Acetyl-HIST1H4A (K5) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Overview

Target: Acetylated lysine 5 on histone H4 (HIST1H4A/K5ac).
Function: Recognizes H4K5ac, a modification associated with transcriptional activation, chromatin decondensation, and DNA repair .

PropertyDetails
Host SpeciesRabbit (monoclonal or polyclonal )
ClonalityRecombinant monoclonal or polyclonal
ReactivityHuman, Mouse, Rat
ApplicationsWB, ChIP, IHC, IF/ICC, ELISA, IP
Specificity ValidationELISA, immunoblotting, peptide competition assays

Specificity Testing

  • ELISA: Demonstrated specificity for K5ac over K8ac, K16me1, K16me3, and unmodified H4 peptides .

  • Western Blot: Detects a single band at 11 kDa in nuclear extracts from HeLa cells treated with histone deacetylase inhibitors (e.g., Trichostatin A) .

  • Cross-Reactivity: No cross-reactivity observed with non-acetylated H4 or other histone variants .

Unique Features

  • The antibody CMA405 (monoclonal) detects K5ac only when K8 is unacetylated, enabling differentiation between newly assembled H4 (K5/K12 diacetylation) and hyperacetylated H4 (K5/K8 acetylation) .

Chromatin Immunoprecipitation (ChIP)

  • Validated in ChIP assays using formaldehyde-fixed chromatin from mouse embryonic fibroblasts (MEFs) .

  • Enriched at transcription start sites (TSS) when combined with H4K16ac .

Immunohistochemistry (IHC)

  • Staining observed in formalin-fixed paraffin-embedded (FFPE) tissues (human colon, cervical carcinoma, mouse liver) at dilutions up to 1:500 .

Immunofluorescence (IF)

  • Localizes to euchromatic regions in HeLa cells, with signal intensity increasing after Trichostatin A treatment .

ApplicationRecommended DilutionSample Type
WB1:500,000–1:1,000,000HeLa nuclear extract
IHC1:200–1:500FFPE tissues
IF/ICC1:50–1:200Cultured cells

Biological Significance of H4K5 Acetylation

  • Transcriptional Activation: K5ac correlates with open chromatin states, facilitating access for transcription machinery .

  • DNA Repair: Mediated by acetyltransferases Tip60 and CBP/p300, K5ac recruits repair factors during non-homologous end joining .

  • Cell Cycle Regulation: K5ac levels fluctuate during the cell cycle, peaking in S phase during chromatin assembly .

Comparative Data Across Sources

SourceClone/ProductKey Findings
Abcam EP1000Y (ab51997)Validated in 108 publications; IP/WB/ChIP-optimized with 11 kDa band confirmation.
Cell Signaling #9672Links K5ac to Esa1p/Tip60-mediated DNA repair and transcriptional co-activation.
Cusabio CSB-RA010429A05acHURecombinant monoclonal antibody with 1:2,000 WB dilution and no cross-reactivity.
Abbexa Polyclonal (ABX340698)Detects K5ac in human samples via IF/ICC at 1:50–1:200 dilutions.

Validation Controls

  • Negative Controls: Rabbit IgG or PBS instead of primary antibody .

  • Positive Controls: Sodium butyrate- or Trichostatin A-treated cells to enhance acetylation .

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. For specific delivery times, please contact your local distributors.
Synonyms
dJ160A22.1 antibody; dJ160A22.2 antibody; dJ221C16.1 antibody; dJ221C16.9 antibody; FO108 antibody; H4 antibody; H4.k antibody; H4/a antibody; H4/b antibody; H4/c antibody; H4/d antibody; H4/e antibody; H4/g antibody; H4/h antibody; H4/I antibody; H4/j antibody; H4/k antibody; H4/m antibody; H4/n antibody; H4/p antibody; H4_HUMAN antibody; H4F2 antibody; H4F2iii antibody; H4F2iv antibody; H4FA antibody; H4FB antibody; H4FC antibody; H4FD antibody; H4FE antibody; H4FG antibody; H4FH antibody; H4FI antibody; H4FJ antibody; H4FK antibody; H4FM antibody; H4FN antibody; H4M antibody; HIST1H4A antibody; HIST1H4B antibody; HIST1H4C antibody; HIST1H4D antibody; HIST1H4E antibody; HIST1H4F antibody; HIST1H4H antibody; HIST1H4I antibody; HIST1H4J antibody; HIST1H4K antibody; HIST1H4L antibody; HIST2H4 antibody; HIST2H4A antibody; Hist4h4 antibody; Histone 1 H4a antibody; Histone 1 H4b antibody; Histone 1 H4c antibody; Histone 1 H4d antibody; Histone 1 H4e antibody; Histone 1 H4f antibody; Histone 1 H4h antibody; Histone 1 H4i antibody; Histone 1 H4j antibody; Histone 1 H4k antibody; Histone 1 H4l antibody; Histone 2 H4a antibody; histone 4 H4 antibody; Histone H4 antibody; MGC24116 antibody
Target Names
HIST1H4A
Uniprot No.

