HIST1H4A Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Function

Histone H4 (encoded by HIST1H4A) is a 103-amino acid protein that, alongside other histones, forms the nucleosome core around which DNA wraps. It plays roles in transcription regulation, DNA repair, and chromosomal stability . The HIST1H4A antibody is raised against specific epitopes of Histone H4 to enable its detection in experimental assays.

Types of HIST1H4A Antibodies

Two primary formats are available:

Catalog NumberTypeHostReactivityApplications
ABIN7181576PolyclonalRabbitHumanELISA, ICC
A14495-2PolyclonalRabbitHuman, Mouse, RatWB, ICC, IF, IHC
MAB20165MonoclonalRabbitHuman, Mouse, RatChIP, Flow Cytometry, IP, ICC, IHC
A14495PolyclonalRabbitHumanELISA, WB

Western Blot (WB)

  • A14495-2 detects Histone H4 in mouse spleen lysates at 1:1,000–1:5,000 dilution .

  • A14495 yields a single band (~11.3 kDa) in HeLa lysates .

Immunocytochemistry (ICC)

  • ABIN7181576 (1:10–1:100) and A14495-2 (1:50–1:200) stain nuclear histones in PANC-1 cells .

Chromatin Immunoprecipitation (ChIP)

  • MAB20165 is optimized for ChIP assays to study histone modifications .

Research Relevance

Histone H4 antibodies are critical for studying chromatin remodeling, gene expression, and epigenetic regulation. Post-translational modifications (e.g., acetylation, methylation) of Histone H4 influence chromatin accessibility and transcriptional outcomes .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery information.
Synonyms
dJ160A22.1 antibody; dJ160A22.2 antibody; dJ221C16.1 antibody; dJ221C16.9 antibody; FO108 antibody; H4 antibody; H4.k antibody; H4/a antibody; H4/b antibody; H4/c antibody; H4/d antibody; H4/e antibody; H4/g antibody; H4/h antibody; H4/I antibody; H4/j antibody; H4/k antibody; H4/m antibody; H4/n antibody; H4/p antibody; H4_HUMAN antibody; H4F2 antibody; H4F2iii antibody; H4F2iv antibody; H4FA antibody; H4FB antibody; H4FC antibody; H4FD antibody; H4FE antibody; H4FG antibody; H4FH antibody; H4FI antibody; H4FJ antibody; H4FK antibody; H4FM antibody; H4FN antibody; H4M antibody; HIST1H4A antibody; HIST1H4B antibody; HIST1H4C antibody; HIST1H4D antibody; HIST1H4E antibody; HIST1H4F antibody; HIST1H4H antibody; HIST1H4I antibody; HIST1H4J antibody; HIST1H4K antibody; HIST1H4L antibody; HIST2H4 antibody; HIST2H4A antibody; Hist4h4 antibody; Histone 1 H4a antibody; Histone 1 H4b antibody; Histone 1 H4c antibody; Histone 1 H4d antibody; Histone 1 H4e antibody; Histone 1 H4f antibody; Histone 1 H4h antibody; Histone 1 H4i antibody; Histone 1 H4j antibody; Histone 1 H4k antibody; Histone 1 H4l antibody; Histone 2 H4a antibody; histone 4 H4 antibody; Histone H4 antibody; MGC24116 antibody
Target Names
HIST1H4A
Uniprot No.

