HIST1H4A (Ab-35) 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 the products within 1-3 business days after receiving your orders. Delivery time 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
HIST1H4A, also known as Histone H4, is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery requiring 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. Research indicates that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  2. Studies suggest that post-translational modifications of histones, trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), play a role in DNA damage repair; H3K36me3 stimulates H4K16ac upon DNA double-strand break; SETD2, LEDGF, and KAT5 are necessary for these epigenetic changes. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Research shows that Omomyc protein co-localized 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 pathologic 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 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 function as 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. Research suggests 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. Imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4 PMID: 20949922
  17. 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. The 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. Findings indicate an important role of histone H4 modifications in bronchial carcinogenesis PMID: 18974389
  34. Results indicate that, 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 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 relative to normal lymphoid tissue. PMID: 19438744
  36. The release of histone H4 by holocrine secretion from the sebaceous gland may play an important 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 role does it play in chromatin structure?

HIST1H4A is one of several genes encoding the histone H4 protein, a core component of nucleosomes. Nucleosomes function as fundamental units of chromatin structure, wrapping and compacting DNA to limit its accessibility to cellular machinery. Histone H4 plays a central role in transcription regulation, DNA repair, DNA replication, and maintaining chromosomal stability .

The protein is highly conserved and exists alongside multiple H4 variants including HIST1H4B, HIST1H4C, HIST1H4D, and others, which all encode identical or nearly identical proteins. The core function of histone H4 involves forming the nucleosome core particle with other histones (H2A, H2B, and H3), around which approximately 146 base pairs of DNA are wound . This structural arrangement is crucial for genome packaging within the nucleus.

Post-translational modifications (PTMs) of histone H4 serve as key regulatory mechanisms for DNA accessibility. These modifications, including acetylation, methylation, and phosphorylation, constitute part of the "histone code" that regulates chromatin dynamics and gene expression .

What are the main applications of HIST1H4A (Ab-35) antibody in epigenetic research?

The HIST1H4A (Ab-35) antibody serves as a versatile tool in epigenetic research with several key applications:

  • ELISA (Enzyme-Linked Immunosorbent Assay): Used for quantitative detection of histone H4 and its modifications in cell or tissue lysates .

  • Immunohistochemistry (IHC): Applied to detect HIST1H4A in tissue sections, providing information about its localization and expression patterns in different cell types .

  • Immunocytochemistry (ICC): Enables visualization of HIST1H4A within cultured cells, typically showing nuclear localization as demonstrated in studies with NTera-2 human testicular embryonic carcinoma cell lines .

  • Western Blotting: Confirms the presence and approximate size (12 kDa) of histone H4 proteins in cell lysates, allowing semi-quantitative analysis of expression levels across different cell types such as Tera-2, Nalm-6, and Jurkat human cell lines .

  • Chromatin Immunoprecipitation (ChIP): Though not explicitly mentioned for Ab-35, certain HIST1H4A antibodies are suitable for ChIP applications, particularly those targeting specific modifications like acetylated lysine residues .

Each application provides unique insights into histone H4 biology, from total protein expression to specific post-translational modifications that regulate chromatin structure and function.

How does the HIST1H4A antibody recognize histone H4 modifications?

HIST1H4A antibodies are designed to recognize specific epitopes within the histone H4 protein. The Ab-35 antibody specifically targets a synthesized peptide derived from amino acids 28-39 of the human histone H4 protein . This region is particularly important for understanding histone modifications and their functional implications.

Different HIST1H4A antibodies are engineered to recognize specific post-translational modifications:

  • Acetylation-specific antibodies: These recognize acetylated lysine residues at specific positions such as Lys8, Lys12, Lys16, Lys31, Lys56, and Lys79 . Acetylation typically correlates with transcriptionally active chromatin.

  • Methylation-specific antibodies: These detect methylated lysine residues, such as mono- or di-methylated Lys20 , which is often associated with transcriptional repression.

The specificity of these antibodies is achieved through careful immunogen design and purification strategies. For instance, some antibodies have reactivity with non-acetylated peptides removed through purification processes . This ensures that the antibody specifically binds to the modified form of the histone rather than the unmodified version.