Target Background

Function
Histone H4 is a core component of nucleosomes. Nucleosomes play a crucial role in packaging and compacting DNA into chromatin, restricting DNA accessibility to cellular machinery that requires DNA as a template. Histones, therefore, play a central role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. The accessibility of DNA is modulated by a complex interplay of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies have shown that PP32 and SET/TAF-Ibeta proteins inhibit 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), play significant roles in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand break. SETD2, LEDGF, and KAT5 are crucial for these epigenetic changes. (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 the 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 are major nuclear proteins whose binding of histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  12. TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is due to the epigenetic mechanism by suppression of acetylation of histone H4. PMID: 21973049
  13. Our data suggest that global histone H3 and H4 modification patterns are potential markers of tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  14. HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  15. Phosphorylation of histone H4 Ser 47 catalyzed by the PAK2 kinase promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by increasing the binding affinity of HIRA to H3.3-H4 and reducing association of CAF-1 with H3.1-H4. PMID: 21724829
  16. The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4. PMID: 20949922
  17. Our findings reveal the molecular mechanisms by which the DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200 which, in turn, reduces gene expression by half. PMID: 20512922
  18. Downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, possibly through alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation level of histone H4 protein are closely related. PMID: 15095300
  21. Peptidylarginine deiminase 4 regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine; data suggest that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones. PMID: 15345777
  22. Lack of biotinylation of K12 in histone H4 is an early signaling event in response to double-strand breaks. PMID: 16177192
  23. Incorporation of acetylated histone H4-K16 into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and impedes the ability of chromatin to form cross-fiber interactions. PMID: 16469925
  24. Apoptosis is associated with global DNA hypomethylation and histone deacetylation events in leukemia cells. PMID: 16531610
  25. BTG2 contributes to retinoic acid activity by favoring differentiation through a gene-specific modification of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  26. Relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. H4 tail and its acetylation have novel roles in mediating recruitment of multiple regulatory factors that can change chromatin states for transcription regulation. PMID: 17548343
  28. Brd2 bromodomain 2 is monomeric in solution and dynamically interacts with H4-AcK12; additional secondary elements in the long ZA loop may be a common characteristic of BET bromodomains. PMID: 17848202
  29. Spermatids Hypac-H4 impairment in mixed atrophy did not deteriorate further by AZFc region deletion. PMID: 18001726
  30. The SET8 and PCNA interaction couples H4-K20 methylation with DNA replication. PMID: 18319261
  31. H4K20 monomethylation and PR-SET7 are important for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more common in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Our findings indicate an important role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results indicate, by acetylation of histone H4 K16 during S-phase, early replicating chromatin domains acquire the H4K16ac-K20me2 epigenetic label that persists on the chromatin throughout mitosis 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 a significant role in innate immunity. PMID: 19536143
  37. Histone modification, including PRC2-mediated repressive histone marker H3K27me3 and active histone marker acH4, may be involved in CD11b transcription during HL-60 leukemia cells reprogramming to terminal differentiation. PMID: 19578722
  38. A role of Cdk7 in regulating elongation is further suggested by enhanced histone H4 acetylation and diminished histone H4 trimethylation on lysine 36 - two marks of elongation - within genes when the kinase was inhibited. PMID: 19667075
  39. Data showed the dynamic fluctuation of histone H4 acetylation levels during mitosis, as well as acetylation changes in response to structurally distinct histone deacetylase inhibitors. PMID: 19805290
  40. Data directly implicate BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714

Show More

Hide All

Database Links

HGNC: 4781

OMIM: 142750

KEGG: hsa:121504

STRING: 9606.ENSP00000367034

UniGene: Hs.143080

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

Q&A

What is Acetyl-HIST1H4A (K5) and why is it significant in epigenetic research?

