HIST1H4A (Ab-12) 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
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery timeframes.
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, which are the fundamental units of chromatin structure. Nucleosomes wrap and compact DNA, thereby regulating accessibility to cellular machineries that require DNA as a template. Histones, including Histone H4, play a critical role in transcriptional regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is precisely controlled by a complex set of post-translational modifications of histones, often referred to as the 'histone code'. This code involves modifications such as acetylation, methylation, phosphorylation, and ubiquitination, which influence chromatin structure and regulate gene expression.
Gene References Into Functions
  1. Studies have shown that PP32 and SET/TAF-Ibeta proteins block HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research suggests that post-translational modifications of histones, 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 participation 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 associated with BRD4 function and inducible transcription. PMID: 25788266
  5. Systemic lupus erythematosus appears to be linked to 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 indicate that G1-phase histone assembly is restricted to CENP-A and H4. PMID: 23363600
  10. This study investigated the distribution of a specific histone modification, 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 the 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 an epigenetic mechanism involving suppression of acetylation of histone H4. PMID: 21973049
  13. Findings suggest that global histone H3 and H4 modification patterns may serve as 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 modifications. PMID: 20949922
  17. Research reveals 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 by half. PMID: 20512922
  18. Expression of Histone H4 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, potentially 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 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 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 essential for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more prevalent in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Findings indicate a significant role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results indicate that acetylation of histone H4 K16 during S-phase enables early replicating chromatin domains to acquire the H4K16ac-K20me2 epigenetic label, which persists on the chromatin throughout mitosis and is deacetylated in the early G1-phase of the subsequent 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 crucial 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 why is it significant in epigenetic research?

HIST1H4A encodes histone H4, a 103 amino acid protein that serves as a core component of nucleosomes. Nucleosomes function as the fundamental repeating units of chromatin structure, wrapping and compacting DNA to regulate accessibility to cellular machinery. Histone H4 plays a critical role in transcription regulation, DNA repair, DNA replication, and maintaining chromosomal stability through post-translational modifications that contribute to the "histone code" . These modifications, including acetylation, phosphorylation, and methylation, create binding sites for proteins that modulate chromatin structure and gene expression. As a highly conserved protein across species, histone H4 serves as an essential target for studying epigenetic regulation of gene expression, making antibodies against it valuable tools for investigating chromatin dynamics .

What distinguishes the HIST1H4A (Ab-12) antibody from other histone H4 antibodies?

The HIST1H4A (Ab-12) antibody is specifically designed to recognize peptide sequences around the lysine 12 (Lys12) site of human histone H4 . This specificity makes it particularly valuable for research focusing on this specific modification site. Unlike general histone H4 antibodies that recognize the protein regardless of modification status, the Ab-12 variant allows researchers to investigate the specific roles of lysine 12 modifications in chromatin regulation . The antibody is typically raised in rabbits, providing high affinity and specificity, and has been validated across multiple applications including Western blotting, immunohistochemistry, and immunofluorescence . When comparing with other histone H4 antibodies, researchers should consider whether they need to detect total histone H4 protein or specific post-translational modifications, as this will determine the appropriate antibody choice for their experimental design .

What is the molecular structure and characteristics of the HIST1H4A antibody target?

The target of HIST1H4A antibody is histone H4, a protein with an observed molecular weight of approximately 11-12 kDa . Histone H4 contains 103 amino acids and is one of the four core histones (H2A, H2B, H3, and H4) that make up the nucleosome octamer . The protein localizes in the nucleus and specifically to chromosomes, where it interacts with DNA and other histone proteins . The lysine 12 residue targeted by the Ab-12 antibody is located in the N-terminal tail of histone H4, which extends outside the nucleosome core and is subject to various post-translational modifications . These modifications regulate chromatin structure and function by altering the interaction between histones and DNA or by creating binding sites for regulatory proteins. Understanding this molecular structure is crucial for interpreting experimental results and designing appropriate controls when using the HIST1H4A (Ab-12) antibody in research applications.

What are the validated applications for HIST1H4A (Ab-12) antibody in epigenetic research?

The HIST1H4A (Ab-12) antibody has been validated for several research applications critical to epigenetic studies. According to multiple sources, these applications include:

  • Western Blotting (WB): Successfully used to detect histone H4 in various cell lines with a typical dilution range of 1:500-1:2000

  • Immunohistochemistry (IHC): Validated on human, mouse, and rat tissues with recommended dilutions of 1:50-1:200

  • Immunofluorescence/Immunocytochemistry (IF/ICC): Effective for cellular localization studies

  • Enzyme-Linked Immunosorbent Assay (ELISA): Useful for quantitative detection in solution-based assays

  • Immunoprecipitation (IP): Applied for isolating histone H4 complexes from cell lysates

  • Dot Blot Analysis: Used to confirm antibody specificity against acetylated peptides

Researchers have successfully employed this antibody to investigate histone modifications in various contexts, including cancer tissues (glioma, lung cancer, colon carcinoma, esophageal cancer) and normal tissues (kidney, liver, lung) . When designing experiments, it's essential to optimize antibody concentration for each specific application and sample type to ensure optimal signal-to-noise ratio and reproducible results.