Target Background

Function
Histone H4 is a core component of the nucleosome. Nucleosomes are responsible for packaging and compacting DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones thus play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is modulated by a complex set of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  • Studies demonstrate that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  • Evidence suggests that post-translational modifications of histones, trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), are involved in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand break; SETD2, LEDGF, and KAT5 are required for these epigenetic changes. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  • 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
  • H4K12ac is regulated by estrogen receptor-alpha and is associated with BRD4 function and inducible transcription PMID: 25788266
  • Systemic lupus erythematosus appears to be associated with an imbalance in histone acetyltransferases and histone deacetylase enzymes, favoring pathologic H4 acetylation. PMID: 25611806
  • Sumoylated human histone H4 prevents chromatin compaction by inhibiting long-range internucleosomal interactions. PMID: 25294883
  • Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  • 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
  • Data indicate that G1-phase histone assembly is restricted to CENP-A and H4. PMID: 23363600
  • This study investigated the distribution of a specific histone modification, H4K12ac, in human sperm and characterized its specific enrichment sites in promoters throughout the whole human genome. PMID: 22894908
  • SRP68/72 heterodimers are identified as major nuclear proteins whose binding of histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  • TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is attributed to the epigenetic mechanism of suppression of acetylation of histone H4. PMID: 21973049
  • Findings suggest that global histone H3 and H4 modification patterns are potential markers of tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  • HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  • 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
  • The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4 modifications. PMID: 20949922
  • Research reveals 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
  • Histone H4 expression is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  • Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, possibly through alteration of gene expression. PMID: 12385581
  • Overexpression of MTA1 protein and acetylation level of histone H4 protein are closely related. PMID: 15095300
  • 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
  • The lack of biotinylation of K12 in histone H4 is an early signaling event in response to double-strand breaks. PMID: 16177192
  • 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
  • Apoptosis is associated with global DNA hypomethylation and histone deacetylation events in leukemia cells. PMID: 16531610
  • BTG2 contributes to retinoic acid activity by favoring differentiation through a gene-specific modification of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  • A relationship exists between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  • 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
  • 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
  • Spermatids Hypac-H4 impairment in mixed atrophy did not deteriorate further by AZFc region deletion. PMID: 18001726
  • The SET8 and PCNA interaction couples H4-K20 methylation with DNA replication. PMID: 18319261
  • H4K20 monomethylation and PR-SET7 are essential for L3MBTL1 function. PMID: 18408754
  • High expression of acetylated H4 is more prevalent in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  • Findings indicate a significant role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  • Results indicate that, through 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
  • Acetylated H4 is overexpressed in diffuse large B-cell lymphoma and peripheral T-cell lymphoma relative to normal lymphoid tissue. PMID: 19438744
  • The release of histone H4 by holocrine secretion from the sebaceous gland may play a crucial role in innate immunity. PMID: 19536143
  • 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
  • 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
  • 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
  • Data directly implicate BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714
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 important in epigenetic research?

HIST1H4A (Histone Cluster 1, H4a) is a core component of nucleosomes, the basic structural units of chromatin. This histone protein plays a critical role in DNA packaging and gene regulation through post-translational modifications. As a fundamental element in the "histone code," HIST1H4A and its modifications influence transcription regulation, DNA repair, DNA replication, and chromosomal stability .

Histone H4 modifications are particularly important in epigenetic research because they serve as markers for chromatin states, with specific modifications (acetylation, methylation, phosphorylation) correlating with gene activation or repression. The ability to detect these modifications using specific antibodies enables researchers to map epigenetic landscapes across the genome.

How do I distinguish between different HIST1H4A antibodies in my experimental design?

Distinguishing between HIST1H4A antibodies requires careful consideration of their specific binding properties:

Modification TargetCommon ApplicationsTypical HostClonalityExample Catalog Numbers
Acetyl-Lys5 (K5)WB, ChIP, IF, IHCRabbitMonoclonal/PolyclonalMAB9549, orb418625
Acetyl-Lys8 (K8)ELISA, IF, ChIP, ICCRabbitPolyclonalListed in search result 1
Acetyl-Lys12 (K12)ELISA, IF, ChIP, ICCRabbitPolyclonalABIN7139153
Acetyl-Lys16 (K16)WB, IF, ChIP, ICCRabbitPolyclonal/MonoclonalABIN7139167, M14495-3
Pan-H4 (no specific modification)WB, IHC, ChIP, IPRabbitPolyclonalab10158, ab7311

When selecting an antibody, consider:

  • The specific modification you're targeting

  • Required applications (ChIP vs. WB vs. IHC)

  • Species reactivity needed (human, mouse, rat)

  • Whether you need a monoclonal (higher specificity) or polyclonal (potentially higher sensitivity)

What are the optimal conditions for using HIST1H4A antibodies in ChIP experiments?

For successful Chromatin Immunoprecipitation with HIST1H4A antibodies:

  • Crosslinking optimization: For histone modifications, use 1% formaldehyde for 10 minutes at room temperature for efficient DNA-protein crosslinking .

  • Sonication parameters: Aim for chromatin fragments between 200-500bp for optimal resolution. This typically requires 10-15 cycles (30 seconds ON/30 seconds OFF) with modern sonicators.

  • Antibody quantity: Use 2-5μg of HIST1H4A antibody per ChIP reaction with 25μg of chromatin . Validation experiments show that antibodies like ab10158 perform effectively at this ratio.

  • Controls: Always include:

    • Input control (non-immunoprecipitated chromatin)

    • IgG negative control (non-specific antibody)

    • Positive control (known target region)

  • Washing stringency: For histone modifications, perform four washes with increasing salt concentration to reduce background while maintaining specific binding.

  • Quantification method: Real-time PCR (Taqman or Sybr green approaches) is recommended for quantifying immunoprecipitated DNA, with primers located in the first kb of the transcribed region .

How can I optimize western blot protocols when using HIST1H4A antibodies?