The binding specificity is critically important when studying the "histone code," as these modifications work in combination to regulate chromatin structure and gene expression.

What experimental controls should be used when working with HIST1H4A antibodies?

Proper experimental controls are essential when working with HIST1H4A antibodies to ensure reliable and interpretable results:

  • Positive Controls:

    • Cell lines known to express HIST1H4A abundantly, such as Tera-2, Nalm-6, or Jurkat cell lines

    • Recombinant HIST1H4A protein or synthetic peptides containing the target epitope

    • Tissues with documented histone H4 expression patterns

  • Negative Controls:

    • Samples treated with non-specific IgG of the same species as the primary antibody

    • Samples where the primary antibody has been pre-absorbed with the immunizing peptide

    • For modification-specific antibodies, samples treated with deacetylases or demethylases

  • Specificity Controls:

    • Western blot analysis to confirm the antibody detects a single band at the expected molecular weight (approximately 12 kDa for histone H4)

    • Peptide competition assays to demonstrate binding specificity

    • Testing samples with known differential expression or modification states

  • Technical Controls:

    • Secondary antibody-only controls to assess background signal

    • Isotype controls to evaluate non-specific binding

    • Dilution series to determine optimal antibody concentration for specific applications

When studying specific modifications, it's particularly important to validate that the antibody distinguishes between modified and unmodified forms of the histone. For instance, an antibody specific to acetylated lysine residues should not detect the unmodified protein .

How can I validate the specificity of HIST1H4A (Ab-35) antibody in my experiments?

Validating antibody specificity is crucial for generating reliable data. For HIST1H4A (Ab-35) antibody, consider these validation approaches:

  • Peptide Competition Assay: Pre-incubate the antibody with excess immunizing peptide before application to your sample. Loss of signal confirms specificity for the target epitope .

  • Western Blot Analysis: Confirm detection of a single band at approximately 12 kDa, the expected molecular weight for histone H4. Multiple bands or bands at unexpected molecular weights may indicate cross-reactivity .

  • Knockout/Knockdown Validation: Use RNA interference (siRNA) or CRISPR-Cas9 systems to reduce or eliminate HIST1H4A expression. A corresponding decrease in signal intensity provides strong evidence of specificity .

  • Cross-Reactivity Testing: Test the antibody against related histone variants to ensure it recognizes the intended target. This is particularly important for HIST1H4A due to the high sequence similarity between histone H4 variants .

  • Multi-antibody Approach: Use multiple antibodies targeting different epitopes of HIST1H4A and compare staining patterns. Concordant results increase confidence in specificity.

  • Modification-Specific Validation: For antibodies targeting modified histones, compare signals in conditions known to increase or decrease the specific modification (e.g., HDAC inhibitors for acetylation) .

  • Mass Spectrometry Validation: For definitive validation, immunoprecipitate with the antibody and analyze the pulled-down proteins using mass spectrometry to confirm target identity.

Proper validation ensures experimental observations reflect true biological phenomena rather than technical artifacts.

How can HIST1H4A antibodies be used to study the relationship between histone modifications and gene expression?

HIST1H4A antibodies provide powerful tools for investigating the complex relationship between histone modifications and gene expression through several sophisticated approaches:

  • ChIP-Seq (Chromatin Immunoprecipitation followed by Sequencing): Using antibodies that recognize specific HIST1H4A modifications, researchers can identify genomic regions associated with particular histone marks. This technique maps the distribution of histone modifications across the genome and correlates them with gene expression data .

  • ChIP-qPCR: For targeted analysis of specific gene promoters or regulatory elements, ChIP followed by quantitative PCR can determine enrichment of modified histones at regions of interest. This approach is particularly useful when studying the H4 box promoter element that regulates H4 gene expression .

  • Co-Immunoprecipitation with Transcription Factors: HIST1H4A antibodies can be used to investigate interactions between modified histones and transcription machinery components. For example, studies have shown that HAT1 (Histone Acetyltransferase 1) coordinates with H4 production and acetylation to regulate gene expression .

  • Sequential ChIP (Re-ChIP): This technique uses two antibodies sequentially to identify genomic regions that simultaneously contain two different histone modifications, providing insights into combinatorial histone codes.