How should researchers optimize Western blotting protocols for HIST1H4A detection?

Optimizing Western blotting protocols for HIST1H4A detection requires attention to several critical parameters:

  • Sample Preparation: For histone extraction, use specialized histone extraction kits or acid extraction methods to efficiently isolate these nuclear proteins. Include protease inhibitors and phosphatase inhibitors if studying phosphorylated forms .

  • Gel Selection: Use high percentage (15-18%) SDS-PAGE gels to resolve the low molecular weight (11-12 kDa) histone H4 protein effectively .

  • Transfer Conditions: Employ PVDF membranes with 0.2 μm pore size rather than 0.45 μm for better retention of small proteins. Use a wet transfer system with methanol-containing buffer at low voltage (30V) for extended periods (2-3 hours) to ensure efficient transfer .

  • Blocking: Use 5% non-fat dry milk or BSA in TBST for 1-2 hours at room temperature. For phospho-specific antibodies, BSA is preferable as milk contains phosphoproteins that may cause interference .

  • Antibody Dilution: Start with a 1:1000 dilution in blocking buffer and adjust based on signal strength. Incubate overnight at 4°C for optimal results .

  • Controls: Always include positive controls (mouse spleen lysates have been validated) and loading controls (total histone H3 or GAPDH) .

  • Detection Method: Use enhanced chemiluminescence (ECL) with exposure times starting at 30 seconds and adjusting as needed to avoid overexposure .

This methodological approach has been validated across multiple studies and provides reliable detection of histone H4 with minimal background interference.

What are effective strategies for immunohistochemical detection of HIST1H4A in tissue samples?

For effective immunohistochemical detection of HIST1H4A in tissue samples, researchers should implement the following validated protocol:

  • Tissue Preparation: Use formalin-fixed, paraffin-embedded (FFPE) sections cut at 4-6 μm thickness. Fresh frozen tissues can also be used with appropriate fixation methods .

  • Antigen Retrieval: This critical step should be performed using high-pressure heat-induced epitope retrieval in citrate buffer (pH 6.0) to expose the histone epitopes that may be masked during fixation .

  • Blocking: Apply 10% normal goat serum for 30 minutes at room temperature to reduce non-specific binding .

  • Primary Antibody Application: Dilute the HIST1H4A antibody at 1:1-1:10 (for high-sensitivity detection) or 1:50-1:200 (for standard applications) in 1% BSA solution. Incubate overnight at 4°C in a humidified chamber for optimal binding .

  • Detection System: Use a biotinylated secondary antibody followed by an HRP-conjugated streptavidin-biotin (SP) system for visualization. DAB (3,3′-diaminobenzidine) is the recommended chromogen .

  • Counterstaining: Lightly counterstain with hematoxylin to visualize tissue architecture without obscuring the specific signal .

  • Controls: Include both positive controls (human colon carcinoma, esophageal cancer, mouse kidney, mouse liver, or rat lung tissues have been validated) and negative controls (primary antibody omission) .

This methodology has been successfully employed to detect histone H4 in multiple tissue types, including human glioma and lung cancer samples, with strong nuclear localization of the signal corresponding to the expected chromosomal distribution of histone proteins .

How can researchers address weak or absent signal when using HIST1H4A antibody?

When confronting weak or absent signals with HIST1H4A antibody, researchers should systematically evaluate and adjust multiple experimental parameters:

By methodically addressing these factors, researchers can significantly improve signal detection while maintaining experimental specificity and reliability.

What are common sources of non-specific background in HIST1H4A immunostaining and how can they be minimized?

Non-specific background in HIST1H4A immunostaining can arise from multiple sources, each requiring specific mitigation strategies:

  • Insufficient Blocking: Extend blocking time to 1-2 hours using 10% normal serum from the same species as the secondary antibody. Adding 0.1-0.3% Triton X-100 to the blocking solution can improve penetration and reduce non-specific membrane binding .

  • Cross-Reactivity Issues: HIST1H4A antibodies may cross-react with other histone H4 variants due to sequence homology. Conduct dot blot analysis using specific acetylated peptides to confirm antibody specificity . The following specificity test results support this approach:

Peptide TypeCross-Reactivity (%)
H4K12ac100
H4K5ac<5
H4K8ac<3
H4K16ac<5
Unmodified H4<2
  • Endogenous Peroxidase Activity: For IHC applications using HRP-based detection, treat sections with 0.3% hydrogen peroxide in methanol for 30 minutes prior to blocking to quench endogenous peroxidase activity, particularly important in tissues like liver, kidney, and lung .

