HIST1H4A (Ab-91) Antibody

Shipped with Ice Packs
In Stock

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 order within 1-3 business days of receiving it. The delivery timeframe may vary based on the purchase method or location. Please consult your local distributors for specific delivery details.
Synonyms
dJ160A22.1 antibody; dJ160A22.2 antibody; dJ221C16.1 antibody; dJ221C16.9 antibody; FO108 antibody; H4 antibody; H4.k antibody; H4/a antibody; H4/b antibody; H4/c antibody; H4/d antibody; H4/e antibody; H4/g antibody; H4/h antibody; H4/I antibody; H4/j antibody; H4/k antibody; H4/m antibody; H4/n antibody; H4/p antibody; H4_HUMAN antibody; H4F2 antibody; H4F2iii antibody; H4F2iv antibody; H4FA antibody; H4FB antibody; H4FC antibody; H4FD antibody; H4FE antibody; H4FG antibody; H4FH antibody; H4FI antibody; H4FJ antibody; H4FK antibody; H4FM antibody; H4FN antibody; H4M antibody; HIST1H4A antibody; HIST1H4B antibody; HIST1H4C antibody; HIST1H4D antibody; HIST1H4E antibody; HIST1H4F antibody; HIST1H4H antibody; HIST1H4I antibody; HIST1H4J antibody; HIST1H4K antibody; HIST1H4L antibody; HIST2H4 antibody; HIST2H4A antibody; Hist4h4 antibody; Histone 1 H4a antibody; Histone 1 H4b antibody; Histone 1 H4c antibody; Histone 1 H4d antibody; Histone 1 H4e antibody; Histone 1 H4f antibody; Histone 1 H4h antibody; Histone 1 H4i antibody; Histone 1 H4j antibody; Histone 1 H4k antibody; Histone 1 H4l antibody; Histone 2 H4a antibody; histone 4 H4 antibody; Histone H4 antibody; MGC24116 antibody
Target Names
HIST1H4A
Uniprot No.

Target Background

Function
Histone H4 is a core component of the nucleosome, the fundamental unit of chromatin structure. Nucleosomes wrap and compact DNA, restricting access to cellular machinery that requires DNA as a template. Histones play a critical role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. DNA accessibility is intricately regulated through various post-translational modifications of histones, collectively referred to as the 'histone code,' along with nucleosome remodeling mechanisms.
Gene References Into Functions
  1. Studies show that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research suggests that post-translational histone modifications, specifically trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), are involved in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand break, and this process requires the involvement of SETD2, LEDGF, and KAT5 (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5). PMID: 28546430
  3. Data indicate that Omomyc protein co-localizes with proto-oncogene protein c-myc (c-Myc), protein arginine methyltransferase 5 (PRMT5), and histone H4 H4R3me2s-enriched chromatin domains. PMID: 26563484
  4. H4K12ac is regulated by estrogen receptor-alpha and is linked to BRD4 function and inducible transcription. PMID: 25788266
  5. Systemic lupus erythematosus appears to be associated with an imbalance in histone acetyltransferases and histone deacetylase enzymes, favoring pathological H4 acetylation. PMID: 25611806
  6. Sumoylated human histone H4 inhibits chromatin compaction by preventing long-range internucleosomal interactions. PMID: 25294883
  7. Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during the reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  8. Increased histone H4 acetylation caused by hypoxia in human neuroblastoma cell lines corresponds to elevated levels of N-myc transcription factor. PMID: 24481548
  9. Research suggests that G1-phase histone assembly is limited to CENP-A and H4. PMID: 23363600
  10. This study focused on the distribution of a specific histone modification, namely H4K12ac, in human sperm and characterized its specific enrichment sites in promoters throughout the human genome. PMID: 22894908
  11. SRP68/72 heterodimers are major nuclear proteins whose binding of histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  12. TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is attributed to an epigenetic mechanism involving the suppression of histone H4 acetylation. PMID: 21973049
  13. Findings indicate that global histone H3 and H4 modification patterns serve as potential markers for tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  14. HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  15. Phosphorylation of histone H4 Ser 47, catalyzed by the PAK2 kinase, promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by enhancing the binding affinity of HIRA to H3.3-H4 and reducing the association of CAF-1 with H3.1-H4. PMID: 21724829
  16. The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4 modifications. PMID: 20949922
  17. Findings reveal the molecular mechanisms by which DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200, which, in turn, reduces gene expression. PMID: 20512922
  18. Histone H4 expression is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, possibly through alterations in gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation levels of histone H4 protein are closely related. PMID: 15095300
  21. Peptidylarginine deiminase 4 (PAD4) regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. Data suggest that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones. PMID: 15345777
  22. The lack of biotinylation of K12 in histone H4 is an early signaling event in response to double-strand breaks. PMID: 16177192
  23. Incorporation of acetylated histone H4-K16 into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and impedes the ability of chromatin to form cross-fiber interactions. PMID: 16469925
  24. Apoptosis is associated with global DNA hypomethylation and histone deacetylation events in leukemia cells. PMID: 16531610
  25. BTG2 contributes to retinoic acid activity by favoring differentiation through a gene-specific modification of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  26. There is a relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. The H4 tail and its acetylation have novel roles in mediating the recruitment of multiple regulatory factors that can alter chromatin states for transcription regulation. PMID: 17548343
  28. Brd2 bromodomain 2 is monomeric in solution and dynamically interacts with H4-AcK12; additional secondary elements in the long ZA loop may be a common characteristic of BET bromodomains. PMID: 17848202
  29. Spermatids Hypac-H4 impairment in mixed atrophy did not deteriorate further by AZFc region deletion. PMID: 18001726
  30. The SET8 and PCNA interaction couples H4-K20 methylation with DNA replication. PMID: 18319261
  31. H4K20 monomethylation and PR-SET7 are crucial for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more common in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Findings indicate an important role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results show that acetylation of histone H4 K16 during S-phase leads to early replicating chromatin domains acquiring the H4K16ac-K20me2 epigenetic label that persists throughout mitosis and is deacetylated in early G1-phase of the next cell cycle. PMID: 19348949
  35. Acetylated H4 is overexpressed in diffuse large B-cell lymphoma and peripheral T-cell lymphoma compared to normal lymphoid tissue. PMID: 19438744
  36. The release of histone H4 by holocrine secretion from the sebaceous gland may play a significant role in innate immunity. PMID: 19536143
  37. Histone modification, including PRC2-mediated repressive histone marker H3K27me3 and active histone marker acH4, may be involved in CD11b transcription during HL-60 leukemia cells reprogramming to terminal differentiation. PMID: 19578722
  38. A role of Cdk7 in regulating elongation is further supported by enhanced histone H4 acetylation and diminished histone H4 trimethylation on lysine 36 - two marks of elongation - within genes when the kinase was inhibited. PMID: 19667075
  39. Data show the dynamic fluctuation of histone H4 acetylation levels during mitosis, as well as acetylation changes in response to structurally distinct histone deacetylase inhibitors. PMID: 19805290
  40. Data directly implicate BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714

