage commitment and developmental programs .

How do experimental approaches to studying H2AZK7me1 differ between embryonic stem cells and cancer contexts?

Experimental approaches to studying H2AZK7me1 differ significantly between embryonic stem cell and cancer research contexts:

In Embryonic Stem Cells:

  • Differentiation Models: Research utilizes controlled differentiation systems such as retinoic acid treatment or shRNA-mediated depletion of pluripotency factors like Nanog to study dynamic changes in H2AZK7me1 during lineage commitment .

  • Functional Assays: Alkaline phosphatase staining, clonogenic assays, and self-renewal competition assays are employed to assess the functional importance of H2AZK7me1 and its methyltransferase SETD6 in maintaining pluripotency .

  • Developmental Gene Focus: ChIP analyses typically target the promoters of developmental and differentiation marker genes (e.g., Fgf5, FoxA2, and Hand2) to understand how H2AZK7me1 regulates lineage-specific expression .

In Cancer Research:

While some methodologies overlap (such as immunohistochemistry and expression analysis), the research questions and biological systems differ substantially. Stem cell research focuses on developmental transitions and differentiation processes, while cancer research emphasizes prognostic value, tumor biology, and potential therapeutic applications .

What technical challenges exist in distinguishing between H2AZK7me1 and other similar histone modifications in experimental systems?

Distinguishing H2AZK7me1 from other similar histone modifications presents several technical challenges:

  • Antibody Cross-Reactivity: Generating antibodies that specifically recognize H2AZK7me1 without cross-reacting with unmethylated H2AZ, H2AZK4me1, or H2AZ acetylated at K7 requires extensive validation. The sequence surrounding K7 in H2AZ shares similarities with other histones, potentially leading to non-specific binding .

  • Co-occurrence of Modifications: H2AZK7me1 has been found to co-occur with H2AZK4me1 in vivo, making it difficult to attribute biological effects specifically to K7 methylation versus K4 methylation or the dual modification . Mass spectrometry detected H2AZK7me1 exclusively in conjunction with H2AZK4me1, suggesting these modifications may function as a coordinated epigenetic mark .

  • Low Abundance: Modified histones often exist at relatively low abundance compared to their unmodified counterparts, making detection challenging. This is especially true for site-specific methylation like H2AZK7me1, which may require highly sensitive detection methods .

  • Similar Molecular Weights: Monomethylation adds only 14 Da to the molecular weight of a protein, making it difficult to distinguish methylated from unmethylated forms by gel electrophoresis alone. Additionally, different methylation sites (e.g., K4 vs. K7) are indistinguishable by simple molecular weight analysis .

  • Mass Spectrometry Resolution: Even with high-resolution mass spectrometry, distinguishing between isomeric peptides with methylation at different lysine residues can be challenging and requires careful MS/MS fragmentation analysis . The research demonstrated this by using detailed MS2 chromatogram analysis to identify specific methylation sites .

  • Mutational Approach Limitations: Site-directed mutagenesis (e.g., K7R) can help assign functional effects to specific lysine residues, but changing an amino acid can alter protein structure and function independently of the loss of methylation, complicating interpretation .

  • Enzyme Specificity: While SETD6 preferentially methylates K7, it can also methylate K4 at lower levels. Similarly, SET7 can methylate multiple sites on H2AZ. This lack of absolute enzyme specificity makes it difficult to manipulate just one modification experimentally .

Addressing these challenges requires integrating multiple approaches, including site-specific antibodies, mass spectrometry, enzyme knockdown/knockout experiments, and careful controls to interpret the specific contribution of H2AZK7me1 to biological processes .

How can researchers optimize mass spectrometry protocols for detecting low abundance H2AZK7me1 in complex biological samples?

Optimizing mass spectrometry protocols for detecting low abundance H2AZK7me1 requires several specialized approaches:

  • Enrichment of Histone Fractions: Begin with acid extraction of histones from nuclei to enrich for all histone proteins before further processing. This reduces sample complexity and increases the relative abundance of histone modifications .