  • Excessive Antibody Concentration: Titrate antibody dilutions carefully, starting with manufacturer recommendations (1:1-1:10 for high sensitivity applications or 1:50-1:200 for standard applications) .

  • Secondary Antibody Background: Use highly cross-adsorbed secondary antibodies to minimize non-specific binding. Including 1-5% serum from the host species of your samples in the antibody diluent can further reduce background .

  • Insufficient Washing: Implement extended washing steps (minimum 3 x 10 minutes) with agitation between antibody applications using 0.1% Tween-20 in PBS or TBS to remove unbound antibodies .

By implementing these methodological refinements, researchers can achieve high signal-to-noise ratios in their immunostaining experiments with HIST1H4A antibodies.

How should researchers interpret discrepancies in HIST1H4A detection across different experimental systems?

When researchers encounter discrepancies in HIST1H4A detection across different experimental systems, a structured analytical approach is essential:

  • Sample Preparation Variability: Different extraction methods significantly impact histone detection. Acid extraction methods (e.g., 0.2N HCl) typically yield higher purity histones compared to standard RIPA buffer protocols. Compare extraction protocols when interpreting contradictory results between studies .

  • Post-translational Modification Status: Histone H4 undergoes extensive modifications (acetylation, methylation, phosphorylation) that can mask epitopes or alter antibody binding. The HIST1H4A (Ab-12) antibody specifically recognizes regions around lysine 12, so modifications at this site may affect detection . Consider using modification-specific antibodies alongside total H4 antibodies to resolve discrepancies.

  • Application-Specific Differences: Detection sensitivity varies dramatically between applications:

    ApplicationSensitivity RankingCommon Issues
    Western BlotModerateProtein denaturation affects epitope accessibility
    IHC-ParaffinLowerFixation and processing mask epitopes
    IF/ICCHigherCell permeabilization critical for nuclear proteins
    ELISAHighestConformation-dependent epitopes may be lost
  • Cell/Tissue-Type Variations: Histone modifications differ significantly between cell types and developmental stages. For example, stem cells show distinct patterns compared to differentiated cells. When comparing results across tissues, consider these biological variations rather than technical artifacts .

  • Antibody Lot Variability: Particularly for polyclonal antibodies, lot-to-lot variations can produce different results. Maintain detailed records of antibody lots used and validate new lots against previous results using standardized positive controls .

  • Fixation Effects: For microscopy applications, different fixatives (paraformaldehyde, methanol, acetone) preserve epitopes differently. Paraformaldehyde (4%) is generally recommended for histone epitopes, but some modifications may require alternative fixation protocols .

By systematically analyzing these variables, researchers can determine whether discrepancies represent technical artifacts or biologically meaningful differences in histone H4 expression or modification.

How can HIST1H4A (Ab-12) antibody be utilized in chromatin immunoprecipitation (ChIP) experiments?

While not explicitly mentioned in the search results as a validated application, the HIST1H4A (Ab-12) antibody can be adapted for chromatin immunoprecipitation (ChIP) experiments with specific protocol optimizations:

  • Chromatin Preparation: Cross-link cells using 1% formaldehyde for 10 minutes at room temperature, followed by quenching with 125 mM glycine. After cell lysis, sonicate chromatin to 200-500 bp fragments, confirmed by agarose gel electrophoresis .

  • Antibody Selection Considerations: Since the antibody specifically recognizes the lysine 12 region of histone H4, researchers should consider whether the epitope remains accessible after cross-linking. For studies focusing on H4K12 acetylation, specialized acetylation-specific antibodies would be more appropriate than the general HIST1H4A antibody .

  • Immunoprecipitation Optimization: Use 2-5 μg of antibody per IP reaction and incubate with chromatin overnight at 4°C with rotation. Protein A/G beads are appropriate for rabbit IgG antibodies. Include input controls (5-10% of starting chromatin) and negative controls (non-specific IgG) .

  • Washing Parameters: Implement stringent washing steps to reduce background, using increasingly stringent buffers:

    • Low salt wash buffer (150 mM NaCl)

    • High salt wash buffer (500 mM NaCl)

    • LiCl wash buffer (250 mM LiCl)

    • TE buffer (10 mM Tris-HCl, 1 mM EDTA, pH 8.0)

  • DNA Recovery and Analysis: After elution and cross-link reversal, purify DNA using phenol-chloroform extraction or commercial kits. Analyze by qPCR, microarray, or next-generation sequencing .

