HIST1H4A (Ab-8) Antibody

Shipped with Ice Packs
In Stock

Description

Target and Specificity

The antibody specifically recognizes the acetylated form of lysine 8 on histone H4 (H4K8ac), a modification linked to chromatin relaxation and transcriptional activation. Key features include:

  • Immunogen: Peptide sequence surrounding Lys8 derived from human histone H4 .

  • Reactivity: Primarily human, with cross-reactivity reported in mouse and rat in some studies .

  • Clonality: Polyclonal, ensuring broad epitope recognition .

PropertyDetailSources
HostRabbit
ConjugationUnconjugated (for flexibility in assays)
Storage-20°C in 50% glycerol/PBS buffer

Applications and Validation

The antibody is validated for diverse techniques critical to epigenetic and chromatin studies:

Table 1: Applications and Recommended Dilutions

ApplicationDilution RangeKey ObservationsSources
Western Blot (WB)1:50–1:5000Detects H4K8ac in whole cell lysates (e.g., HeLa, K562) .
Immunohistochemistry (IHC)1:10–1:200Stains nuclear H4K8ac in human glioma and placenta tissues .
Immunofluorescence (IF)1:1–1:10Visualizes nuclear acetylation patterns in HeLa cells .
Chromatin Immunoprecipitation (ChIP)5 µg–2 µg/IPEnriches active gene promoters (e.g., EIF4A2) in K562 cells .

Validation Highlights:

  • ChIP Efficiency: Demonstrated enrichment of DNA from active promoters (e.g., EIF4A2) and depletion from inactive regions (e.g., MYOD1) in K562 cells .

  • Specificity Tests: Dot blot assays confirm no cross-reactivity with unmodified H4 or other acetylated lysines (e.g., H4K5, H4K12) .

Cross-Reactivity and Limitations

While highly specific to H4K8ac, potential caveats include:

  • Limited Cross-Species Reactivity: Primarily validated in human and rodent models; no data for non-vertebrate species .

  • Off-Target Risks: Minimal cross-reactivity with other histone modifications (e.g., H2AK5ac) noted in dot blot experiments .

Research Findings and Use Cases

The antibody has been instrumental in studying epigenetic mechanisms:

Table 2: Notable Research Applications

Study FocusMethodOutcomeSources
Chromatin AccessibilityChIP-seqIdentified H4K8ac enrichment at active gene promoters in leukemia cells (K562) .
Cancer EpigeneticsIHCDetected H4K8ac in glioma and placental tissues, linking acetylation to tumor biology .
Gene RegulationIFVisualized nuclear acetylation dynamics in HeLa cells during chromatin remodeling .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
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 nucleosomes. Nucleosomes are structures that wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones therefore play a crucial role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
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 modifications of histones, including 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; SETD2, LEDGF, and KAT5 are essential for these epigenetic changes. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Data indicates 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 associated with 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 prevents chromatin compaction by inhibiting long-range internucleosomal interactions. PMID: 25294883
  7. Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  8. An increase in histone H4 acetylation caused by hypoxia in human neuroblastoma cell lines corresponds to increased levels of N-myc transcription factor in these cells. PMID: 24481548
  9. Data suggest that G1-phase histone assembly is restricted 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 whole 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 due to the epigenetic mechanism by suppression of acetylation of histone H4. PMID: 21973049
  13. Our findings suggest that global histone H3 and H4 modification patterns are potential markers of 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 increasing the binding affinity of HIRA to H3.3-H4 and reducing 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. PMID: 20949922
  17. Our findings reveal the molecular mechanisms whereby the DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200 which, in turn, reduces gene expression by half. PMID: 20512922
  18. 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 alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation level of histone H4 protein are closely related. PMID: 15095300
  21. Peptidylarginine deiminase 4 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. 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. Relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. H4 tail and its acetylation have novel roles in mediating recruitment of multiple regulatory factors that can change 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 important 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. Our findings indicate an important role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results indicate, by acetylation of histone H4 K16 during S-phase, early replicating chromatin domains acquire the H4K16ac-K20me2 epigenetic label that persists on the chromatin throughout mitosis & 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 relative 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 suggested 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 showed 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 what epitope does the (Ab-8) antibody recognize?