  • Targeted Immunoprecipitation: Use H2AZ-specific antibodies to enrich for the histone variant before mass spectrometry analysis. This can be followed by a second enrichment using methylation-specific antibodies if available .

  • Chemical Derivatization: Modify histones with propionic anhydride before and after trypsin digestion to convert lysines to propionyl-lysines. This prevents trypsin from cleaving after every lysine and creates larger, more readily identifiable peptide fragments containing the modifications of interest .

  • Sample Fractionation: Implement strong cation exchange (SCX) or hydrophilic interaction liquid chromatography (HILIC) fractionation prior to LC-MS/MS analysis to separate peptides based on charge or hydrophilicity, reducing sample complexity .

  • Multiple Reaction Monitoring (MRM): Develop targeted MRM assays specific for H2AZK7me1-containing peptides. This approach increases sensitivity by monitoring specific precursor-to-fragment ion transitions characteristic of the modified peptide .

  • Parallel Reaction Monitoring (PRM): Utilize high-resolution mass spectrometers with PRM capabilities to monitor all fragment ions from a selected precursor ion, increasing specificity and sensitivity for H2AZK7me1 detection .

  • Internal Standards: Include isotopically labeled synthetic peptides corresponding to the H2AZK7me1 modification as internal standards for accurate quantification. The research used biotinylated peptides with specific modifications (mono-, di-, or tri-methylated H2AZK7) for in vitro methylation assays that could be adapted for MS quantification .

  • Enhanced Chromatographic Separation: Optimize liquid chromatography parameters, including extended gradient times and specialized columns, to improve separation of isobaric peptides that differ only in the position of methylation (e.g., K4me1 vs. K7me1) .

  • MS/MS Fragmentation Optimization: Fine-tune collision energies and fragmentation methods (CID, HCD, ETD) to maximize diagnostic fragment ions that distinguish between methylation at different sites. The research demonstrated this by using detailed MS2 chromatogram analysis to identify specific methylation sites with distinct molecular masses (e.g., 766.49 for K7 methylation) .

  • Data Analysis Refinement: Employ specialized software and manual validation to accurately assign methylation sites, particularly when analyzing complex mixtures of modified peptides with similar masses and retention times .

By combining these approaches, researchers can significantly improve the detection sensitivity and quantification accuracy of low abundance H2AZK7me1 in complex biological samples.

What are the current hypotheses regarding the molecular mechanisms by which H2AZK7me1 influences gene expression?

Several hypotheses have been proposed regarding the molecular mechanisms by which H2AZK7me1 influences gene expression:

  • Cooperative Repression with H3K27me3: H2AZK7me1 co-localizes with the repressive H3K27me3 mark near the transcription start sites of developmental genes in mESCs . This co-occurrence suggests these marks may cooperatively repress gene expression, potentially through recruitment of similar or coordinated repressive complexes.

  • Inhibition of Activating Modifications: Methylation of H2AZ at K7 likely prevents acetylation at the same residue. Since H2AZ acetylation is associated with transcriptional activation, H2AZK7me1 may repress gene expression by blocking these activating marks . The research noted that "methylation of H2AZ at lysines 4 and 7 could prevent subsequent acetylation at these sites, thereby impairing the reported transcriptional functions associated with the acetylation of H2AZ" .

  • Recruitment of Specific Reader Proteins: H2AZK7me1 may serve as a binding site for specific "reader" proteins that recognize this modification and subsequently recruit additional factors involved in transcriptional repression. The research suggests that "the eventual identification of H2AZ- or H2AZK7me1-specific readers will undoubtedly help concretize the molecular mechanism of chromatin signaling by H2AZK7me1" .

  • Nucleosome Stability Regulation: The methylation of H2AZ may alter nucleosome stability or positioning, affecting the accessibility of DNA to transcription factors and the transcriptional machinery. This is supported by H2AZ's known role in PRC2 chromatin association .