Show More

Hide All

Database Links

HGNC: 4781

OMIM: 142750

KEGG: hsa:121504

STRING: 9606.ENSP00000367034

UniGene: Hs.143080

Involvement In Disease
Chromosomal aberrations involving HISTONE H4 is a cause of B-cell non-Hodgkin lymphomas (B-cell NHL). Translocation t(3;6)(q27;p21), with BCL6.
Protein Families
Histone H4 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is HIST1H4A and why is it significant in epigenetic research?

HIST1H4A is a gene that encodes histone H4, one of the most conserved cell cycle-related histones in eukaryotic organisms. Histone H4 is crucial for chromatin structure as it forms part of the histone octamer around which DNA is wrapped. Post-translational modifications of histone H4, particularly acetylation, play fundamental roles in regulating chromatin assembly, DNA repair, transcriptional silencing, and cellular differentiation. The significance of HIST1H4A in epigenetic research stems from its central role in chromatin dynamics, where modifications at specific residues serve as important epigenetic marks that influence gene expression and genome stability. Histone H4 lysine 91 is particularly noteworthy as it is located at the interface between H3/H4 tetramers and H2A/H2B dimers, meaning its modification can directly affect nucleosome structure and stability .

What epitope does the HIST1H4A (Ab-91) antibody recognize and how does this impact its research applications?

The HIST1H4A (Ab-91) antibody recognizes a peptide sequence surrounding lysine 91 (Lys91) of human histone H4 . This specificity is significant because Lys91 is located in the globular core domain of histone H4 rather than in the NH2-terminal tail where most well-studied modifications occur. Acetylation at Lys91 has been shown to be involved in chromatin assembly processes and maintenance of genomic stability. The antibody's specificity for this region makes it particularly valuable for research investigating core domain modifications that affect nucleosome stability and histone-DNA interactions. This epitope recognition enables researchers to study a modification site that influences fundamental processes like DNA repair, transcriptional silencing, and chromatin assembly through techniques such as ChIP, Western blotting, and immunohistochemistry .

How does HIST1H4A differ from other histone H4 variants and why should researchers be aware of these differences?

HIST1H4A represents one of several histone H4 encoding genes in mammals. While the protein sequence of histone H4 is extremely conserved across variants (with all human H4 proteins having identical amino acid sequences), the genes differ in their promoter regions, regulatory elements, and expression patterns during the cell cycle. At least 12 different promoters regulate the transcription of mouse histone H4, and similar complexity exists in humans . The HIST1H4A gene belongs to the histone cluster 1 on chromosome 6 in humans.

Researchers should be aware of these differences because even though the protein products are identical, the regulation of expression differs between variants. For example, studies have shown that different H4 promoters contain varying transcription factor binding sites, with 10 of 12 mouse histone H4 promoters containing C/EBP-binding sites that can be transactivated by C/EBPβ . This differential regulation becomes particularly important when studying cell cycle-dependent histone expression, cellular differentiation, or when interpreting gene expression data. When using antibodies against histone H4, researchers should remember that they are likely detecting all H4 variants simultaneously unless the antibody is specifically directed against a unique post-translational modification pattern .