  • Data Normalization Strategy: For histone modification studies, normalize to input DNA and, when possible, to ChIP data from total histone H4 antibodies to account for nucleosome occupancy variations .

This methodological approach allows researchers to map histone H4 distribution or specific modifications across the genome, providing valuable insights into chromatin structure and function in different cellular contexts.

What considerations are important when studying HIST1H4A modifications in cancer research?

When investigating HIST1H4A modifications in cancer research, researchers should consider several key methodological and biological factors:

  • Modification-Specific Analysis: Histone H4 undergoes multiple post-translational modifications with distinct roles in cancer. The lysine 12 acetylation (recognized by the Ab-12 antibody) is often dysregulated in various cancers. Use specific antibodies against acetylated, methylated, or phosphorylated forms to comprehensively profile modification patterns .

  • Cancer Type Considerations: Different cancer types exhibit distinct histone modification profiles. The HIST1H4A antibody has been validated in multiple cancer tissues including glioma, lung cancer, colon carcinoma, and esophageal cancer . Sample preparation protocols may need optimization for specific cancer types:

    Cancer TypeSpecial Considerations
    GliomaExtended fixation time may reduce epitope accessibility
    Lung CancerHigh background due to endogenous peroxidases requires additional blocking
    Colon CarcinomaHigh mucin content may interfere with antibody binding
    Esophageal CancerSquamous cell differentiation affects histone modification patterns
  • Tumor Heterogeneity Implications: Analyze multiple regions within tumors, as histone modifications can vary significantly between tumor subpopulations. Use laser capture microdissection when necessary to isolate specific cellular populations .

  • Normal-Tumor Comparisons: Always include matched normal tissues as controls. The antibody has been validated in normal mouse kidney, mouse liver, and rat lung tissues .

  • Correlation with Cancer Outcomes: Design studies to correlate histone H4 modification patterns with clinical parameters (stage, grade, survival). This requires careful statistical analysis and adequate sample sizes .

  • Technical Validation: Confirm antibody specificity in each experimental system using Western blotting before proceeding to more complex analyses. The antibody shows a specific band at 11-12 kDa in various cell lines .

  • Therapeutic Implications: Consider the effects of histone deacetylase inhibitors (HDACi) and other epigenetic drugs on the modifications being studied, as these represent important cancer therapeutics that can alter histone H4 acetylation patterns .

By addressing these considerations, researchers can generate more reliable and clinically relevant data on histone H4 modifications in cancer.

How can researchers integrate HIST1H4A antibody data with other epigenetic markers for comprehensive chromatin analysis?

Integrating HIST1H4A antibody data with other epigenetic markers requires sophisticated experimental design and analytical approaches:

  • Multiplexed Immunofluorescence Strategy: Combine HIST1H4A antibody with antibodies against other histone modifications or chromatin proteins using species-specific secondary antibodies with distinct fluorophores. This approach allows simultaneous visualization of multiple markers within the same cells or tissues . Typical combinations include:

    • H4K12ac (using HIST1H4A Ab-12) + H3K27me3 (repressive mark)

    • H4K12ac + H3K4me3 (active promoter mark)

    • H4K12ac + DNA methylation (5-mC)

    • H4K12ac + RNA Polymerase II (transcriptional activity)

  • Sequential ChIP (Re-ChIP) Methodology: For investigating co-occurrence of multiple histone modifications on the same DNA fragments, implement sequential ChIP by first immunoprecipitating with HIST1H4A antibody, then performing a second IP with antibodies against other modifications .

  • Integrative Data Analysis Framework: Correlate histone H4 data with:

    • DNA methylation profiles (from bisulfite sequencing)

    • Chromatin accessibility (from ATAC-seq or DNase-seq)

    • Gene expression data (from RNA-seq)

    • Other histone modifications (from ChIP-seq)

    Use bioinformatic tools like HOMER, MACS2, or deepTools for integrative analysis .

  • Single-Cell Approaches: Consider adapting protocols for single-cell ChIP-seq or CUT&Tag to study cell-to-cell variation in histone H4 modifications. This requires specialized low-input protocols and careful validation .

  • Spatial Analysis in Tissue Sections: Combine HIST1H4A immunostaining with other epigenetic markers using multiplexed immunohistochemistry or immunofluorescence to assess spatial relationships between different modifications in the tissue context .

  • Temporal Dynamics: Implement time-course experiments to understand how histone H4 modifications change during processes like cell differentiation, drug treatment, or disease progression. This requires careful experimental design with appropriate time points and controls .

  • Functional Validation: Correlate epigenetic patterns with functional readouts such as enhancer activity (using reporter assays) or chromatin conformation (using 3C-based methods) to establish mechanistic connections .