  • Chromatin State Analysis: Integrating ChIP-Seq data from multiple histone modification-specific antibodies can define chromatin states associated with active transcription, repression, or poised regulatory elements.

  • Single-Cell Approaches: Combining HIST1H4A antibodies with single-cell technologies allows researchers to investigate cell-to-cell variability in histone modifications and correlate this with gene expression heterogeneity.

Through these approaches, researchers have discovered that acetylation of histone H4 is generally associated with transcriptionally active chromatin, while specific methylation patterns can correlate with either activation or repression depending on the residue modified and the degree of methylation .

What are the best practices for using HIST1H4A antibodies in ChIP experiments?

Chromatin Immunoprecipitation (ChIP) with HIST1H4A antibodies requires careful attention to protocol details for optimal results:

  • Cross-linking Optimization: Formaldehyde cross-linking should be optimized for histone targets (typically 0.75-1% formaldehyde for 10-15 minutes). Over-fixation can mask epitopes and reduce antibody accessibility to histone modifications .

  • Chromatin Fragmentation: For histone ChIP, aim for DNA fragments between 200-500 bp. Sonication conditions should be carefully optimized and verified by gel electrophoresis before proceeding with immunoprecipitation.

  • Antibody Selection and Validation:

    • Verify that the antibody recognizes the specific modification of interest

    • Ensure the antibody is ChIP-grade (many HIST1H4A antibodies are specifically validated for ChIP applications)

    • Consider using multiple antibodies targeting the same modification for validation

  • Input Controls: Always include an input control (typically 5-10% of chromatin used for IP) to normalize for differences in starting material and to calculate percent enrichment.

  • Negative Controls: Include IgG controls matched to the host species of the primary antibody to assess non-specific binding.

  • Blocking Strategy: Use BSA and sheared salmon sperm DNA in blocking solutions to reduce non-specific interactions.

  • Washing Stringency: Optimize washing conditions to maintain specific interactions while removing background. Typically, increasing salt concentration in wash buffers improves specificity.

  • Elution and Reversal of Cross-links: Complete elution of chromatin from beads and efficient reversal of cross-links are crucial for maximum recovery of target DNA.

  • Quantification Method: qPCR primers should target regions with known modifications as positive controls (e.g., actively transcribed genes for H4 acetylation marks) and regions without the modification as negative controls.

  • Sequential ChIP Considerations: When investigating co-occurrence of modifications, optimize antibody concentrations and elution conditions to maintain epitope availability for the second IP step.

Following these best practices will maximize the specificity and sensitivity of ChIP experiments using HIST1H4A antibodies, providing reliable insights into the genomic distribution of histone H4 and its modifications .

How can researchers distinguish between different histone H4 variants using antibodies?

  • Variant-Unique Epitopes: Though rare in H4 variants, some antibodies target regions containing amino acid differences between variants. These must be carefully validated for specificity .

  • Post-translational Modification Patterns: Different H4 variants may exhibit distinct patterns of post-translational modifications. Antibodies recognizing specific modifications like acetylation at Lys8, Lys12, Lys16, or methylation at Lys20 can help identify variant-specific modification profiles .

  • Combinatorial Antibody Approaches: Using antibodies targeting different epitopes or modifications in multiplexed immunofluorescence or sequential immunoprecipitation can help distinguish variant-specific patterns.

  • Mass Spectrometry Validation: For definitive variant identification, immunoprecipitation followed by mass spectrometry analysis can distinguish variants based on unique peptide fragments.

  • Expression Timing Analysis: Some H4 variants show differential expression during the cell cycle. By synchronizing cells and examining temporal expression patterns, researchers can distinguish variants with distinct cell cycle regulation .

  • Chromatin Association Patterns: Different H4 variants may associate preferentially with specific genomic regions. ChIP-seq analysis using variant-enriched samples can help map variant distribution across the genome.

  • Genetic Engineering Approaches: Introducing tagged versions of specific H4 variants allows their distinction from endogenous proteins using tag-specific antibodies.

Due to the challenge of distinguishing between H4 variants with antibodies alone, researchers often complement antibody-based approaches with molecular techniques that detect variant-specific mRNAs or employ mass spectrometry for protein-level discrimination .

What methodological approaches can resolve contradictory results in histone modification studies using HIST1H4A antibodies?