What are the validated applications for HIST1H4A (Ab-91) antibody and what sample types work best with each technique?

The HIST1H4A (Ab-91) antibody has been validated for several key applications in epigenetic research:

  • Western Blotting (WB): The antibody effectively detects histone H4 in protein lysates from both human and mouse samples. For optimal results, researchers should use nuclear extracts or histone-enriched fractions rather than whole cell lysates to increase specificity and reduce background .

  • Chromatin Immunoprecipitation (ChIP): The antibody successfully precipitates chromatin fragments containing histone H4 with Lys91 modifications. ChIP experiments with this antibody are valuable for mapping the genomic locations where this specific modification occurs. This application works best with cross-linked chromatin from cultured cells or tissue samples .

  • Immunohistochemistry (IHC): The antibody can detect histone H4 in fixed tissue sections, allowing for the spatial analysis of histone modifications in complex tissues .

  • Enzyme-Linked Immunosorbent Assay (ELISA): The antibody functions effectively in ELISA applications for quantitative detection of histone H4 proteins or peptides with the specific modification .

For all applications, the antibody shows strong reactivity with human and mouse samples, making it versatile for comparative studies across these species . When designing experiments, researchers should optimize antibody concentrations for each specific application and sample type, as the optimal dilution may vary based on the technique and the abundance of the target histone modification.

What is the optimal protocol for chromatin immunoprecipitation (ChIP) experiments using the HIST1H4A (Ab-91) antibody?

For optimal ChIP experiments using HIST1H4A (Ab-91) antibody, researchers should follow this methodological approach:

Sample Preparation and Chromatin Isolation:

  • Cross-link protein-DNA complexes in living cells with 1% formaldehyde for 10 minutes at room temperature.

  • Quench the cross-linking reaction with 125 mM glycine for 5 minutes.

  • Isolate nuclei using cell lysis buffer and resuspend in nuclear lysis buffer.

  • Sonicate chromatin to generate fragments of 200-500 bp in length, verifying fragment size by agarose gel electrophoresis.

Immunoprecipitation:

  • Pre-clear chromatin with protein A/G beads and non-immune IgG.

  • Incubate pre-cleared chromatin with HIST1H4A (Ab-91) antibody (2-5 μg per reaction) overnight at 4°C with rotation.

  • Include appropriate controls: IgG negative control and a positive control antibody against a well-characterized histone mark.

  • Add protein A/G beads and incubate for 2-3 hours at 4°C.

  • Perform stringent washing steps to remove non-specific binding.

DNA Recovery and Analysis:

  • Reverse cross-links by incubating samples at 65°C overnight.

  • Treat with RNase A and proteinase K.

  • Purify DNA using phenol-chloroform extraction or commercial kits.

  • Analyze enriched DNA by qPCR, sequencing, or other downstream applications.

This protocol has been effectively used in studies examining histone H4 modifications in the context of transcriptional activation during cell differentiation, as demonstrated in prior research on histone H4 promoter binding by transcription factors . Researchers should always include appropriate controls and validate results through complementary methods such as Western blotting to confirm specificity of the antibody for the target modification .

How should researchers optimize Western blot conditions for detecting H4K91 acetylation using the HIST1H4A (Ab-91) antibody?

Optimizing Western blot conditions for detecting H4K91 acetylation using the HIST1H4A (Ab-91) antibody requires careful attention to several methodological aspects:

Sample Preparation:

  • Extract histones using acid extraction methods (0.2N HCl or 0.4N H₂SO₄) to enrich for basic nuclear proteins.

  • Include histone deacetylase inhibitors (e.g., sodium butyrate, trichostatin A) in lysis buffers to preserve acetylation marks.

  • Quantify protein concentration using Bradford or BCA assay, loading 5-15 μg of histone-enriched extract per lane.

Gel Electrophoresis and Transfer:

  • Use specialized gel systems for low molecular weight proteins (15-18% SDS-PAGE or Triton-Acid-Urea gels).

  • Include positive controls such as commercial histone extracts with known modification status.

  • Transfer to PVDF membranes (rather than nitrocellulose) at lower voltage (30V) overnight at 4°C to ensure efficient transfer of small proteins.

Antibody Incubation and Detection:

  • Block membranes with 5% BSA (not milk, which contains histones and deacetylases).

  • Incubate with HIST1H4A (Ab-91) antibody at optimized dilution (typically 1:500 to 1:2000) in TBST with 1% BSA overnight at 4°C.

  • Use high-sensitivity detection methods such as enhanced chemiluminescence (ECL) or fluorescent secondary antibodies.

  • Consider using antibodies against total histone H4, histone H3, or actin as loading controls.

Critical Controls:

  • Include a non-modified histone H4 control to demonstrate specificity.

  • Use lysates from cells treated with HDAC inhibitors as positive controls for increased acetylation.