Acetyl-HIST1H4A (K5) refers to the acetylation of lysine 5 on histone H4, one of the core histones in nucleosomes. This post-translational modification is significant in epigenetic research because it plays a critical role in chromatin structure and function. H4K5ac is predominantly associated with euchromatin (transcriptionally active regions) and contributes to chromatin decondensation and transcriptional regulation . Additionally, H4K5ac is associated with newly assembled chromatin since H4 in predeposition complexes is diacetylated at K5 and K12 by histone acetyltransferases . This makes H4K5ac a valuable marker for studying processes like DNA replication, chromatin assembly, and cellular responses to DNA damage.

How do I select the appropriate Acetyl-HIST1H4A (K5) antibody for my experiment?

When selecting an Acetyl-HIST1H4A (K5) antibody, consider these key factors:

  • Application compatibility: Confirm the antibody has been validated for your specific application (Western blot, ChIP, immunofluorescence, etc.) .

  • Species reactivity: Ensure the antibody recognizes your species of interest. Many antibodies target human H4K5ac, but cross-reactivity with mouse and rat should be verified if working with these models .

  • Clonality: Monoclonal antibodies offer higher specificity and reproducibility, while polyclonal antibodies might provide greater sensitivity .

  • Modification specificity: Some antibodies recognize H4K5ac only when neighboring residues are in specific states (e.g., CMA405 detects K5ac only when K8 is unacetylated) .

  • Validation data: Review the validation data, including specificity tests against other histone modifications and controls like peptide competition assays .

What are the common applications for Acetyl-HIST1H4A (K5) antibodies in basic research?

Acetyl-HIST1H4A (K5) antibodies are utilized in several fundamental research applications:

  • Western blotting: To detect and quantify H4K5ac levels in cell or tissue lysates. Typically appears as a band at approximately 11-12 kDa .

  • Immunofluorescence/Immunocytochemistry (IF/ICC): To visualize the nuclear localization and distribution patterns of H4K5ac in fixed cells, usually showing specific staining in nuclei .

  • Chromatin Immunoprecipitation (ChIP): To identify genomic regions enriched with H4K5ac, often combined with quantitative PCR or sequencing (ChIP-seq) .

  • ELISA: To quantitatively measure H4K5ac levels in purified histone preparations .

  • Immunohistochemistry (IHC): To examine H4K5ac distribution in tissue sections, providing insights into epigenetic states in different cell types within a tissue context .

What controls should I include when using Acetyl-HIST1H4A (K5) antibodies?

When using Acetyl-HIST1H4A (K5) antibodies, include these essential controls:

  • Positive control: Cell lines treated with histone deacetylase inhibitors like sodium butyrate, which increases global histone acetylation levels .

  • Negative control: Use of appropriate IgG matching the host species of the primary antibody .

  • Specificity control: Peptide competition assays using acetylated and non-acetylated peptides to confirm antibody specificity.

  • Biological controls: Cell lines or conditions known to have different H4K5ac levels. For example, comparing proliferating cells (higher H4K5ac) with quiescent cells.

  • Loading control: For Western blots, include total H4 antibody or other stable proteins to normalize loading variations.

  • Cross-reactivity control: Testing against other acetylated lysines on histone H4 (K8, K12, K16) to confirm specificity for K5 acetylation.

How can I distinguish between newly assembled H4 and hyperacetylated H4 using specific antibodies?

Distinguishing between newly assembled H4 (diacetylated at K5 and K12) and hyperacetylated H4 (acetylated at multiple residues including K5, K8, K12, and K16) requires antibodies with high specificity for particular acetylation patterns:

  • Context-dependent antibodies: Some antibodies, like CMA405, recognize H4K5ac only when the neighboring K8 is unacetylated . This unique feature allows researchers to specifically detect newly assembled H4 (diacetylated at K5 and K12) and distinguish it from hyperacetylated H4 (where both K5 and K8 are acetylated).