Contradictory results in histone modification studies are not uncommon and can stem from various methodological differences. Here are approaches to resolve such discrepancies:

  • Antibody Validation Comparison: Different antibodies targeting the same modification can yield varying results based on epitope recognition and specificity. Cross-validate findings using multiple antibodies from different suppliers and with different clonality (monoclonal vs. polyclonal) .

  • Standardization of Experimental Protocols:

    • Implement consistent cell fixation methods (duration, concentration)

    • Standardize chromatin preparation procedures

    • Use identical immunoprecipitation conditions

    • Normalize data analysis approaches

  • Context-Dependent Analysis: Histone modifications can vary with:

    • Cell cycle phase: Synchronize cells to eliminate cell cycle-dependent variation

    • Cell type: Compare results using identical cell types and passages

    • Growth conditions: Standardize culture conditions, including media composition and cell density

  • Quantitative Methods Assessment:

    • Compare different quantification techniques (Western blot vs. ELISA vs. mass spectrometry)

    • Implement absolute quantification methods using stable isotope-labeled standards

    • Apply consistent normalization strategies across studies

  • Integrated Multi-Omics Approach: Combine ChIP-seq data with:

    • RNA-seq to correlate modifications with transcriptional output

    • ATAC-seq to assess chromatin accessibility

    • DNA methylation analysis to understand interplay between histone and DNA modifications

  • Functional Validation Studies:

    • Use targeted histone modification enzymes (writers/erasers) to manipulate specific marks

    • Implement CRISPR-based epigenome editing to specifically alter modifications at target loci

    • Assess phenotypic consequences of modification changes to validate biological significance

  • Technical Replication and Blinding: Implement technical replicates across different laboratories and use blinded analysis to reduce experimental bias.

By systematically addressing these aspects, researchers can identify the source of contradictions and develop a more accurate understanding of histone H4 modifications and their functional implications .

How does HAT1 coordinate with HIST1H4A in regulating cell proliferation and chromatin dynamics?

HAT1 (Histone Acetyltransferase 1) plays a crucial role in coordinating with HIST1H4A to regulate cell proliferation and chromatin dynamics through several interconnected mechanisms:

  • Dual Functions of HAT1: Beyond its canonical role as a cytoplasmic histone H4 acetyltransferase, HAT1 forms a specific complex that binds directly to promoters of H4 genes, creating a feed-forward circuit that coordinates histone production and acetylation .

  • H4 Promoter Binding and Regulation: HAT1 binds specifically to the promoters of H4 genes through a specialized genetic element called the "H4 box." This element is restricted to H4 gene promoters throughout the accessible genome and is critical for transactivation of the H4 promoter. Deletion of this H4-box significantly diminishes luciferase activity in reporter assays, demonstrating its importance in H4 gene expression .

  • Acetate Sensitivity: The H4 promoter, particularly HIST1H4E, shows acetate sensitivity. Treatment with exogenous sodium acetate stimulates increased luciferase activity when the promoter is fused to a reporter gene. This effect is abolished when the H4-box is deleted, indicating that the H4-box mediates acetate-dependent transcriptional activation .

  • S-Phase Progression: HAT1 expression is critical for S-phase progression. Depletion of HAT1 leads to a modest accumulation of cells in G1 phase (5.2% increase), suggesting that HAT1 plays a role in cell cycle regulation through its effect on histone production .

  • Maintenance of Histone Acetylation: HAT1 is essential for maintaining H3 lysine 9 acetylation at proliferation-associated genes, including histone genes themselves. This creates a self-reinforcing regulatory loop where HAT1 promotes the expression of histones, which then become substrates for acetylation by HAT1 .

  • Metabolic Integration: HAT1 coordinates histone production and acetylation with glucose metabolism, suggesting that it serves as a link between cellular metabolic state and chromatin dynamics. This connection allows cells to adjust chromatin structure and gene expression in response to available energy resources .

This intricate regulatory network enables cells to coordinate the energetically demanding processes of chromatin duplication and modification with cell proliferation and metabolic status, ensuring proper chromatin assembly during DNA replication .

What techniques can be combined with HIST1H4A antibody staining to gain insights into spatial organization of chromatin?