  • Consider using samples with known mutations at H4K91 as negative controls to validate specificity.

Following these optimized conditions will help researchers obtain clear, specific signals for H4K91 acetylation while minimizing background and non-specific binding. This is particularly important when studying core domain modifications like H4K91ac, which may be less abundant than some tail modifications and can be challenging to detect without proper optimization .

How does H4K91 acetylation interact with other histone modifications to regulate chromatin structure and gene expression?

H4K91 acetylation functions within a complex network of histone modifications that collectively regulate chromatin structure and gene expression. This particular modification site is distinctive because it is located in the globular core domain at the interface between histone H3/H4 tetramers and H2A/H2B dimers, rather than in the more commonly studied histone tails .

The interaction between H4K91 acetylation and other modifications involves several key mechanisms:

  • Nucleosome Stability Regulation: H4K91 acetylation has been shown to destabilize the histone octamer by disrupting the electrostatic interactions at the interface between the H3/H4 tetramer and H2A/H2B dimers. This structural impact works in concert with tail modifications that regulate higher-order chromatin folding, creating a multi-level system of chromatin accessibility regulation .

  • Histone Deposition Coordination: H4K91 acetylation by type B histone acetyltransferases occurs during chromatin assembly, working in coordination with H4 tail acetylations (particularly at K5 and K12) that are also associated with histone deposition during DNA replication and repair .

  • Transcriptional Regulation Crosstalk: While tail modifications like H3K4me3 and H3K27ac directly interact with transcription factors and chromatin remodelers to regulate gene expression, H4K91ac may influence these processes indirectly by affecting nucleosome stability and positioning. Research suggests potential crosstalk between core domain modifications like H4K91ac and tail modifications in regulating transcriptional states .

  • DNA Repair Pathway Integration: H4K91 acetylation has been implicated in DNA repair processes, working alongside modifications like γH2AX and H4K16ac that are known to regulate the DNA damage response. Mutations at H4K91 confer sensitivity to DNA damaging agents, suggesting coordinated function with other histone modifications in maintaining genome integrity .

Understanding these complex interactions requires sophisticated experimental approaches, including sequential ChIP (re-ChIP) to detect co-occurrence of modifications, mass spectrometry to identify modification patterns, and genetic studies manipulating specific histone residues to determine functional relationships between different modifications .

What challenges exist in distinguishing between different H4 acetylation sites when using antibody-based detection methods?

Distinguishing between different histone H4 acetylation sites presents several significant challenges when using antibody-based detection methods:

  • Epitope Similarity and Cross-Reactivity: The amino acid sequences surrounding different lysine residues in histone H4 can be similar, potentially leading to antibody cross-reactivity. For example, antibodies generated against H4K91ac might cross-react with other acetylated lysines if the surrounding peptide sequences share homology. Researchers must validate antibody specificity through multiple approaches including peptide competition assays and using samples with known modification patterns .

  • Modification Density Effects: Histone H4 can be simultaneously modified at multiple lysine residues (K5, K8, K12, K16, K91), and the presence of adjacent modifications can sterically hinder antibody binding. This "epitope masking" can lead to false negative results where a modification is present but not detected because nearby modifications prevent antibody access .

  • Context-Dependent Antibody Performance: The performance of histone modification antibodies, including those against H4 acetylation sites, can vary dramatically depending on the experimental context (ChIP vs. Western blot vs. immunofluorescence). An antibody that works well in Western blot may perform poorly in ChIP experiments due to differences in how the epitope is presented in these different conditions .

  • Lot-to-Lot Variability: Polyclonal antibodies like HIST1H4A (Ab-91) can exhibit significant lot-to-lot variation in specificity and sensitivity. This necessitates careful characterization of each new antibody lot using appropriate controls and validation procedures .

  • Detection of Low-Abundance Modifications: Core domain modifications like H4K91ac may be less abundant than some tail modifications, making their detection more challenging and requiring more sensitive detection methods or enrichment procedures .

To address these challenges, researchers should:

  • Use complementary detection methods such as mass spectrometry to validate antibody-based findings

  • Perform extensive controls including peptide competition assays

  • Consider developing site-specific approaches such as using genetically engineered histones with specific mutations as negative controls

  • Implement quantitative approaches that can account for differences in antibody affinity when comparing different modification sites

How does C/EBPβ-mediated transcriptional regulation of histone H4 influence cell proliferation and differentiation?

C/EBPβ-mediated transcriptional regulation of histone H4 plays a critical role in coordinating cell proliferation and differentiation, particularly during processes like adipocyte differentiation. The mechanism involves several interconnected pathways:

  • Direct Transcriptional Activation of Histone H4 Genes: C/EBPβ directly binds to specific C/EBP-binding sites in histone H4 gene promoters. Research has identified that 10 of 12 mouse histone H4 promoters contain C/EBP-binding sites and can be transactivated by C/EBPβ. This was confirmed through multiple experimental approaches including electrophoretic mobility shift assays (EMSA), chromatin immunoprecipitation (ChIP), and reporter gene assays .