HIST1H4A refers to Histone H4, one of the core histone proteins that, together with H2A, H2B, and H3, form the nucleosome core particle around which DNA wraps. The HIST1H4A (Ab-8) antibody specifically recognizes and binds to the peptide sequence surrounding the lysine 8 (Lys8) residue of human histone H4 . This site is a critical target for post-translational modifications, particularly acetylation, which plays important roles in chromatin structure and gene expression regulation.

The specificity of this antibody stems from its development using a peptide immunogen derived from the region around Lys8 of human histone H4 . As a polyclonal antibody raised in rabbits, it contains a mixture of immunoglobulins that recognize different epitopes of the target protein, providing robust detection capabilities. The antibody has been affinity-purified to enhance its specificity toward the target epitope .

What experimental applications is the HIST1H4A (Ab-8) antibody validated for?

The HIST1H4A (Ab-8) antibody has been validated for multiple experimental applications commonly used in epigenetic and chromatin research. According to the product specifications, this antibody can be successfully employed in:

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of histone H4 in solution .

  • Western Blotting (WB): For detecting histone H4 in protein extracts separated by gel electrophoresis .

  • Immunohistochemistry (IHC): For visualizing histone H4 in tissue sections .

  • Immunofluorescence (IF): For cellular localization studies of histone H4 .

  • Chromatin Immunoprecipitation (ChIP): For isolating genomic regions associated with histone H4, particularly useful for studying histone modifications in the context of gene regulation .

The antibody's versatility across multiple applications makes it a valuable tool for comprehensive studies of histone H4 biology and its modifications. Researchers should optimize conditions for each application through titration experiments to determine the optimal antibody concentration for their specific experimental system.

How should the HIST1H4A (Ab-8) antibody be stored and handled to maintain activity?

Proper storage and handling of the HIST1H4A (Ab-8) antibody are crucial for maintaining its activity and specificity. Based on standard protocols for antibody handling and the provided information:

The antibody should be stored at -20°C for long-term preservation . For routine use, small aliquots can be prepared to avoid repeated freeze-thaw cycles, which can degrade antibody quality. When working with the antibody, it should be kept on ice or at 4°C to minimize degradation.

The concentration of the antibody is lot-specific, and researchers should refer to the datasheet included with the product for the exact concentration information . This information is critical for calculating the appropriate dilution for different applications. Generally, antibody dilutions should be prepared in appropriate buffers containing protein stabilizers (such as BSA) and preservatives to prevent microbial growth during storage.

When preparing working dilutions, use clean, nuclease-free tubes and high-quality water or buffer. Document the date of preparation, dilution factor, and lot number for experimental reproducibility and troubleshooting purposes.

How can HIST1H4A (Ab-8) antibody be optimized for chromatin immunoprecipitation (ChIP) experiments?

Chromatin immunoprecipitation is a powerful technique for studying histone modifications and their association with specific genomic regions. When using the HIST1H4A (Ab-8) antibody for ChIP experiments, several optimization steps are crucial:

First, crosslinking conditions must be carefully optimized. For histone H4 modifications, standard formaldehyde crosslinking (1% for 10 minutes at room temperature) is typically sufficient, but this may require adjustment depending on the specific cell type and experimental question . Overfixation can mask epitopes and reduce antibody binding efficiency, while insufficient fixation may lead to poor chromatin recovery.

For ChIP-seq applications, antibody specificity and efficiency are critical factors. Research has demonstrated that histone H4 acetylation can be effectively detected around DNA double-strand breaks (DSBs) using ChIP methodologies, with acetylation peaks detected approximately 0.6 kb from nuclease-induced DSBs in yeast and up to 1.5 kb in mammalian cells . When analyzing ChIP-seq data, the MACS algorithm has been successfully employed to identify significant histone H4 acetylation peaks, and tools like EdgeR can quantify differential acetylation between experimental conditions .

Input normalization and appropriate controls are essential for accurate interpretation of results. Include both negative controls (IgG or no-antibody controls) and positive controls (regions known to be enriched for the modification) in each experiment. Consider using spike-in normalization methods for quantitative comparisons between different conditions or treatments.