To gain comprehensive insights into the spatial organization of chromatin using HIST1H4A antibodies, researchers can implement several advanced combinatorial techniques:

  • Super-Resolution Microscopy with Immunofluorescence: Techniques like Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED), or Stochastic Optical Reconstruction Microscopy (STORM) can be combined with HIST1H4A antibody staining to visualize chromatin organization beyond the diffraction limit. This approach has been successfully used with histone antibodies to reveal previously undetectable chromatin structures .

  • Multi-Color Immunofluorescence: Combining HIST1H4A antibodies with antibodies against other histone modifications or chromatin-associated proteins enables visualization of their spatial relationships. For example, co-staining with antibodies against active (H3K4me3) and repressive (H3K27me3) marks can identify bivalent chromatin domains .

  • Proximity Ligation Assay (PLA): This technique detects proteins in close proximity (<40 nm) and can be used to identify interactions between HIST1H4A and other chromatin components or regulatory proteins. PLA provides spatial information while maintaining the cellular context.

  • Chromatin Electron Microscopy with Immunogold Labeling: Using HIST1H4A antibodies conjugated to gold particles for electron microscopy provides ultrastructural details of chromatin organization at nanometer resolution.

  • DNA-FISH with Immunofluorescence: Combining fluorescence in situ hybridization (FISH) for specific DNA sequences with HIST1H4A antibody staining reveals relationships between histone modifications and specific genomic loci.

  • Live-Cell Imaging with Tagged Antibody Fragments: Using fluorescently labeled antibody fragments (Fabs) directed against HIST1H4A modifications allows dynamic tracking of chromatin reorganization in living cells.

  • 3D-Structured Illumination Microscopy (3D-SIM): This technique provides three-dimensional information about chromatin organization when combined with HIST1H4A antibody staining, revealing spatial relationships between different chromatin compartments.

  • Chromatin Accessibility Assays with Immunoprecipitation: Combining ATAC-seq or DNase-seq with ChIP using HIST1H4A antibodies enables correlation between chromatin accessibility and specific histone modifications.

These combinatorial approaches provide multidimensional insights into chromatin organization, helping researchers understand how histone H4 modifications contribute to nuclear architecture and genome function .

How can post-translational modifications of HIST1H4A be accurately quantified in different cell states?

Accurate quantification of HIST1H4A post-translational modifications across different cell states requires robust methodological approaches:

  • Mass Spectrometry-Based Approaches:

    • Targeted MS: Selected Reaction Monitoring (SRM) or Parallel Reaction Monitoring (PRM) allows precise quantification of specific histone modifications

    • Untargeted MS: Data-Independent Acquisition (DIA) provides comprehensive profiling of all detectable modifications

    • Chemical Derivatization: Propionylation of unmodified and monomethylated lysines prior to tryptic digestion improves detection of histone peptides

    • SILAC/TMT Labeling: Stable isotope labeling enables accurate comparative quantification across different cell states

  • Antibody-Based Quantification Methods:

    • Quantitative Western Blotting: Using fluorescent secondary antibodies with internal loading controls provides semi-quantitative data on modification levels

    • ELISA: Sandwich or competitive ELISAs using modification-specific antibodies enable absolute quantification

    • Multiplexed Bead-Based Assays: Allow simultaneous quantification of multiple histone modifications from limited sample amounts

  • Imaging-Based Quantification:

    • Quantitative Immunofluorescence: Standardized imaging protocols with calibration controls enable relative quantification of modifications in single cells

    • High-Content Imaging: Automated microscopy combined with image analysis algorithms provides population-level statistics on modification distributions

  • Genomic Approaches:

    • ChIP-seq with Spike-in Normalization: Exogenous reference chromatin (e.g., from Drosophila) allows normalization across samples for accurate comparison

    • CUT&RUN or CUT&Tag: These techniques provide higher signal-to-noise ratios than traditional ChIP, enabling more accurate quantification of genomic distribution

  • Single-Cell Analysis:

    • Mass Cytometry (CyTOF): Metal-conjugated antibodies allow quantification of multiple histone modifications at single-cell resolution

    • Single-Cell ChIP-seq: Emerging techniques enable profiling of histone modifications in individual cells

For optimal accuracy, consider these methodological considerations:

  • Include appropriate internal standards for normalization

  • Implement technical and biological replicates

  • Use multiple orthogonal techniques to validate quantitative findings

  • Control for cell cycle effects, as histone modifications can vary throughout the cell cycle

These approaches enable precise measurement of dynamic changes in HIST1H4A modifications across different cellular states, providing insights into epigenetic regulation of cell function .