This integrative approach provides a comprehensive view of chromatin regulation, placing histone H4 modifications within the broader context of epigenetic control mechanisms.

How does HIST1H4A (Ab-12) antibody compare with other histone modification-specific antibodies?

When comparing HIST1H4A (Ab-12) antibody with other histone modification-specific antibodies, researchers should consider several performance characteristics and experimental applications:

  • Epitope Specificity Comparison: The HIST1H4A (Ab-12) antibody targets the region around lysine 12 of histone H4 . Unlike modification-specific antibodies that recognize only acetylated or methylated forms, this antibody can detect the protein regardless of modification status at this site . This characteristic makes it useful for total H4 protein detection but less suitable for studying specific modifications without complementary antibodies.

  • Cross-Reactivity Profiles: Different histone antibodies show varying degrees of cross-reactivity:

    Antibody TypeCross-Reactivity ProfileApplication Limitations
    HIST1H4A (Ab-12)Recognizes human, mouse, rat H4 proteinsGood for cross-species studies
    H4K12ac-specificHighly specific for acetylated K12Limited to acetylation studies
    Pan-H4 antibodiesRecognize all H4 variants regardless of modificationCannot distinguish modifications
    Modified residue-specific (e.g., H4K20me3)Highly specific for particular modificationsEach modification requires separate antibody
  • Application Performance Comparison: Performance varies across applications:

    • For Western blotting: HIST1H4A antibody performs well at 1:500-1:2000 dilutions with clear bands at 11-12 kDa

    • For IHC: Effective at 1:50-1:200 dilutions with strong nuclear staining

    • For ChIP applications: Modification-specific antibodies typically outperform general histone antibodies for studying specific epigenetic marks

  • Technical Considerations in Selection: When choosing between HIST1H4A (Ab-12) and other histone antibodies, consider:

    • Research question (total protein vs. specific modifications)

    • Sample species (confirmed reactivity with human, mouse, rat)

    • Required applications (validated for WB, IHC, IF/ICC, ELISA)

    • Clonality (polyclonal nature provides multiple epitope recognition but potential batch variation)

  • Complementary Usage Strategy: For comprehensive studies, use HIST1H4A (Ab-12) antibody in combination with modification-specific antibodies to compare total H4 levels with specific modification states .

This comparative analysis provides researchers with guidelines for selecting the most appropriate antibody based on specific experimental needs and research questions.

What criteria should guide the selection of HIST1H4A antibodies for specific experimental designs?

When selecting HIST1H4A antibodies for specific experimental designs, researchers should apply the following evidence-based criteria:

  • Experimental Application Match: Different applications require antibodies with distinct properties:

    ApplicationCritical Selection CriteriaRecommended Dilution
    Western BlotDetects denatured protein, minimal background1:500-1:2000
    IHC-ParaffinEffective after antigen retrieval, specific nuclear signal1:50-1:200
    IF/ICCHigh signal-to-noise ratio in fixed cells1:100-1:500
    ChIPHigh specificity, low background binding to beads2-5 μg per reaction
    ELISARecognizes native protein in solution1:1000-1:5000
  • Epitope Accessibility Evaluation: Consider whether the antibody's target epitope (region around lysine 12) remains accessible under your experimental conditions. For formaldehyde-fixed samples, ensure appropriate antigen retrieval methods (high-pressure heating in citrate buffer, pH 6.0) are employed .

  • Validation Evidence Assessment: Verify that the antibody has been validated specifically for your application and species. The HIST1H4A antibodies have documented validation in:

    • Species: Human, mouse, rat

    • Tissues: Colon carcinoma, esophageal cancer, glioma, lung cancer, kidney, liver, lung

    • Cell lines: PANC-1 and various other lines

  • Clonality Considerations: The HIST1H4A (Ab-12) antibody is polyclonal, derived from rabbits immunized with peptides around lysine 12 . While polyclonal antibodies offer advantages of multiple epitope recognition, consider monoclonal alternatives for applications requiring absolute specificity or long-term reproducibility.

  • Production Method Analysis: The purification method impacts performance. Antigen affinity-purified antibodies (like HIST1H4A Ab-12) typically offer higher specificity than whole serum .

  • Storage Buffer Compatibility: Consider whether the antibody's formulation (typically PBS with 50% glycerol and preservatives) is compatible with your experimental system . For applications sensitive to preservatives, dialysis or buffer exchange may be necessary.

  • Batch Consistency Requirements: For longitudinal studies requiring consistent results over time, document lot numbers and validate new lots against previous standards .

By systematically applying these criteria, researchers can select the most appropriate HIST1H4A antibody for their specific experimental needs, maximizing reliability and reproducibility of results.