  • Cell Cycle Regulation During Mitotic Clonal Expansion (MCE): In 3T3-L1 preadipocyte differentiation, C/EBPβ is required for mitotic clonal expansion, a process where growth-arrested preadipocytes synchronously reenter the cell cycle. C/EBPβ undergoes sequential phosphorylation and activation at the G1/S boundary, coinciding with histone H4 expression. This temporal coordination is critical, as histone synthesis is tightly coupled with DNA replication during S phase .

  • Functional Consequences of C/EBPβ-Histone H4 Regulation:

    • Knockdown of C/EBPβ using stealth RNAi results in decreased histone H4 expression

    • This reduction in histone H4 expression causes more cells to remain in G0/G1 phase and fewer cells to transition to S phase

    • The inhibition of cell cycle progression ultimately impairs terminal differentiation into adipocytes

    • Conversely, overexpression of C/EBPβ increases histone H4 expression and promotes cell cycle progression

  • Molecular Mechanism: C/EBPβ binds to specific sequences in histone H4 promoters, particularly in the hist4h4 promoter region between -125 to -117, which resembles the classic C/EBP-binding consensus sequence. This binding initiates transcription of histone H4, providing the necessary histone proteins for chromatin assembly during DNA replication .

This regulatory mechanism establishes a critical link between a master transcriptional regulator (C/EBPβ), cell cycle progression (through histone H4 expression), and cellular differentiation. The significance extends beyond adipocyte differentiation, as similar C/EBPβ-mediated regulation of histone genes may occur in other tissues where C/EBPβ plays important roles in proliferation and differentiation .

What are the most common technical challenges when using HIST1H4A (Ab-91) antibody in ChIP experiments, and how can researchers overcome them?

Researchers working with HIST1H4A (Ab-91) antibody in ChIP experiments frequently encounter several technical challenges that can be addressed through specific optimization strategies:

  • Low Signal-to-Noise Ratio

    • Challenge: High background or weak specific signal due to the relatively low abundance of core histone modifications compared to tail modifications.

    • Solutions:

      • Increase chromatin amount (up to 25-30 μg per IP reaction)

      • Optimize antibody concentration through titration experiments (2-5 μg recommended)

      • Implement additional pre-clearing steps with protein A/G beads

      • Use more stringent washing conditions with increasing salt concentrations (150 mM to 500 mM NaCl)

  • Cross-Reactivity Issues

    • Challenge: The antibody may recognize other acetylated lysine residues in histones, particularly when using standard ChIP protocols.

    • Solutions:

      • Perform peptide competition assays to verify specificity

      • Include appropriate negative controls (IgG and unmodified H4 peptide)

      • Consider using native ChIP (without crosslinking) which may preserve epitope structure better

      • Validate findings with alternative approaches such as CUT&RUN or CUT&Tag

  • Inconsistent Chromatin Fragmentation

    • Challenge: Over- or under-sonication affects ChIP efficiency for core histone modifications.

    • Solutions:

      • Carefully optimize sonication conditions for each cell type

      • Aim for 200-500 bp fragments (verify by agarose gel electrophoresis)

      • Consider enzymatic fragmentation methods as alternatives

      • Implement standardized protocols with precise timing and power settings

  • Epitope Masking

    • Challenge: The H4K91 site may be inaccessible in certain chromatin contexts due to nucleosome structures or adjacent modifications.

    • Solutions:

      • Try different crosslinking protocols (reducing formaldehyde concentration or time)

      • Include detergents (0.1% SDS or 1% Triton X-100) in IP buffer

      • Consider two-step crosslinking with protein-protein crosslinkers followed by formaldehyde

      • Use epitope retrieval techniques adapted from immunohistochemistry

  • Reproducibility Issues

    • Challenge: Variation between experiments and antibody lots.

    • Solutions:

      • Standardize all buffers and procedures

      • Perform biological replicates with the same antibody lot

      • Include spike-in controls (e.g., Drosophila chromatin) for normalization

      • Implement quantitative PCR with multiple primer sets for validation

By systematically addressing these challenges through methodical optimization, researchers can significantly improve the quality and reproducibility of ChIP experiments using the HIST1H4A (Ab-91) antibody, enabling more reliable studies of H4K91 acetylation patterns throughout the genome.

How can researchers validate the specificity of HIST1H4A (Ab-91) antibody for H4K91 acetylation versus other histone acetylation marks?