  • Crosstalk with Histone Ubiquitylation: The observation of a slow-migrating band detected by the H2AZK7me1 antibody suggests potential cross-talk between methylation and ubiquitylation of H2AZ . This interaction could create complex regulatory circuits controlling gene expression.

  • Developmental Gene Regulation Program: The loss of both H2AZK7me1 and H3K27me3 from developmental gene promoters upon cellular differentiation suggests these marks function together in an epigenetic program that regulates expression of lineage commitment genes . The research states that "the loss of both H2AZK7me1 and H3K27me3 from Fgf5 upon cellular differentiation implies that H2AZK7me1 could be involved with H3K27me3 in an epigenetic program that regulates the expression of lineage commitment genes" .

  • Context-Dependent Effects: While global levels of H2AZK7me1 increase during differentiation, the mark is specifically lost from certain developmental gene promoters, suggesting its effects on gene expression may be highly context-dependent and influenced by local chromatin environment .

These hypotheses are not mutually exclusive, and the actual mechanism likely involves multiple modes of action depending on the genomic context and cellular state. Further research, particularly the identification of specific H2AZK7me1 readers, is needed to fully elucidate these mechanisms .

What are the most promising approaches for identifying the readers and erasers of the H2AZK7me1 mark?

Identifying the readers and erasers of the H2AZK7me1 mark requires specialized techniques that can capture proteins specifically interacting with this modification. The most promising approaches include:

  • Modified Peptide Pull-downs: Synthesize biotinylated peptides corresponding to the N-terminal tail of H2AZ with and without K7 methylation. Use these as baits to capture proteins from nuclear extracts that specifically bind to the methylated form. The research already utilized biotinylated and mono-, di-, or tri-methylated H2AZK7 peptides for in vitro studies , which could be adapted for pull-down experiments.

  • CRISPR-based Proximity Labeling: Combine CRISPR-mediated targeting with proximity labeling enzymes (BioID, APEX2) fused to reader domains known to bind methylated lysines. Target these constructs to genomic regions enriched for H2AZK7me1 to identify proteins in proximity to this mark in living cells.

  • Chemical Crosslinking Mass Spectrometry (XLMS): Use crosslinking agents to capture transient interactions between H2AZK7me1 and its binding partners, followed by mass spectrometry identification. This approach can preserve weak or dynamic interactions that might be lost in standard pull-down assays.

  • Quantitative Interaction Proteomics: Compare the interactomes of wild-type H2AZ versus K7R mutant (cannot be methylated) or K7Q mutant (mimics acetylation) to identify differential binding partners dependent on the K7 methylation state.

  • Domain-focused Protein Arrays: Screen libraries of known chromatin-associated domains (e.g., PHD fingers, chromodomains, Tudor domains) for specific binding to H2AZK7me1 peptides. These domains are common readers of histone methylation marks.

  • Enzymatic Activity Assays for Erasers: Develop in vitro demethylation assays using purified H2AZK7me1 substrates and candidate lysine demethylases. The research noted that "whether H2AZK7me1 is demethylated by a demethylase or evicted by a histone exchange complex remains to be uncovered" , highlighting the need for such assays.

  • Genomic Screens for Erasers: Perform CRISPR screens to identify enzymes whose loss leads to increased H2AZK7me1 levels, potentially indicating a role in removing this mark.

  • Histone Dynamics Studies: Use techniques like SNAP-tag labeling or FRAP (Fluorescence Recovery After Photobleaching) to study the dynamics of H2AZK7me1-containing nucleosomes, potentially revealing mechanisms of histone exchange that might function alongside enzymatic demethylation.

  • Candidate Approach Based on Known Methylation Readers/Erasers: Test known methyl-lysine readers (e.g., members of the MBT, PHD, Tudor protein families) and erasers (e.g., LSD1/KDM1A, JmjC domain proteins) for their ability to recognize or remove H2AZK7me1.