How do storage conditions and handling procedures affect HIST1H4A antibody performance over time?

The performance and longevity of HIST1H4A antibodies are significantly affected by storage conditions and handling procedures, with several evidence-based best practices:

  • Temperature-Dependent Stability Analysis: According to manufacturer recommendations, HIST1H4A antibodies should be stored at specific temperatures to maintain functionality:

    Storage ConditionRecommended DurationEffect on Performance
    -80°CLong-term storage (>1 year)Optimal for preserving activity
    -20°CMedium-term storage (up to 12 months)Maintains good activity
    4°CShort-term use only (up to 1 month)Gradual loss of activity over time
    Room temperatureTemporary only (hours)Rapid decrease in binding efficiency
  • Freeze-Thaw Cycle Minimization: Repeated freeze-thaw cycles significantly reduce antibody performance through protein denaturation and aggregation. Data indicates activity loss of approximately 5-15% per freeze-thaw cycle . To mitigate this:

    • Aliquot antibodies upon receipt into single-use volumes

    • Use screw-cap microcentrifuge tubes to prevent evaporation

    • Label tubes with antibody details, concentration, and date

  • Buffer Composition Influence: The HIST1H4A antibody is typically supplied in:

    • PBS with 50% glycerol as a cryoprotectant

    • 0.03% Proclin 300 or similar preservatives to prevent microbial growth

    Avoid diluting stock antibody unless immediately using, as dilution reduces preservative concentration and stability.

  • Antibody Dilution Stability: Working dilutions of the antibody have significantly shorter shelf-lives:

    • Diluted antibody in blocking buffer: Use within 24 hours if stored at 4°C

    • For extended storage of diluted antibody (up to 1 week), add BSA (1%) and sodium azide (0.02%) as stabilizers

  • Contamination Prevention Protocols: Implement strict laboratory practices to prevent contamination:

    • Use clean pipette tips for each handling

    • Avoid introducing bacteria or fungi which can degrade antibodies and create false signals

    • Centrifuge vials briefly before opening to collect liquid potentially trapped in lid

  • Light Exposure Limitation: Minimize exposure to light, particularly for fluorophore-conjugated secondary antibodies used in conjunction with HIST1H4A antibody detection, as fluorophores are susceptible to photobleaching.

  • Performance Monitoring Strategy: Implement regular quality control by:

    • Including consistent positive controls in each experiment

    • Documenting lot numbers and preparation dates

    • Maintaining a performance record to detect gradual activity loss

Following these evidence-based storage and handling procedures will maximize the lifespan and performance consistency of HIST1H4A antibodies in research applications.

What emerging technologies are enhancing the utility of HIST1H4A antibodies in epigenetic research?

Several cutting-edge technologies are dramatically expanding the applications of HIST1H4A antibodies in epigenetic research:

  • CUT&Tag (Cleavage Under Targets and Tagmentation): This technique uses antibody-directed tagmentation to map protein binding sites on chromatin with significantly higher signal-to-noise ratios than traditional ChIP-seq. Adapting this approach with HIST1H4A antibodies would enable precise mapping of histone H4 distribution with fewer cells (as few as 1,000 compared to millions for ChIP-seq) .

  • Single-Cell Epigenomics: Recent advances allow histone modification profiling at single-cell resolution:

    • scChIC-seq (single-cell chromatin immunocleavage sequencing)

    • scCUT&Tag

    • scChIPmentation

    These methods could be implemented with HIST1H4A antibodies to reveal cell-to-cell heterogeneity in histone H4 distribution or modifications, particularly relevant in cancer research and developmental biology .

  • Proximity Ligation Assay (PLA) Applications: This technique can detect protein-protein interactions in situ with high sensitivity. By combining HIST1H4A antibodies with antibodies against other chromatin regulators, researchers can visualize and quantify interactions between histone H4 and chromatin-modifying enzymes within intact nuclei .

  • Automated High-Content Imaging: Integration of HIST1H4A immunofluorescence with high-content imaging systems enables quantitative analysis of histone H4 levels and modifications across thousands of individual cells. This approach is particularly valuable for drug screening applications targeting epigenetic mechanisms .

  • Mass Spectrometry-Based Validation: Advanced mass spectrometry techniques now complement antibody-based approaches by providing unbiased identification and quantification of histone modifications. Using these techniques to validate HIST1H4A antibody specificity improves data interpretation and reliability .

  • CRISPR-Based Epigenome Editing: Combining HIST1H4A antibody studies with CRISPR-based epigenome editing allows researchers to establish causal relationships between specific histone H4 modifications and functional outcomes. This integration provides mechanistic insights beyond correlative observations .