  • Sequential ChIP (ChIP-reChIP): Perform initial ChIP with antibodies against H4K5ac, followed by a second IP with antibodies against other acetylation marks. Newly assembled chromatin would be enriched for H4K5ac and H4K12ac but not H4K8ac or H4K16ac.

  • Mass spectrometry approach: Using quantitative mass spectrometry to analyze immunoprecipitated histones can provide precise identification of combinatorial modification patterns, revealing the proportions of differently modified H4 molecules.

  • Pulse-chase experiments: Combine metabolic labeling of newly synthesized histones with immunoprecipitation using H4K5ac antibodies to specifically track newly assembled chromatin versus existing modified chromatin.

What are the mechanistic differences between H4K5ac and other H4 acetylation marks in transcriptional regulation?

The mechanistic differences between H4K5ac and other H4 acetylation marks in transcriptional regulation involve distinct functions, readers, and genomic distribution patterns:

  • Functional differences:

    • H4K5ac and H4K12ac are primarily associated with newly assembled chromatin and histone deposition during DNA replication .

    • H4K8ac and H4K16ac are more directly involved in active transcription, with enrichment around transcription start sites as revealed by ChIP-seq analyses .

    • H4K16ac uniquely prevents the formation of compact 30-nm chromatin fibers and inhibits ATP-dependent chromatin remodeling, functions not shared by H4K5ac.

  • Bromodomain recognition:

    • Different bromodomain-containing proteins show varying affinities for specifically acetylated lysines on H4.

    • BRD4 preferentially binds to H4K5ac and H4K8ac, while BRDT recognizes H4K5ac in combination with other acetylation marks.

  • Genomic distribution:

    • ChIP-seq analysis reveals different enrichment patterns, with H4K8ac and H4K16ac showing stronger association with transcription start sites compared to H4K5ac .

    • H4K5ac is more broadly distributed and associated with replication-dependent histone deposition throughout the genome.

  • Contextual effects:

    • H4K5ac functions may depend on neighboring modifications, creating a complex "histone code" that influences transcription factor binding and chromatin structure.

How do H4K5ac patterns change during DNA damage response and what implications does this have for repair mechanisms?

H4K5ac undergoes dynamic changes during DNA damage response (DDR), with significant implications for repair mechanisms:

  • Initial response to damage:

    • Upon DNA damage, there is often a global decrease in H4K5ac levels as part of chromatin compaction to prevent further damage.

    • This is followed by localized increases of H4K5ac at damage sites to facilitate access for repair proteins.

  • Double-strand break (DSB) repair pathway choice:

    • H4K5ac levels influence the choice between homologous recombination (HR) and non-homologous end joining (NHEJ) repair pathways.

    • Higher levels of H4K5ac create a more open chromatin environment that favors HR, which requires greater chromatin accessibility.

  • Cell cycle-dependent regulation:

    • The relationship between H4K5ac and DNA repair varies across the cell cycle.

    • In S phase, when newly assembled chromatin (marked by H4K5ac and H4K12ac) is abundant, H4K5ac patterns contribute to replication-coupled repair mechanisms.

  • Interaction with repair machinery:

    • H4K5ac serves as a binding platform for bromodomain-containing proteins involved in the DNA damage response.

    • The acetylation-dependent recruitment of chromatin remodelers facilitates access to damaged DNA for repair proteins.

  • Restoration phase:

    • Following successful repair, proper restoration of chromatin structure requires precise regulation of H4K5ac and other histone modifications.

    • Failure to reset proper acetylation patterns may lead to epigenetic instability and increased mutation rates.

What are the optimal conditions for using Acetyl-HIST1H4A (K5) antibodies in ChIP experiments?

Optimizing ChIP experiments with Acetyl-HIST1H4A (K5) antibodies requires careful attention to several parameters:

  • Crosslinking conditions:

    • For most H4K5ac ChIP applications, 1% formaldehyde for 10 minutes at room temperature provides adequate crosslinking.

    • Excessive crosslinking may mask epitopes and reduce antibody accessibility.

  • Chromatin fragmentation:

    • Micrococcal nuclease digestion is often preferred for histone modification ChIP, as demonstrated in protocols using Acetyl-HIST1H4A (K5) antibodies .