  • Differential Gene Expression in SETD6-depleted Cells: Analyze gene expression changes in cells depleted of SETD6 to identify potential regulatory pathways affected by H2AZK7me1 loss, providing clues to proteins that might function downstream of this mark .

The research highlights that "the eventual identification of H2AZ- or H2AZK7me1-specific readers will undoubtedly help concretize the molecular mechanism of chromatin signaling by H2AZK7me1" , underscoring the importance of these approaches for understanding the complete regulatory circuit of this epigenetic modification.

How might researchers integrate H2AZK7me1 ChIP-seq data with other epigenomic datasets to gain comprehensive insights into its regulatory functions?

Integrating H2AZK7me1 ChIP-seq data with other epigenomic datasets requires sophisticated computational approaches to reveal comprehensive insights into its regulatory functions:

  • Multi-mark Co-occupancy Analysis: Compare the genome-wide distribution of H2AZK7me1 with other histone modifications, particularly H3K27me3 which co-localizes with H2AZK7me1 at developmental gene promoters . Identify regions of overlap and exclusivity to map the combinatorial chromatin landscape.

  • Chromatin State Segmentation: Apply algorithms like ChromHMM or Segway to integrate H2AZK7me1 data with other histone modifications, creating a comprehensive chromatin state map that defines functional genomic elements (e.g., poised promoters, repressed enhancers).

  • Transcription Factor Binding Correlation: Integrate with transcription factor ChIP-seq datasets to identify factors that preferentially associate with or are excluded from H2AZK7me1-marked regions, potentially revealing functional interactions.

  • RNA Expression Correlation: Combine with RNA-seq data to establish relationships between H2AZK7me1 occupancy and gene expression levels across different cellular states, particularly during differentiation when H2AZK7me1 levels change dynamically .

  • Chromatin Accessibility Integration: Overlay with ATAC-seq or DNase-seq data to determine how H2AZK7me1 correlates with chromatin accessibility and nucleosome positioning, especially at developmental gene promoters.

  • Three-dimensional Chromatin Organization: Integrate with Hi-C or other chromosome conformation capture datasets to examine how H2AZK7me1-marked regions participate in higher-order chromatin structures and long-range interactions.

  • Methyltransferase Dependency Networks: Compare H2AZK7me1 ChIP-seq profiles between wild-type and SETD6-depleted cells to identify regions most dependent on SETD6 activity, potentially highlighting direct versus indirect regulation .

  • Differentiation Dynamics Analysis: Perform time-course ChIP-seq during cellular differentiation to track the dynamic changes in H2AZK7me1 occupancy at specific genomic loci, correlating these changes with other epigenetic marks and gene expression changes .

  • PRC2 Complex Co-occupancy: Integrate with ChIP-seq data for PRC2 complex components (e.g., SUZ12, EED) which co-localize with H2AZ at developmental gene promoters to examine their relationship with H2AZK7me1 specifically .

  • Motif Enrichment Analysis: Identify DNA sequence motifs enriched at H2AZK7me1 sites to predict transcription factors that might influence H2AZK7me1 deposition or function.

  • Cell Type-Specific Comparisons: Compare H2AZK7me1 profiles across different cell types, particularly contrasting pluripotent stem cells with differentiated lineages, to identify cell type-specific functions .

  • Pathway and Ontology Enrichment: Analyze the biological pathways and gene ontology terms associated with H2AZK7me1-marked genes to understand the broader biological processes regulated by this modification.

The research suggests that "genome-wide studies, such as H2AZ and H2AZK7me1 ChIPseq, may reveal more precisely their biological functions" , highlighting the value of these integrative approaches. Such comprehensive analyses would provide a systems-level understanding of how H2AZK7me1 functions within the broader epigenetic landscape to regulate gene expression and cellular identity.

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.