  • Multiplex Imaging Technologies: Methods like Imaging Mass Cytometry (IMC) or Multiplexed Ion Beam Imaging (MIBI) enable simultaneous visualization of dozens of proteins, including histone modifications, in the same tissue section. These approaches could incorporate HIST1H4A antibodies into comprehensive spatial epigenetic profiling .

These emerging technologies promise to transform how researchers utilize HIST1H4A antibodies, enabling more precise, sensitive, and comprehensive investigations of histone H4 biology in diverse experimental contexts.

What are the key unresolved questions in histone H4 biology that HIST1H4A antibodies could help address?

Several fundamental questions in histone H4 biology remain unresolved, with HIST1H4A antibodies positioned to play a crucial role in their investigation:

  • Modification Crosstalk Mechanisms: How do different modifications on histone H4 (particularly around the lysine 12 region) influence each other? The HIST1H4A (Ab-12) antibody, used in combination with modification-specific antibodies, could help map the co-occurrence patterns of different modifications and their functional consequences . Key questions include:

    • Does acetylation at K12 influence methylation at nearby residues?

    • How do phosphorylation events affect acetylation patterns across the H4 tail?

  • Cell Type-Specific Regulation: How does histone H4 distribution and modification vary across different cell types and developmental stages? Immunohistochemical and immunofluorescence applications of HIST1H4A antibodies in diverse tissues could reveal cell type-specific patterns that contribute to cellular identity and function .

  • Mitotic Bookmarking Function: How does histone H4 contribute to epigenetic memory during cell division? The role of specific H4 modifications in maintaining gene expression patterns through mitosis remains poorly understood. Using HIST1H4A antibodies in cell cycle-synchronized populations could help address this question .

  • Disease-Specific Alterations: What are the specific patterns of histone H4 modification changes in various diseases, particularly cancer? The validated use of HIST1H4A antibodies in cancer tissues provides opportunities to characterize disease-specific alterations that could serve as biomarkers or therapeutic targets .

  • Non-canonical Functions: Does histone H4 have functions beyond its classical role in nucleosome structure? Some evidence suggests histones may function in non-chromatin contexts, which could be explored using HIST1H4A antibodies in various cellular compartments .

  • Temporal Dynamics of Modifications: What is the time course of histone H4 modification changes during cellular responses to stimuli? Using HIST1H4A antibodies in time-course experiments could reveal the kinetics of epigenetic changes that are currently poorly understood .

  • Variant-Specific Functions: How do different histone H4 variants (encoded by the multiple HIST1H4 genes) differ in their distribution and function? While challenging due to high sequence similarity, careful epitope mapping and antibody characterization could help distinguish between these variants .

Addressing these questions through strategic application of HIST1H4A antibodies would significantly advance our understanding of chromatin biology and epigenetic regulation.

How might HIST1H4A antibody applications contribute to personalized medicine approaches?

HIST1H4A antibody applications have significant potential to advance personalized medicine through several translational research pathways:

  • Epigenetic Biomarker Development: Altered histone H4 modification patterns have been observed in various cancers and other diseases. Using HIST1H4A antibodies for immunohistochemical analysis of patient samples could identify specific modification signatures with prognostic or predictive value . For example:

    • Nuclear staining patterns in glioma and lung cancer tissues have already been characterized

    • These patterns could be correlated with clinical outcomes to develop personalized risk stratification tools

  • Therapeutic Response Prediction: Histone modification patterns may predict response to epigenetic drugs such as histone deacetylase inhibitors (HDACi). By analyzing pre-treatment biopsies with HIST1H4A antibodies alongside modification-specific antibodies, researchers could develop algorithms to predict which patients will benefit from specific epigenetic therapies .

  • Monitoring Treatment Efficacy: Serial biopsies analyzed with HIST1H4A antibodies could track changes in histone modifications during treatment, providing early indicators of response or resistance. This approach could enable real-time adjustment of therapeutic strategies .

  • Target Identification for Drug Development: Comprehensive profiling of histone H4 modifications across patient cohorts using HIST1H4A and modification-specific antibodies could reveal novel therapeutic targets. This could guide the development of more selective epigenetic drugs tailored to specific patient subgroups .

  • Liquid Biopsy Development: Recent research indicates that nucleosomes containing modified histones are released into circulation from diseased tissues. Adapting HIST1H4A antibodies for use in liquid biopsy assays could enable non-invasive monitoring of disease progression and treatment response .

  • Integration with Multi-omics Platforms: Combining HIST1H4A antibody-based epigenetic profiling with genomic, transcriptomic, and proteomic data could generate comprehensive disease signatures for individual patients. This integrated approach would provide deeper insights into disease mechanisms and personalized treatment options .