What are the considerations for using HIST1H4A (Ab-8) antibody in studying histone acetylation dynamics?

Histone H4 acetylation is a dynamic process involving both histone acetyltransferases (HATs) and histone deacetylases (HDACs). When studying these dynamics with the HIST1H4A (Ab-8) antibody, several factors should be considered:

Temporal resolution is critical when studying histone modification dynamics. Research has shown that histone H4 acetylation levels change rapidly in response to cellular signals and environmental conditions . Time-course experiments should be designed with appropriate sampling intervals to capture these dynamic changes. For instance, studies have detected peaks in H3 and H4 N-terminal tail acetylation two hours after DNA damage induction .

When investigating acetylation dynamics, experimental designs should consider the roles of both HATs and HDACs. Studies have demonstrated that both acetylation and deacetylation of H4 N-tail lysines are necessary for preventing CAG expansions, suggesting that dynamic turnover, rather than a static modification state, is biologically important . HDAC inhibitors like sodium butyrate (NaBu) and Trichostatin A (TSA) can be valuable tools for manipulating histone acetylation levels in experimental systems .

To comprehensively analyze acetylation dynamics, complementary techniques should be employed alongside antibody-based detection. Mass spectrometry can provide site-specific quantification of multiple modifications simultaneously, while genetic approaches using HAT or HDAC mutants can reveal functional relationships between enzymes and their histone substrates .

How does HIST1H4A (Ab-8) antibody perform in context-specific histone modification research?

The context-dependent nature of histone modifications requires careful experimental design when using the HIST1H4A (Ab-8) antibody in different biological systems:

In neuroscience research, histone H4 acetylation has been linked to behavioral variability. Studies have analyzed acetylation levels across eight different brain regions using ChIP methodologies with histone H4 antibodies, revealing region-specific patterns of acetylation . When designing such experiments, it's essential to carefully dissect and process different brain regions to minimize cross-contamination.

For cellular model systems, like the murine embryonic stem cells (mESCs) used to study chromatin-related diseases such as Rahman syndrome, specific considerations apply . In pluripotent stem cells, chromatin is generally more accessible, which may affect antibody binding efficiency. When using CRISPR-engineered cell lines expressing modified histones, researchers should validate antibody specificity against the engineered constructs, especially if tags have been added to the histone proteins .

In disease-related research, such as studies on HIV-1 integration that involves histone H4 tail interactions with viral integrase, careful consideration of experimental conditions is necessary . When studying histone modifications in the context of viral infections or other pathologies, researchers should account for potential changes in chromatin accessibility, nucleosome positioning, and the presence of competing binding proteins that may affect antibody performance.

What validation approaches should be employed to confirm HIST1H4A (Ab-8) antibody specificity?

Antibody validation is crucial for ensuring experimental reliability, particularly for histone antibodies where cross-reactivity with similar epitopes is a concern:

Peptide competition assays represent a gold standard for validating antibody specificity. In this approach, the antibody is pre-incubated with excess immunizing peptide (the peptide sequence around Lys8 of histone H4) before application to the experimental sample. If the antibody is specific, this pre-incubation should block binding and eliminate the signal .

Genetic validation using knockout or knockdown systems provides compelling evidence for antibody specificity. For histone H4, complete knockout is typically lethal, but conditional systems or targeted mutations at the Lys8 residue can be useful. Additionally, comparing signals in wild-type cells versus cells expressing mutant histones (e.g., K8R mutations that prevent acetylation) can validate modification-specific antibodies .

Orthogonal detection methods should be employed to corroborate findings. Mass spectrometry analysis of immunoprecipitated material can confirm the presence of the target histone and modification. Similarly, comparing results obtained with antibodies from different sources that recognize the same epitope can increase confidence in the specificity of detection .

How can the HIST1H4A (Ab-8) antibody be used in combination with other techniques to study chromatin dynamics?

Integrating multiple technical approaches with antibody-based detection provides more comprehensive insights into chromatin biology:

For studying protein-protein interactions involving histone H4, the antibody can be used in co-immunoprecipitation experiments followed by mass spectrometry or western blotting to identify interaction partners. This approach has been valuable in identifying proteins that interact with acetylated histones, such as bromodomain-containing proteins that recognize acetylated lysine residues .