Validating antibody specificity is crucial for accurate interpretation of experimental results, particularly for histone modification antibodies where cross-reactivity is a common concern. To validate the specificity of HIST1H4A (Ab-91) antibody for H4K91 acetylation versus other histone acetylation marks, researchers should implement a comprehensive multi-method approach:

  • Peptide Competition Assays

    • Conduct Western blot or immunoprecipitation experiments in the presence of increasing concentrations of:

      • H4K91ac-modified peptides (should block signal)

      • Unmodified H4 peptides containing K91 (should not block signal)

      • Peptides with acetylation at other lysine residues (e.g., H4K5ac, H4K8ac, H4K12ac) (should not block signal if antibody is specific)

    • A truly specific antibody will show signal reduction only with the H4K91ac peptide

  • Dot Blot Analysis with Modified Peptide Arrays

    • Use commercially available or custom-synthesized peptide arrays containing:

      • H4K91ac peptides at different concentrations

      • H4 peptides with acetylation at other sites (K5, K8, K12, K16)

      • Other histone peptides with acetylated lysines in similar sequence contexts

    • Quantify and compare binding affinity across different modified peptides

  • Genetic Validation Using Mutant Histones

    • Express mutant histone H4 with K91R or K91Q substitutions (mimicking unacetylated or acetylated states)

    • If antibody is specific, it should:

      • Not recognize K91R mutants

      • Show reduced or no signal in Western blots or immunofluorescence with these mutants

    • This approach provides the most stringent biological validation of specificity

  • Mass Spectrometry Correlation

    • Perform immunoprecipitation with the antibody followed by mass spectrometry

    • Analyze the enriched peptides to confirm they contain acetylated K91

    • Compare the modification profile of immunoprecipitated histones with known modification patterns

    • This approach can identify unexpected cross-reactivities with modifications not tested in peptide arrays

  • HDAC Inhibitor and HAT Studies

    • Treat cells with HDAC inhibitors (increases acetylation) or HAT inhibitors (decreases acetylation)

    • Monitor changes in antibody signal by Western blot or immunofluorescence

    • Compare changes in H4K91ac signal with other acetylation marks

    • A specific antibody should show distinct patterns of change compared to antibodies against other acetylation sites

By implementing this multi-faceted validation approach, researchers can establish the specificity parameters of the HIST1H4A (Ab-91) antibody and clearly document its performance characteristics across different experimental contexts, enabling confident interpretation of results in histone modification studies .

What controls should be included when using HIST1H4A (Ab-91) antibody to study the relationship between histone modifications and chromatin assembly?

When using HIST1H4A (Ab-91) antibody to study the relationship between histone modifications and chromatin assembly, researchers should implement a comprehensive set of controls to ensure experimental validity and interpretability:

Essential Experimental Controls:

  • Antibody Specificity Controls

    • Include peptide competition assays with H4K91ac peptides versus unmodified peptides

    • Use histone H4K91 mutants (K91R or K91Q) as negative and positive mimetics

    • Include known positive samples (cell types with validated H4K91ac) and negative samples (where possible)

  • Chromatin Assembly-Specific Controls

    • Cell Cycle Synchronization Verification: Include flow cytometry analysis to confirm cell cycle stage, as histone deposition occurs primarily during S phase

    • Replication Timing Controls: Compare early versus late replicating genomic regions, which should show different patterns of newly assembled chromatin

    • Pulse-Chase Controls: For studies of new versus old histones, include pulse-chase experiments with labeled histones (e.g., SNAP-tag H4) to distinguish assembly timing

  • Treatment Controls for Validating Biological Function

    • HDAC Inhibitors: Treat cells with trichostatin A (TSA) or sodium butyrate to increase global acetylation levels

    • DNA Damage Agents: Include samples treated with agents like methyl methanesulfonate (MMS) or UV, as H4K91 acetylation is implicated in DNA repair

    • Replication Inhibitors: Use hydroxyurea or aphidicolin to block replication and observe effects on H4K91ac in chromatin assembly

  • Technical Controls for ChIP Experiments

    • Input Control: Include a portion of pre-immunoprecipitation chromatin (5-10%)

    • IgG Control: Use non-specific IgG matching the host species of the primary antibody

    • Additional Histone Mark Controls: Include ChIPs for established deposition-related marks (H4K5ac, H4K12ac) and comparison marks (H3K4me3, H3K27ac)

    • Genomic Region Controls: Analyze both euchromatic and heterochromatic regions as internal controls for specificity

  • Controls for Protein-Protein Interactions

    • When studying histone chaperones or assembly factors:

      • Include co-immunoprecipitation with known H4 binding partners (e.g., CAF-1, HIRA)

      • Use sequential ChIP (re-ChIP) to verify co-occurrence of H4K91ac with deposition factors

      • Include proximity ligation assays to verify spatial relationships between H4K91ac and assembly machinery

Experimental Framework for Comprehensive Analysis:

Control TypeSpecific ControlsPurposeInterpretation
Antibody ValidationPeptide competition, H4K91 mutantsConfirm specificitySignal should disappear with competing peptide or in K91R mutants
Cell CycleSynchronized vs. asynchronous cellsLink to replication timingH4K91ac should show cell cycle-dependent patterns
Chromatin AssemblyReplication inhibition, H4 chaperone knockdownsEstablish dependency on assemblyChanges in H4K91ac pattern after disrupting assembly
Genomic ContextEuchromatin vs. heterochromatin regionsDetermine distribution patternsMay show preference for newly assembled regions
Biological FunctionDNA damage response, transcriptional changesConnect to downstream effectsH4K91ac changes should correlate with functional outcomes

By systematically implementing these controls, researchers can establish robust connections between H4K91 acetylation and chromatin assembly processes while minimizing the risk of experimental artifacts or misinterpretation of results .