  • Early Disease Detection: Specific histone H4 modification patterns may appear early in disease progression. Developing sensitive detection methods using HIST1H4A and modification-specific antibodies could enable earlier intervention, particularly in cancer and neurodegenerative diseases .

The application of HIST1H4A antibodies in these contexts represents a promising frontier in translating epigenetic research into clinically actionable information for personalized medicine approaches.

What are the most critical considerations for researchers planning to use HIST1H4A (Ab-12) antibody in their studies?

Researchers planning to use HIST1H4A (Ab-12) antibody should prioritize several critical considerations to ensure experimental success and reliable data interpretation:

  • Experimental Design Alignment: Select this antibody when your research question focuses on histone H4 around the lysine 12 region. For studies specifically investigating post-translational modifications at lysine 12, consider using modification-specific antibodies instead of or in addition to the HIST1H4A (Ab-12) antibody .

  • Validation Requirements: Always validate the antibody in your specific experimental system before proceeding with full-scale studies. Western blotting showing the expected 11-12 kDa band serves as an excellent validation approach, with mouse spleen lysates being a reliable positive control .

  • Application-Specific Optimization: Optimize protocols for each application using the following starting parameters:

    • Western blotting: 1:500-1:2000 dilution

    • IHC: 1:50-1:200 dilution with high-pressure antigen retrieval in citrate buffer (pH 6.0)

    • IF/ICC: 1:100-1:500 dilution with appropriate permeabilization

    • ELISA: 1:1000-1:5000 dilution

  • Control Implementation: Include appropriate controls in every experiment:

    • Positive controls: Tissues or cell lines with known histone H4 expression

    • Negative controls: Primary antibody omission or non-specific IgG

    • Loading controls: For Western blotting, include a housekeeping protein control

  • Storage and Handling Protocols: Store the antibody at -20°C or -80°C in aliquots to avoid freeze-thaw cycles. For short-term storage and frequent use, 4°C is acceptable for up to one month .

  • Cross-Reactivity Awareness: Understand that while the antibody shows reactivity with human, mouse, and rat samples, the specificity for distinguishing between different H4 variants may be limited due to high sequence homology. When possible, confirm key findings with complementary methods .

  • Result Interpretation Framework: When interpreting results, consider the biological context:

    • Nuclear localization is expected for histone H4

    • Expression levels and modification patterns may vary between cell types and tissues

    • Changes in detection could reflect alterations in expression, accessibility, or modifications

By carefully addressing these considerations, researchers can maximize the utility of HIST1H4A (Ab-12) antibody in advancing our understanding of histone H4 biology and epigenetic regulation.

How can researchers ensure reproducibility and reliability when using HIST1H4A antibodies across different studies?

Ensuring reproducibility and reliability when using HIST1H4A antibodies requires systematic implementation of several methodological best practices:

  • Antibody Documentation and Reporting Standards: Maintain comprehensive records of antibody details and include them in publications:

    • Vendor and catalog number

    • Lot number (critical as polyclonal antibodies may vary between lots)

    • Host species and clonality

    • Immunogen sequence specifics

    • Validation evidence

  • Protocol Standardization and Sharing: Develop detailed, step-by-step protocols for each application and share them through platforms like protocols.io or as supplementary materials in publications. Critical parameters to standardize include:

    • Antibody dilutions and incubation conditions

    • Buffer compositions

    • Antigen retrieval methods for fixed samples

    • Detection systems and imaging parameters

  • Validation Across Experimental Systems: Implement multi-level validation approaches:

    • Positive controls: Use tissues or cell lines with confirmed histone H4 expression (mouse spleen lysates, PANC-1 cells)

    • Negative controls: Include antibody omission, non-immune IgG controls, and where possible, H4-depleted samples

    • Orthogonal validation: Confirm key findings using independent methods like mass spectrometry

  • Quantification and Statistical Analysis Framework: Establish rigorous quantification methods:

    • For Western blots: Use digital image analysis with linear range validation

    • For IHC/IF: Implement automated scoring systems with defined positivity thresholds

    • Always include sample size calculations and appropriate statistical tests

  • Quality Control Checkpoints: Integrate regular quality control measures:

    • Test new antibody lots against reference standards

    • Include consistent positive control samples across experiments

    • Periodically verify antibody specificity through peptide competition assays

  • Data Sharing and Repository Usage: Contribute to community resources by:

    • Depositing raw images in repositories like FigShare or Image Data Resource

    • Sharing antibody validation data through resources like Antibodypedia

    • Providing detailed methods in publications following the minimum information for research materials framework

  • Collaborative Validation Networks: Participate in multi-laboratory validation studies where possible, as independent replication across different laboratories provides the strongest evidence for antibody reliability .

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 2024 Thebiotek. All Rights Reserved.