What are the challenges in detecting HIST1H4A modifications in fixed versus live cell imaging?

Detecting HIST1H4A modifications presents distinct challenges in fixed versus live cell imaging contexts, each requiring specific technical considerations:

Challenges in Fixed Cell Imaging:

  • Epitope Masking: Fixation methods can alter protein conformation or create cross-links that mask epitopes. Formaldehyde fixation particularly affects histone-DNA interactions, potentially hiding certain HIST1H4A modifications .

  • Fixative Compatibility: Different modifications respond variously to fixatives. For instance, some acetylation marks are better preserved with paraformaldehyde, while others may require methanol fixation. This creates challenges when attempting to visualize multiple modifications simultaneously .

  • Permeabilization Balance: Effective antibody penetration requires sufficient permeabilization, but excessive treatment can extract histones or disrupt nuclear architecture, leading to artifacts.

  • Background Autofluorescence: Fixed cells often exhibit increased autofluorescence, which can mask low-abundance histone modifications and reduce signal-to-noise ratios.

  • Modification Stability: Some histone modifications are labile and may be lost during fixation and processing, leading to underestimation of their abundance.

Challenges in Live Cell Imaging:

  • Antibody Delivery: Full-size antibodies cannot penetrate intact cell membranes, necessitating alternative approaches like fluorescently tagged antibody fragments (Fabs) or nanobodies, which have limited availability for specific HIST1H4A modifications.

  • Signal Strength: Live imaging typically produces weaker signals than fixed samples, making detection of low-abundance modifications challenging.

  • Photobleaching and Phototoxicity: Extended imaging of living cells can cause photobleaching of fluorophores and phototoxicity, limiting observation duration and potentially altering cellular physiology.

  • Temporal Dynamics: HIST1H4A modifications change rapidly in response to cellular signals, requiring high temporal resolution imaging that can be technically demanding.

  • Specificity Verification: Confirming the specificity of signals in living cells is more challenging than in fixed samples where counterstaining and co-localization are more readily performed.

Comparative Technical Solutions:

ChallengeFixed Cell SolutionLive Cell Solution
Epitope accessibilityEpitope retrieval methods (heat, pH)Smaller antibody fragments (Fabs, nanobodies)
Signal strengthSignal amplification (TSA)Brighter fluorophores, sensitive detectors
SpecificityMultiple antibody validationFluorescent protein fusions to modification readers
ResolutionSuper-resolution microscopyLattice light-sheet microscopy
Temporal analysisSequential timepointsContinuous imaging with minimal phototoxicity

Understanding these distinct challenges helps researchers select appropriate techniques based on their specific experimental questions regarding HIST1H4A modifications .

How to optimize signal-to-noise ratio when using HIST1H4A antibodies in immunofluorescence?

Optimizing signal-to-noise ratio in HIST1H4A immunofluorescence experiments requires systematic attention to multiple experimental parameters:

  • Fixation Protocol Optimization:

    • Test multiple fixation methods (4% paraformaldehyde, methanol, or combination approaches)

    • Optimize fixation duration (typically 10-15 minutes for PFA)

    • Consider dual fixation with formaldehyde followed by methanol for certain modifications

    • Perform antigen retrieval when necessary (citrate buffer at pH 6.0 or Tris-EDTA at pH 9.0)

  • Antibody Selection and Dilution:

    • Compare different antibodies targeting the same modification

    • Perform titration experiments to determine optimal antibody concentration

    • For HIST1H4A (Ab-35), recommended dilutions for immunohistochemistry range from 1:10 to 1:100

    • Consider using directly conjugated primary antibodies to eliminate secondary antibody background

  • Blocking Strategy:

    • Use species-appropriate serum (5-10%) combined with BSA (1-3%)