How should researchers interpret variations in H4K91 acetylation patterns across different genomic regions and cell types?

Interpreting variations in H4K91 acetylation patterns requires careful consideration of multiple factors that influence the biological significance of this histone modification. Researchers should apply the following analytical framework when evaluating H4K91ac data across genomic regions and cell types:

  • Genomic Context Integration

    • Chromatin State Correlation: Compare H4K91ac distributions with known chromatin states (active, repressed, bivalent). H4K91ac has been linked to chromatin assembly processes, so researchers should examine whether its enrichment correlates with regions of active replication, repair, or transcription .

    • Co-occurrence with Other Modifications: Analyze how H4K91ac patterns overlap with or diverge from other histone modifications (H3K4me3, H3K27ac, H3K9me3). The unique position of K91 at the histone octamer interface suggests it may function differently from tail modifications .

    • Nucleosome Stability Considerations: Regions with high H4K91ac may exhibit different nucleosome stability characteristics. Researchers should correlate H4K91ac patterns with nucleosome positioning data, as this modification can destabilize the histone octamer by disrupting interactions between H3/H4 tetramers and H2A/H2B dimers .

  • Cell Type-Specific Analysis

    • Proliferation Rate Effects: Fast-dividing cell types may show distinct H4K91ac patterns compared to terminally differentiated cells. Compare patterns between proliferating cells (e.g., embryonic stem cells, cancer cell lines) and post-mitotic cells (e.g., neurons) .

    • Lineage-Specific Patterns: Different cell lineages may utilize H4K91ac differently for specialized chromatin functions. Examine whether pattern variations correlate with lineage-specific transcriptional programs or chromatin organizations .

    • Disease State Comparisons: In disease models, altered H4K91ac patterns may indicate dysregulation of chromatin assembly or repair pathways. Compare normal and pathological samples to identify disease-associated changes .

  • Functional Correlation Approaches

    • Transcriptional Output Correlation: Analyze how H4K91ac enrichment relates to gene expression levels using RNA-seq data from matching samples. While not necessarily a direct transcriptional regulator, H4K91ac may influence expression through effects on chromatin structure .

    • DNA Repair Pathway Association: Given the sensitivity of H4K91 mutants to DNA damaging agents, examine H4K91ac patterns at sites of induced DNA damage or in cells with defects in specific repair pathways .

    • Replication Timing Analysis: Compare H4K91ac patterns with replication timing data, as this modification is associated with chromatin assembly during DNA replication .

  • Quantitative Interpretation Guidelines

    • Signal Intensity Considerations: H4K91ac may be less abundant than some tail modifications, so signal intensity should be normalized appropriately when making comparisons.

    • Peak Shape Analysis: Sharp peaks versus broad domains of H4K91ac may indicate different functional roles (e.g., regulatory element marking versus broadly assembled new chromatin).

    • Temporal Dynamics: When possible, analyze H4K91ac patterns across time points (e.g., during differentiation or cell cycle progression) to capture dynamic changes that may not be evident in static comparisons .

By applying this multifaceted analytical approach, researchers can develop nuanced interpretations of H4K91ac variation that connect this core domain modification to its biological functions in chromatin assembly, DNA repair, and genome stability across different cellular contexts .

What is the relationship between H4K91 acetylation and DNA damage response pathways?

The relationship between H4K91 acetylation and DNA damage response (DDR) pathways represents a critical intersection between histone modifications and genome stability maintenance. Research has revealed several important connections:

  • Structural Importance in Chromatin Stability

    • H4K91 is positioned at the interface between the H3/H4 tetramer and H2A/H2B dimers within the nucleosome structure. Acetylation at this site neutralizes the positive charge of lysine, potentially weakening the electrostatic interactions that stabilize the nucleosome .

    • This structural role is significant because chromatin destabilization and increased accessibility are essential early steps in DNA damage detection and repair processes. The strategic location of H4K91 makes its modification particularly impactful for allowing repair machinery to access damaged DNA .

  • Experimental Evidence Linking H4K91ac to DNA Repair

    • Studies using yeast models with H4K91 mutations (K91R or K91Q) have demonstrated increased sensitivity to DNA damaging agents, providing direct genetic evidence for the involvement of this residue in DNA damage responses .

    • Cells with mutations at H4K91 show phenotypes consistent with defects in both chromatin assembly and DNA repair, suggesting these processes are mechanistically linked through this histone modification site .

  • Temporal Dynamics During Repair Processes

    • Current research suggests that H4K91 acetylation may occur during two distinct phases of DNA repair:

      • During initial chromatin relaxation to facilitate damage recognition and processing

      • During repair-coupled nucleosome assembly to restore chromatin structure after repair

    • These dynamics align with the known involvement of H4 acetylation in new histone deposition during replication-coupled and replication-independent nucleosome assembly .