Combining ChIP with sequencing technologies (ChIP-seq) enables genome-wide mapping of histone H4 occupancy and modifications. This approach has been successfully used to identify genomic regions with differential histone H4 acetylation in response to treatments with HDAC inhibitors like sodium butyrate . For higher resolution, techniques like CUT&RUN or CUT&Tag, which don't require crosslinking, may provide complementary information.

Live-cell imaging approaches using fluorescently-tagged histone readers can complement antibody-based detection of histone modifications. While the HIST1H4A (Ab-8) antibody is primarily used for fixed samples, researchers can correlate antibody staining patterns with live-cell dynamics by using reader domains specific for acetylated lysines fused to fluorescent proteins .

What are common sources of variability in HIST1H4A (Ab-8) antibody experiments?

Several factors can introduce variability in experiments using the HIST1H4A (Ab-8) antibody:

Sample preparation methods significantly impact antibody performance, particularly for nuclear proteins like histones. Incomplete cell lysis, inadequate nuclear extraction, or inappropriate fixation can limit antibody access to epitopes. For histone modifications, excessive formaldehyde crosslinking may mask epitopes, while insufficient fixation may result in epitope loss during processing . Standardizing sample preparation protocols is essential for reproducible results.

Neighboring modifications on the histone tail can affect antibody binding through epitope masking or altered protein conformation. For example, phosphorylation of nearby residues might influence antibody recognition of acetylated Lys8 . When interpreting results, researchers should consider the potential impact of combinatorial modifications, particularly in experimental conditions that might induce multiple types of histone modifications simultaneously.

How should researchers interpret contradictory results from HIST1H4A (Ab-8) antibody experiments?

When faced with contradictory results, systematic troubleshooting and critical analysis are necessary:

First, evaluate technical factors that might contribute to discrepancies. For western blotting, inconsistent results might stem from differences in protein extraction methods, gel running conditions, or transfer efficiency. For immunofluorescence or immunohistochemistry, fixation methods, antigen retrieval procedures, and detection systems can significantly influence outcomes . Standardizing these technical parameters is essential for resolving contradictions.

Biological context must be carefully considered when interpreting seemingly contradictory results. Research has shown that histone H4 acetylation is dynamically regulated and context-dependent, with levels varying across different cell types, tissues, and physiological states . What appears contradictory might reflect genuine biological differences between experimental systems or conditions.

Orthogonal approaches should be employed to resolve contradictions. If ChIP-seq and immunofluorescence results appear inconsistent, for example, complementary techniques like mass spectrometry can provide independent verification. Additionally, genetic approaches, such as manipulating the relevant histone acetyltransferases or deacetylases, can help clarify the specificity and biological relevance of antibody-detected signals .

What statistical approaches are recommended for analyzing data from HIST1H4A (Ab-8) antibody experiments?

For ChIP-seq data analysis, specialized statistical frameworks have been successfully applied to histone H4 acetylation studies. The MACS (Model-based Analysis of ChIP-Seq) algorithm is commonly used for peak calling, while tools like EdgeR can identify statistically significant differences in acetylation levels between experimental conditions . When comparing acetylation levels between groups, appropriate normalization methods should be applied to account for technical variations in sequencing depth and IP efficiency.

When analyzing immunofluorescence or immunohistochemistry data, quantitative image analysis with appropriate controls is essential. Researchers should establish objective criteria for positive staining, collect data from multiple fields of view or tissue sections, and apply appropriate statistical tests based on the data distribution . For experiments comparing treatment groups, power analyses should guide sample size determination to ensure sufficient statistical power.

For western blotting, quantification should include multiple biological replicates with appropriate loading controls. Statistical comparisons should account for the semi-quantitative nature of western blotting, and non-parametric tests may be more appropriate if data do not meet the assumptions of parametric tests . Researchers should also consider the dynamic range limitations of detection methods when interpreting quantitative differences.

How is HIST1H4A (Ab-8) antibody being applied in disease-related epigenetic research?