    • Include detergents like Triton X-100 (0.1-0.3%) to improve penetration

    • Consider specialized blocking reagents for reduced background (e.g., Image-iT FX)

    • Extend blocking time (1-2 hours at room temperature or overnight at 4°C)

  • Washing Optimization:

    • Increase number and duration of washes (at least 3-5 washes of 5-10 minutes each)

    • Use gentle agitation during washing

    • Include detergent (0.05-0.1% Tween-20) in wash buffers

  • Advanced Detection Strategies:

    • Implement tyramide signal amplification (TSA) for low-abundance modifications

    • Use quantum dots or bright organic fluorophores (e.g., Alexa Fluor 647)

    • Consider spectral unmixing for multiplexed detection of closely spaced emission spectra

  • Microscopy Parameters:

    • Use narrow bandwidth emission filters to reduce spectral bleed-through

    • Implement deconvolution to improve signal clarity

    • Apply appropriate background subtraction algorithms during image analysis

    • Consider confocal or super-resolution techniques for improved signal localization

  • Controls for Optimization:

    • Include secondary-only controls to assess non-specific binding

    • Use peptide competition to confirm signal specificity

    • Include positive controls (cell lines with known high expression, like NTera-2)

    • Apply nuclear counterstains (DAPI) to confirm nuclear localization of signals

By systematically optimizing these parameters, researchers can significantly improve signal-to-noise ratios in HIST1H4A immunofluorescence experiments, leading to more reliable and interpretable results .

What are common pitfalls in interpreting HIST1H4A antibody results in chromatin studies?

  • Cross-Reactivity Misinterpretation:

    • Pitfall: HIST1H4A antibodies may cross-react with other H4 variants due to high sequence homology between HIST1H4A, HIST1H4B, HIST1H4C, and other variants .

    • Solution: Validate specificity through peptide competition assays, knockout controls, and mass spectrometry confirmation. Consider parallel experiments with different antibodies targeting the same modification.

  • Modification-Specificity Ambiguity:

    • Pitfall: Antibodies claiming to recognize specific modifications (e.g., acLys16) may detect that modification in contexts beyond HIST1H4A or fail to distinguish between similar modifications.

    • Solution: Verify modification-specificity using synthetic peptides with defined modifications and unmodified controls. Use orthogonal techniques like mass spectrometry to confirm modification identity .

  • Epitope Masking Effects:

    • Pitfall: Certain histone modifications or protein interactions may mask antibody epitopes, leading to false negatives, especially in densely packed chromatin regions.

    • Solution: Compare results across multiple extraction conditions and fixation protocols. Consider native versus cross-linked ChIP approaches to address epitope accessibility issues.

  • Context-Dependent Modification Patterns:

    • Pitfall: Histone modification patterns vary with cell cycle phase, differentiation state, and response to environmental signals, leading to inconsistent results across experiments.

    • Solution: Control for cell cycle effects through synchronization or cell cycle phase analysis. Document experimental conditions thoroughly and consider single-cell approaches to capture heterogeneity .

  • Antibody Batch Variation:

    • Pitfall: Different lots of the same antibody can show variation in specificity and sensitivity, creating reproducibility challenges.

    • Solution: Record lot numbers, perform batch validation before major studies, and maintain reference samples for inter-batch calibration.

  • Quantification Limitations:

    • Pitfall: Semi-quantitative techniques like Western blotting or immunofluorescence may not accurately reflect the true abundance of modifications.

    • Solution: Implement absolute quantification methods like targeted mass spectrometry. Use appropriate normalization strategies and include calibration standards .

  • Interpretation of Causality:

    • Pitfall: Correlative observations between histone modifications and functional outcomes are often interpreted as causal relationships.

    • Solution: Perform intervention studies that specifically manipulate the modification of interest (e.g., CRISPR-based epigenome editing) to establish causality.

  • ChIP Signal Interpretation:

    • Pitfall: ChIP signals may reflect antibody accessibility rather than true enrichment of the target modification.

    • Solution: Include spike-in controls, compare multiple antibodies, and integrate accessibility data (ATAC-seq, DNase-seq) to distinguish true enrichment from accessibility effects .

Awareness of these pitfalls enables more rigorous experimental design and more accurate interpretation of HIST1H4A antibody results in chromatin studies .

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.