    • Aim for chromatin fragments of 150-500 bp for optimal resolution.

  • Antibody amount and incubation:

    • Typically, 5 μg of anti-Acetyl-HIST1H4A (K5) antibody per ChIP reaction is recommended .

    • Overnight incubation at 4°C with rotation ensures adequate antibody-chromatin interaction.

  • Washing stringency:

    • Include progressively stringent wash steps to reduce background without compromising specific signal.

    • For H4K5ac, standard RIPA buffer washes followed by LiCl and TE washes are generally effective.

  • Controls:

    • Include input chromatin control (typically 5-10% of starting material).

    • Use normal rabbit IgG as a negative control to assess non-specific binding .

    • Consider using cell treatments that alter H4K5ac levels (e.g., HDAC inhibitors) as biological controls.

  • Quantification method:

    • For targeted analysis, quantitative PCR with primers for known H4K5ac-enriched regions.

    • For genome-wide analysis, ChIP-seq with appropriate sequencing depth (minimum 20 million uniquely mapped reads).

How can I effectively troubleshoot weak or non-specific signals in Western blots using Acetyl-HIST1H4A (K5) antibodies?

ProblemPossible CauseSolution
No signalInsufficient histone extractionUse acidic extraction methods specifically designed for histones
Epitope masked or degradedInclude protease and HDAC inhibitors in lysis buffers
Antibody concentration too lowIncrease antibody concentration (recommended range: 0.1-1 μg/mL)
High backgroundInsufficient blockingIncrease blocking time or try alternative blocking agents
Secondary antibody concentration too highDilute secondary antibody further
Non-specific bindingAdd 0.1% Tween-20 to antibody diluent
Multiple bandsCross-reactivity with other acetylated histonesVerify antibody specificity with peptide competition assays
Histone degradationAdd protease inhibitors during sample preparation
Weak signalLow abundance of H4K5acTreat cells with HDAC inhibitors like sodium butyrate
Poor transfer efficiencyOptimize transfer conditions for low molecular weight proteins
Inconsistent resultsVariable acetylation levelsStandardize cell culture conditions and harvesting protocols
Antibody lot variationValidate each new antibody lot with positive controls

For optimal results with Acetyl-HIST1H4A (K5) antibodies in Western blot:

  • Use PVDF membrane for better protein retention

  • Run under reducing conditions using Immunoblot Buffer Group 1

  • Include both acetylated and non-acetylated control samples

  • Ensure adequate transfer of low molecular weight histones (11-12 kDa)

What are the critical parameters for immunofluorescence staining when using Acetyl-HIST1H4A (K5) antibodies?

For optimal immunofluorescence staining with Acetyl-HIST1H4A (K5) antibodies, attention to these critical parameters is essential:

  • Fixation method:

    • Paraformaldehyde (4%) for 10-15 minutes is generally recommended for preserving nuclear architecture.

    • Methanol fixation may expose epitopes better in some cell types but can disrupt nuclear morphology.

  • Permeabilization:

    • Adequate nuclear permeabilization is crucial for antibody access to nuclear epitopes.

    • Use 0.2-0.5% Triton X-100 for 10 minutes at room temperature.

  • Antigen retrieval:

    • Some protocols benefit from antigen retrieval using citrate buffer (pH 6.0) heating.

    • This may be particularly important for detecting H4K5ac in cells with compact chromatin.

  • Blocking conditions:

    • Thorough blocking (1 hour) with 5% normal serum from the same species as the secondary antibody.

    • Addition of 0.1-0.3% BSA can reduce non-specific binding.

  • Antibody concentration and incubation:

    • Optimal dilution ranges from 1:50 to 1:200 for most Acetyl-HIST1H4A (K5) antibodies .

    • For HeLa cells, 0.1 μg/mL has been validated .

    • Extending primary antibody incubation to 3 hours at room temperature or overnight at 4°C improves signal quality .

  • Washing steps:

    • Multiple PBS washes (at least 3×5 minutes) after both primary and secondary antibody incubations.

    • Include 0.05% Tween-20 in wash buffers to reduce background.