The HIST1H4A (Ab-8) antibody and similar histone H4 antibodies are increasingly employed in studying disease mechanisms with epigenetic components:

In neurological disorders, histone H4 acetylation patterns have been linked to behavioral phenotypes and neurodegeneration. Research using histone H4 antibodies has revealed that HDAC inhibitors can reduce behavioral inter-individual variability in zebrafish models, suggesting potential therapeutic applications for conditions with behavioral components . When designing such studies, researchers should consider brain region-specific effects and the temporal dynamics of epigenetic modifications.

Cancer epigenetics research frequently utilizes histone modification antibodies to investigate dysregulated gene expression. Altered histone H4 acetylation patterns have been observed in various cancer types, often correlating with disease progression or treatment response . For such applications, careful validation in the specific cancer cell types or tissues is necessary, as the nuclear architecture and chromatin accessibility may differ substantially from normal cells.

Viral infection models, particularly for HIV-1, have benefited from histone H4 research. Studies have demonstrated that the human H4 tail can stimulate HIV-1 integration through binding to the carboxy-terminal domain of viral integrase . This research highlights the potential of histone modification studies in understanding host-pathogen interactions and developing novel therapeutic approaches. When designing such experiments, researchers should consider how viral proteins might compete with or alter normal histone modification patterns.

What are the latest methodological advances in using antibodies like HIST1H4A (Ab-8) for chromatin studies?

Recent technological developments have expanded the utility of histone antibodies in chromatin research:

Single-cell epigenomic techniques now allow researchers to analyze histone modifications at the individual cell level, revealing heterogeneity that would be masked in bulk population studies. Adaptations of ChIP protocols for small cell numbers or even single cells can be combined with the HIST1H4A (Ab-8) antibody to study cell-to-cell variation in histone H4 modifications . These approaches are particularly valuable for heterogeneous tissues or cell populations where subpopulations might exhibit distinct epigenetic states.

Multiplexed detection methods enable simultaneous analysis of multiple histone modifications from the same sample. Techniques like Co-ChIP, sequential ChIP, or mass cytometry combined with histone antibodies allow researchers to investigate combinatorial modification patterns and their functional relationships . When designing such experiments, careful validation of antibody compatibility and potential epitope masking effects is essential.

Genome engineering approaches, such as those used to create cellular models for Rahman syndrome, can be combined with histone antibodies to study the functional consequences of specific histone variants or modifications . CRISPR-based methods for introducing specific histone mutations or for targeting enzymes that modify histones to particular genomic loci represent powerful approaches for investigating causal relationships between histone modifications and biological outcomes.

What are the current limitations of HIST1H4A (Ab-8) antibody research and potential solutions?

Despite its utility, research using the HIST1H4A (Ab-8) antibody faces several challenges:

Cross-reactivity with other histone variants or modifications remains a concern, particularly given the high sequence conservation among histone proteins. While the antibody is designed to be specific for the region around Lys8 of histone H4, comprehensive validation in each experimental system is advisable . Emerging technologies like programmable antibodies or synthetic nanobodies with enhanced specificity may address these limitations in the future.

The semi-quantitative nature of antibody-based detection presents challenges for precise quantification of histone modifications. Integration with quantitative mass spectrometry approaches can provide complementary, absolute quantification of modification levels . Additionally, the development of internal standards and calibration methods for immunoassays may improve quantitative accuracy.

Context-dependent epitope accessibility in different chromatin states may influence antibody binding efficiency. Advanced sample preparation methods that control for chromatin accessibility differences or complementary approaches like native ChIP or CUT&RUN, which don't require crosslinking, may help address these challenges .

What future research directions will benefit from HIST1H4A (Ab-8) antibody applications?

Several emerging research areas are likely to benefit from continued application of histone H4 antibodies:

Single-cell epigenomics represents a frontier for histone modification research. Adapting techniques like CUT&TAG for single-cell applications, combined with the specificity of antibodies like HIST1H4A (Ab-8), will enable unprecedented insights into epigenetic heterogeneity within tissues and cell populations . These approaches will be particularly valuable for understanding developmental processes, disease progression, and treatment responses.

Spatial epigenomics, which aims to map histone modifications in the context of tissue architecture, represents another promising direction. Combining immunohistochemistry using the HIST1H4A (Ab-8) antibody with spatial transcriptomics methods could reveal location-specific epigenetic regulation in complex tissues .

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.