Optimizing western blot protocols for HIST1H4A detection requires specific adjustments:

  • Sample preparation:

    • For histones, use specialized extraction methods (acid extraction with 0.2N HCl or commercial histone extraction kits)

    • Load 10-20μg of whole cell lysate or 1-2μg of purified histones

  • Gel selection:

    • Use 15-18% SDS-PAGE gels to resolve the low molecular weight (11-12 kDa) histone proteins

    • Consider specialized Triton-Acid-Urea (TAU) gels for separating differentially modified histones

  • Transfer conditions:

    • Use PVDF membrane (0.2μm pore size) for better retention of small proteins

    • Transfer at lower voltage (30V) overnight at 4°C for efficient transfer

  • Antibody dilution:

    • Primary antibody: 1:500-1:2000 for most HIST1H4A antibodies

    • Incubation: Overnight at 4°C for optimal results

  • Detection optimization:

    • Use enhanced chemiluminescence or fluorescent secondary antibodies

    • Expected band at approximately 11-14 kDa

  • Validation: Test specificity using histone deacetylase inhibitors (e.g., sodium butyrate, TSA) to increase acetylation levels as positive controls .

Why might I observe multiple bands or unexpected patterns when using HIST1H4A antibodies?

Multiple bands or unexpected patterns with HIST1H4A antibodies could result from:

  • Post-translational modifications: Histone H4 undergoes numerous modifications (acetylation, methylation, phosphorylation) that can alter migration patterns. For example, hyperacetylation can cause slower migration .

  • Cross-reactivity: Some antibodies may cross-react with other histone variants due to high sequence homology. The search results indicate that many HIST1H4A antibodies recognize multiple histone H4 family members (HIST1H4A through HIST1H4L) .

  • Proteolytic degradation: Incomplete protease inhibition during sample preparation can result in degradation products appearing as lower molecular weight bands.

  • Antibody specificity issues: Especially for modification-specific antibodies, the specificity for a particular modification may be compromised by similar epitopes on other histones or proteins.

  • Ubiquitination or other large modifications: Larger modifications can cause significant mobility shifts, resulting in higher molecular weight bands.

To address these issues:

  • Include positive and negative controls for the specific modification

  • Use purified histones as reference standards

  • Test with different antibody lots or sources

  • Perform peptide competition assays to confirm specificity

How do I interpret contradictory results between different HIST1H4A antibody-based assays?

When facing contradictory results between different HIST1H4A antibody-based assays:

  • Consider epitope accessibility differences: Different assays (ChIP, IF, WB) have different sample preparation methods that may affect epitope accessibility. For example, an antibody might work well in ChIP but poorly in IHC due to fixation effects on epitope structure .

  • Evaluate antibody specificity:

    • Modification-specific antibodies (e.g., acLys16) may have different cross-reactivity profiles

    • Some antibodies may recognize multiple modifications or be sensitive to neighboring modifications

  • Analyze experimental conditions:

    • Different buffers, pH, and salt concentrations between assays can affect antibody binding

    • Fixation methods in IHC/IF can mask epitopes

  • Validation approach:

    • Use orthogonal techniques (mass spectrometry, ELISA) to validate findings

    • Test with cells treated with HDAC inhibitors or HAT inhibitors to modulate acetylation levels

    • Use genetic models (knockout/knockdown of histone modifying enzymes) to confirm specificity

  • Quantitative considerations:

    • ChIP provides relative enrichment data while IF/IHC provides spatial information

    • WB can be more quantitative but loses spatial information

A systematic approach comparing results across techniques with appropriate controls is essential for accurate interpretation.

How can I use HIST1H4A antibodies to investigate chromatin dynamics during cell cycle progression?

Investigating chromatin dynamics during cell cycle progression using HIST1H4A antibodies requires a multi-faceted approach:

  • Cell synchronization techniques:

    • Double thymidine block for G1/S boundary

    • Nocodazole treatment for M phase

    • Serum starvation/reintroduction for G0/G1

  • Temporal analysis strategy:

    • Collect synchronized cells at defined time points

    • Track modification patterns using modification-specific antibodies:

      • H4K5ac and H4K12ac: typically associated with newly synthesized histones during S phase

      • H4K20me1: elevated at G2/M transition

      • H4K20me2/me3: accumulates as cells move into M/G1 phase

  • Multimodal analysis:

    • ChIP-seq with phase-specific cells to map genome-wide distributions

    • Immunofluorescence microscopy for spatial organization in nucleus

    • Flow cytometry with cell cycle markers and H4 modification antibodies for quantitative assessment

  • Functional validation:

    • Inhibit specific modifying enzymes at defined cell cycle stages

    • Measure consequences on cell cycle progression

    • Correlate with transcriptional and replication timing data

This approach allows for comprehensive mapping of dynamic H4 modification patterns throughout the cell cycle and their functional implications.