  • Secondary antibody selection:

    • Use highly cross-adsorbed secondary antibodies to minimize non-specific binding.

    • For Acetyl-HIST1H4A (K5) rabbit antibodies, anti-rabbit IgG conjugated to fluorophores like NorthernLights 557 has been validated .

  • Nuclear counterstain:

    • DAPI is recommended for nuclear visualization and colocalization analysis with H4K5ac signals .

    • Ensure DAPI concentration and incubation time are optimized to avoid oversaturation.

How can I quantitatively analyze H4K5ac levels in ChIP-seq data and integrate with other epigenetic marks?

Quantitative analysis of H4K5ac ChIP-seq data and integration with other epigenetic marks requires sophisticated computational approaches:

  • Quality control and preprocessing:

    • Assess sequencing quality using FastQC.

    • Filter low-quality reads and trim adapters before alignment.

    • Map reads to reference genome using Bowtie2 or BWA.

    • Remove PCR duplicates to prevent bias in quantification.

  • Peak calling and signal quantification:

    • Use MACS2 or SICER for peak calling, with input DNA as control.

    • For H4K5ac, which can show broad domains, consider broad peak calling parameters.

    • Generate normalized coverage tracks (bigWig format) for visualization and quantification.

  • Differential binding analysis:

    • Compare H4K5ac enrichment between conditions using DiffBind or MAnorm.

    • Normalize for sequencing depth and potential global changes in acetylation levels.

    • Apply appropriate statistical thresholds (FDR < 0.05) for identifying significant changes.

  • Genomic feature association:

    • Analyze distribution of H4K5ac relative to genomic features (promoters, enhancers, gene bodies).

    • Use tools like HOMER or ChIPseeker for annotation of H4K5ac peaks.

    • Create metaplots and heatmaps showing H4K5ac distribution around transcription start sites.

  • Integration with other epigenetic marks:

    • Perform correlation analysis between H4K5ac and other histone modifications.

    • Use ChromHMM or EpiSig for chromatin state modeling based on multiple marks.

    • Apply multivariate analysis to identify combinatorial patterns of histone modifications.

    • Compare H4K5ac with H4K8ac and H4K16ac to distinguish newly assembled chromatin from transcriptionally active regions .

  • Functional interpretation:

    • Perform gene ontology and pathway enrichment analysis of genes associated with H4K5ac peaks.

    • Integrate with gene expression data to correlate H4K5ac changes with transcriptional outcomes.

    • Analyze motif enrichment within H4K5ac peaks to identify potential regulatory factors.

How should I interpret discrepancies between H4K5ac antibodies from different sources in my experiments?

When faced with discrepancies between H4K5ac antibodies from different sources, consider these analytical approaches:

What is the relationship between H4K5ac and other histone modifications in different cellular contexts?

The relationship between H4K5ac and other histone modifications varies across cellular contexts, creating complex regulatory networks:

  • Co-occurrence patterns:

    • H4K5ac frequently co-occurs with H4K12ac in newly assembled chromatin .

    • H4K5ac may be mutually exclusive with H4K20me3, as these marks are associated with opposite chromatin states (open vs. condensed) .

    • In transcriptionally active regions, H4K5ac often coincides with H3K4me3 and H3K27ac.

  • Cell cycle-dependent relationships:

    • During S phase, newly deposited histones show enrichment of H4K5ac and H4K12ac, which are subsequently modified as chromatin matures .

    • This pattern differs from the stable H4K8ac and H4K16ac enrichment at transcription start sites throughout the cell cycle .

    • In mitotic cells, global reduction of H4K5ac accompanies chromosome condensation.

  • Development and differentiation contexts:

    • Stem cells show distinct patterns of H4K5ac distribution compared to differentiated cells.

    • During differentiation, dynamic changes in H4K5ac correlate with developmental gene regulation.

    • Tissue-specific patterns of H4K5ac reflect specialized gene expression programs.

  • Disease states:

    • Cancer cells often exhibit altered patterns of H4K5ac and other histone modifications.

    • Similar to H4K16ac and H4K20me3, H4K5ac levels may have diagnostic and prognostic value in certain cancers .

    • Neurodegenerative disorders show disruptions in the balance between histone acetylation and deacetylation, affecting H4K5ac levels.