  • Pathway-Specific Involvement

    • While comprehensive mapping across all DNA repair pathways is still emerging, current evidence suggests H4K91ac may have pathway-specific roles:

      • In non-homologous end joining (NHEJ), H4K91ac may facilitate the access and function of end-processing factors

      • In homologous recombination (HR), the modification may be involved in the extensive chromatin remodeling required for strand invasion and recombination

      • In nucleotide excision repair (NER), H4K91ac might contribute to the accessibility of damaged sites and subsequent restoration of chromatin structure

  • Regulatory Mechanisms

    • The specific histone acetyltransferases (HATs) and deacetylases (HDACs) that regulate H4K91 acetylation in the context of DNA damage are still being fully characterized

    • Type B HATs, which are involved in the process of chromatin assembly, appear to play a role in H4K91 acetylation

    • The regulatory pathway likely involves damage sensing kinases (ATM, ATR) that initiate signaling cascades leading to recruitment of chromatin modifiers to damage sites

This relationship between H4K91ac and DNA damage responses highlights how core domain histone modifications can regulate fundamental nuclear processes through both structural and signaling mechanisms. Understanding these connections has important implications for both basic chromatin biology and for conditions where DNA repair is compromised, such as cancer and aging .

How does the current understanding of HIST1H4A function and regulation inform potential therapeutic approaches for diseases with epigenetic dysregulation?

The emerging understanding of HIST1H4A function and regulation, particularly regarding H4K91 acetylation, opens several avenues for potential therapeutic approaches targeting diseases with epigenetic dysregulation:

  • Cancer Treatment Strategies

    • Synthetic Lethality Approaches: Cancer cells with mutations in chromatin assembly or DNA repair pathways may be hypersensitive to perturbations in H4K91 acetylation. Research on H4K91 mutants has shown increased sensitivity to DNA damaging agents, suggesting that modulating this modification could selectively target cancer cells with pre-existing repair deficiencies (e.g., BRCA-mutant tumors) .

    • Combination Therapy Potential: Understanding how H4K91ac influences chromatin structure during DNA repair could inform more effective combinations of epigenetic modulators with traditional genotoxic therapies. For example, drugs affecting HATs or HDACs that regulate H4K91ac could be used to sensitize cancer cells to radiation or chemotherapy .

    • Biomarker Development: Patterns of H4K91 acetylation could serve as biomarkers for chromatin assembly defects in cancer, potentially guiding treatment selection or identifying patients likely to respond to specific therapies .

  • Neurodegenerative Disease Applications

    • Chromatin Stability Maintenance: Neurodegenerative diseases often involve accumulation of DNA damage and chromatin disorganization. Targeting the pathways that regulate H4K91ac might help maintain chromatin stability in neurons, which are particularly vulnerable to DNA damage due to their post-mitotic state and high metabolic activity .

    • Transcriptional Regulation: The effects of H4K91ac on nucleosome stability could influence transcriptional programs relevant to neurodegeneration. Modulating this modification might help restore proper gene expression patterns in affected neurons .

  • Developmental Disorder Insights

    • Chromatin Assembly Modulation: Disorders caused by defects in chromatin assembly might benefit from therapies targeting the pathways that regulate H4K91ac. The importance of this modification in nucleosome assembly suggests it could be leveraged to correct assembly defects .

    • Cell Differentiation Regulation: The role of histone H4 in cell differentiation processes, as evidenced by C/EBPβ-mediated regulation during adipogenesis, suggests that modulating H4 modifications could help correct aberrant differentiation in developmental disorders .

  • Therapeutic Modulation Strategies

    • Targeted Enzyme Inhibitors: Development of specific inhibitors for HATs or HDACs that regulate H4K91ac could provide more precise tools for modulating this modification without the broad effects of current epigenetic drugs .

    • Synthetic Histone Mimetics: Engineered histone proteins or peptides that mimic the effects of H4K91 acetylation could potentially be used to modulate chromatin structure in a targeted manner .

    • Gene Therapy Approaches: For severe disorders caused by mutations in histone genes or their regulatory elements, gene therapy to correct these defects could restore proper histone modification patterns .

  • Translational Challenges and Opportunities

    • Specificity Barriers: The conserved nature of histones and their modifications presents challenges for achieving specificity in therapeutic targeting. Research on the unique context of H4K91 at the histone octamer interface could reveal specific structural features that enable more selective targeting .

    • Delivery Systems: Advanced delivery systems would be needed to target drugs to specific chromatin regions or cell types where H4K91ac modulation would be most beneficial .

    • Combinatorial Approaches: Given the complex interplay between different histone modifications, effective therapies may need to simultaneously target multiple modifications including H4K91ac .

The therapeutic potential of targeting H4K91 acetylation and related pathways represents an emerging frontier in epigenetic medicine. As our understanding of HIST1H4A regulation in chromatin assembly, DNA repair, and transcriptional control continues to evolve, so too will opportunities to translate this knowledge into novel treatments for diseases with epigenetic components .

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.