What strategies can I use to validate the specificity of modification-specific HIST1H4A antibodies?

Validating modification-specific HIST1H4A antibodies requires multiple complementary approaches:

  • Peptide competition assays:

    • Pre-incubate antibody with excess modified peptide (specific modification)

    • Pre-incubate with unmodified peptide (control)

    • Compare signal reduction in WB/ChIP/IF to confirm specificity

  • Genetic manipulation approaches:

    • CRISPR/Cas9 to create point mutations at specific modification sites (e.g., K16R to prevent acetylation)

    • Knockdown/knockout of specific histone modifying enzymes (e.g., HDAC inhibitors for acetylation sites)

    • Overexpression of modifying enzymes to increase target modification

  • Pharmacological manipulation:

    • Treat cells with HDAC inhibitors (TSA, sodium butyrate) to increase acetylation

    • Use specific enzyme inhibitors targeting relevant modification pathways

    • Validate with dose-response and time-course experiments

  • Orthogonal technical validation:

    • Mass spectrometry to confirm modification status

    • Multiplex ChIP using different antibodies targeting the same modification

    • Sequential ChIP (re-ChIP) to confirm co-occurrence of modifications

    • Compare results across multiple antibody sources/clones

  • Cross-reactivity assessment:

    • Test against synthetic peptide arrays containing various histone modifications

    • Evaluate signal with modified vs. unmodified recombinant histones

    • Assess reactivity across species to leverage evolutionary conservation

These comprehensive validation strategies ensure the reliability of experimental results when using modification-specific HIST1H4A antibodies.

How can I incorporate HIST1H4A antibodies into single-cell epigenomics approaches?

Incorporating HIST1H4A antibodies into single-cell epigenomics requires adaptation of traditional methods:

  • Single-cell CUT&Tag/CUT&RUN modifications:

    • Immobilize single cells on concanavalin A-coated magnetic beads

    • Use HIST1H4A antibodies (particularly modification-specific ones) as primary antibodies

    • Employ pA-Tn5 fusion proteins for tagmentation

    • Incorporate cell barcoding strategies during library preparation

    • This approach provides genome-wide profiles of H4 modifications at single-cell resolution

  • Single-cell immunofluorescence quantification:

    • Use high-content imaging platforms with HIST1H4A antibodies

    • Employ computational image analysis for quantification

    • Correlate with other single-cell markers (transcription factors, RNA)

    • Provides spatial context missing from sequencing approaches

  • Mass cytometry (CyTOF) integration:

    • Conjugate HIST1H4A antibodies with heavy metal isotopes

    • Combine with cell cycle markers and other epigenetic modifications

    • Enables high-dimensional analysis of histone modification patterns

    • Allows correlation with cellular phenotypes in heterogeneous populations

  • Single-cell multi-omics strategies:

    • Combine scCUT&Tag using HIST1H4A antibodies with scRNA-seq

    • Integrate with chromatin accessibility data (scATAC-seq)

    • Develop computational methods to align epigenetic and transcriptomic data

    • Creates comprehensive single-cell epigenetic landscapes

These approaches enable researchers to resolve cellular heterogeneity in histone modification patterns that would be masked in bulk analyses.

What are the considerations for using HIST1H4A antibodies in live-cell imaging experiments?

Using HIST1H4A antibodies for live-cell imaging presents unique challenges and considerations:

  • Antibody delivery strategies:

    • Cell-penetrating peptide conjugation to antibodies

    • Microinjection of fluorescently labeled antibodies

    • Electroporation for temporary membrane permeabilization

    • Specialized protein transfection reagents

  • Alternative approaches:

    • Use modification-specific nanobodies instead of conventional antibodies

    • Consider recombinant modification-specific binding domains (e.g., bromodomains for acetylation)

    • Express fluorescently tagged readers of histone modifications rather than direct antibody labeling

  • Technical optimizations:

    • Test different fluorophore conjugations for optimal signal-to-noise ratio

    • Titrate antibody concentration to minimize interference with normal cellular functions

    • Employ rapid image acquisition to capture dynamic changes

    • Use spinning disk or light sheet microscopy for reduced phototoxicity

  • Controls and validation:

    • Compare live imaging results with fixed-cell immunofluorescence

    • Validate with cells treated with modification-altering compounds (HDAC inhibitors)

    • Use non-binding antibody controls to assess background

    • Include genetic controls (modification site mutants)

  • Limitations to consider:

    • Antibodies may interfere with normal chromatin dynamics

    • Access to densely packed heterochromatin may be limited

    • Resolution constraints for visualizing individual nucleosomes

    • Potential functional interference with histone-protein interactions

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.