  • Response to environmental stimuli:

    • Stress conditions can rapidly alter H4K5ac distribution as part of adaptive transcriptional responses.

    • Nutritional status influences global histone acetylation, including H4K5ac, through metabolic regulation of acetyl-CoA availability.

What are the emerging roles of H4K5ac in regulating non-coding RNA expression and three-dimensional chromatin organization?

Emerging research reveals complex roles for H4K5ac in non-coding RNA regulation and chromatin architecture:

  • Long non-coding RNA (lncRNA) regulation:

    • H4K5ac enrichment at lncRNA promoters correlates with their expression, similar to protein-coding genes.

    • Dynamic changes in H4K5ac can mediate tissue-specific expression of regulatory lncRNAs.

    • Some lncRNAs themselves affect H4K5ac deposition through interactions with histone acetyltransferases or deacetylases.

  • Enhancer RNA (eRNA) production:

    • Enrichment of H4K5ac at active enhancers, often in combination with H3K27ac, correlates with eRNA transcription.

    • eRNAs may subsequently influence broader H4K5ac distribution through feedback mechanisms.

    • The presence of H4K5ac at enhancers facilitates chromatin accessibility for transcription factor binding.

  • Topologically associating domains (TADs):

    • H4K5ac distribution patterns often respect TAD boundaries, suggesting coordination with three-dimensional chromatin organization.

    • Changes in H4K5ac levels can precede or accompany alterations in chromatin interaction frequencies.

    • The presence of H4K5ac may influence the flexibility of chromatin fibers, affecting long-range interactions.

  • Chromatin accessibility and nucleosome positioning:

    • H4K5ac contributes to local chromatin accessibility by weakening histone-DNA interactions.

    • This modification can alter nucleosome stability and positioning, particularly when combined with other acetylation marks.

    • Advanced techniques combining H4K5ac ChIP with ATAC-seq or DNase-seq provide insights into the relationship between this modification and chromatin accessibility.

  • Nuclear compartmentalization:

    • Regions with high H4K5ac often localize to the active A compartment in the nucleus.

    • During cell differentiation or response to stimuli, changes in H4K5ac can accompany repositioning of genomic regions between compartments.

    • The relationship between H4K5ac and nuclear organization reveals higher-order regulation beyond sequence-specific effects.

What emerging technologies will advance our understanding of H4K5ac dynamics and function?

Several cutting-edge technologies are poised to revolutionize our understanding of H4K5ac biology:

  • Single-cell epigenomics:

    • Single-cell ChIP-seq and CUT&Tag approaches will reveal cell-to-cell variation in H4K5ac patterns.

    • Integration with single-cell transcriptomics will clarify the relationship between H4K5ac heterogeneity and gene expression variability.

    • These approaches will be particularly valuable for understanding H4K5ac dynamics in heterogeneous tissues and during development.

  • Live-cell imaging of H4K5ac:

    • Development of acetylation-specific intrabodies and fluorescent probes for real-time visualization of H4K5ac in living cells.

    • FRET-based sensors to monitor dynamic changes in H4K5ac in response to stimuli with high temporal resolution.

    • These techniques will provide unprecedented insights into the kinetics of H4K5ac establishment and removal.

  • Mass spectrometry innovations:

    • Improved sensitivity to detect combinatorial histone modifications including H4K5ac.

    • Development of targeted approaches for quantifying specific histone proteoforms.

    • Integration with crosslinking techniques to identify proteins that specifically interact with H4K5ac-modified chromatin.

  • Genomic engineering of acetylation sites:

    • CRISPR-based approaches for site-specific manipulation of H4K5ac levels.

    • Creation of acetylation-mimetic or acetylation-deficient mutants to dissect functional consequences.

    • These interventions will help establish causality between H4K5ac and specific cellular processes.

  • Computational modeling:

    • Machine learning algorithms to predict H4K5ac patterns from DNA sequence and other epigenetic features.

    • Structural modeling of how H4K5ac affects nucleosome and chromatin fiber dynamics.

    • Systems biology approaches integrating multiple data types to place H4K5ac in broader regulatory networks.

These emerging technologies will address current knowledge gaps and provide more comprehensive understanding of H4K5ac function in genome regulation, development, and disease.

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.